1
|
Cui Z, He J, Li A, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Novel insights into non-coding RNAs and their role in hydrocephalus. Neural Regen Res 2026; 21:636-647. [PMID: 39688559 DOI: 10.4103/nrr.nrr-d-24-00963] [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: 08/20/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
A large body of evidence has highlighted the role of non-coding RNAs in neurodevelopment and neuroinflammation. This evidence has led to increasing speculation that non-coding RNAs may be involved in the pathophysiological mechanisms underlying hydrocephalus, one of the most common neurological conditions worldwide. In this review, we first outline the basic concepts and incidence of hydrocephalus along with the limitations of existing treatments for this condition. Then, we outline the definition, classification, and biological role of non-coding RNAs. Subsequently, we analyze the roles of non-coding RNAs in the formation of hydrocephalus in detail. Specifically, we have focused on the potential significance of non-coding RNAs in the pathophysiology of hydrocephalus, including glymphatic pathways, neuroinflammatory processes, and neurological dysplasia, on the basis of the existing evidence. Lastly, we review the potential of non-coding RNAs as biomarkers of hydrocephalus and for the creation of innovative treatments.
Collapse
Affiliation(s)
- Zhiyue Cui
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - An Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan Province, China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke 's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Ren Y, Liang J, Chen B, Liu X, Chen J, Liu X, Chen Y. LncRNA AC100865.1 regulates macrophage adhesion and ox-LDL intake through miR-7/GDF5 pathway. Cell Signal 2025; 131:111748. [PMID: 40096934 DOI: 10.1016/j.cellsig.2025.111748] [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: 03/13/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES Cardiovascular disease (CVD) accounts for over 40 % of deaths related to diseases among residents. Atherosclerosis (AS) and its associated thrombosis are the primary causes of CVD. LncRNA AC100865.1, a newly identified lncRNA, has shown potential as a diagnostic biomarker for AS. This study aims to evaluate the therapeutic value of lncRNA AC100865.1 in AS. METHODS Real-time PCR was conducted to assess the relative expression of lncRNA AC100865.1 in Peripheral Blood Mononuclear Cell (PBMC) samples from 50 CVD patients and 50 healthy controls. lncRNA AC100865.1 was overexpressed in RAW264.7 cells to measure its effects on adhesion and oxidized low-density lipoprotein (ox-LDL) uptake. Flow cytometry was utilized to identify the pathway mediating these processes. The luciferase assay and knockout rescue experiments were performed to elucidate the downstream signaling pathways involved. RESULTS lncRNA AC100865.1 expression was found to be downregulated in CVD patients. Overexpression of lncRNA AC100865.1 significantly enhanced the adhesion capacity of RAW264.7 cells. Luciferase reporter assays and flow cytometry indicated that this effect is mediated through the miR-7/GDF5/p38/LFA-1 pathway. Furthermore, lncRNA AC100865.1 notably increased ox-LDL uptake by macrophages via upregulation of CD36 expression. CONCLUSION Overexpression of lncRNA AC100865.1 enhances the adhesion of RAW264.7 cells through the miR-7/GDF5/p38/LFA-1 pathway and increases ox-LDL uptake by elevating CD36 levels. These findings suggest that circulating lncRNA AC100865.1 may serve not only as an early diagnostic marker for CVD but also as a potential therapeutic target, offering new prospects for CVD treatment.
Collapse
Affiliation(s)
- Yong Ren
- Central Medical Laboratory, Zhengzhou Yihe Hospital, Zhengzhou, PR China
| | - Jiarong Liang
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China
| | - Baofeng Chen
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China
| | - Xiangyang Liu
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China
| | - Jinfeng Chen
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China
| | - Xiangying Liu
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China
| | - Yunxian Chen
- Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, PR China.
| |
Collapse
|
3
|
Liu X, Zhang L, Chen J, Shao W. Decoding intricate interactions between m6A modification with mRNAs and non-coding RNAs in cervical cancer: Molecular mechanisms and clinical implications. Cell Signal 2025; 131:111745. [PMID: 40107480 DOI: 10.1016/j.cellsig.2025.111745] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent RNA modification that is regulated by three regulatory factors: "writers", "erasers" and "readers". m6A modification regulates RNA stability and other mechanisms, including translation, cleavage, and degradation. Current research has demonstrated that m6A methylation is involved in the regulation of occurrence and development of cancers by controlling the expression of cancer-related genes. This review summarizes the role of m6A modification on messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) in cervical cancer (CC). We highlight the dual role of m6A regulatory factors, which act as oncogenes or tumor suppressors depending on the cellular context and downstream targets. Additionally, we examine how ncRNAs reciprocally regulate m6A modification in two ways: by guiding the deposition or removal of m6A modifications on RNA targets, and by modulating the expression of m6A regulatory factors. These interactions further contribute to tumor progression. Furthermore, the therapeutic potential of targeting m6A modification has been emphasized in CC. Moreover, recent advances in small-molecule inhibitors targeting m6A regulators and RNA-based therapies which may offer new treatment strategies have been summarized. Finally, we discuss the current challenges in m6A modification research and provide suggestions for future research directions. This review aims to deepen the understanding of m6A modification in CC and contribute to the development of targeted and personalized treatment strategies.
Collapse
Affiliation(s)
- Xuefei Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China; First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Lizhi Zhang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Ji Chen
- Department of Obstetrics, The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
4
|
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
|
5
|
Wu L, Sun J, Wang L, Chen Z, Guan Z, Du L, Qu R, Liu C, Shao Y, Hua Y. Whole-transcriptome sequencing in neural and non-neural tissues of a mouse model identifies miR-34a as a key regulator in SMA pathogenesis. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102490. [PMID: 40125274 PMCID: PMC11930137 DOI: 10.1016/j.omtn.2025.102490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025]
Abstract
Spinal muscular atrophy (SMA) is a severe neurodegenerative disorder caused by deficiency of survival of motor neuron (SMN). While significant progress has been made in SMA therapy by rescuing SMN expression, limited knowledge about SMN downstream genes has hindered the development of alternative therapies. Here, we conducted whole-transcriptome sequencing of spinal cord, heart, and liver tissues of a severe SMA mouse model at early postnatal ages to explore critical coding and non-coding RNAs (ncRNAs). A large number of differentially expressed RNAs (DE-RNAs) were obtained, including 2,771 mRNAs, 382 microRNAs (miRNAs), 1,633 long ncRNAs, and 1,519 circular RNAs. Through in-depth data mining, we unveiled deregulation of miR-34a in all tissues. Analysis of competitive endogenous RNA networks of DE-RNAs identified multiple novel targets of miR-34a including Spag5 mRNA, lncRNA00138536, and circRNA007386. Further in vitro studies using mouse myoblast and human cardiomyocyte cell lines showed that knockdown of SMN upregulated miR-34a-5p and overexpression of miR-34a-5p alone disrupted cell-cycle progression through regulating its targets, recapitulating gene expression patterns observed in cardiac tissue of SMA mice. Our results identified a critical miRNA involved in SMA pathology, which sheds insights into the molecular basis of widespread tissue abnormalities observed in severe forms of SMA.
Collapse
Affiliation(s)
- Liucheng Wu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Laboratory Animal Center, Nantong University, Nantong 226001, China
- Institute of Neuroscience, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Li Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Zhiheng Chen
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Zeyuan Guan
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Institute of Neuroscience, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Lili Du
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ruobing Qu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Institute of Neuroscience, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Chun Liu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Yixiang Shao
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Yimin Hua
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
6
|
Sun YG, Yu ZK, Chen X, Zhang SY, Wu WJ, Liu K, Cheng L. circHIPK2 promotes malignant progression of laryngeal squamous cell carcinoma through the miR-889-3p/MCTS1/IL-6 axis. Transl Oncol 2025; 56:102390. [PMID: 40222337 PMCID: PMC12018564 DOI: 10.1016/j.tranon.2025.102390] [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: 08/19/2024] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor of the head and neck with a poor prognosis. The role of circRNAs in LSCC remains largely unknown. In this study, quantitative real-time PCR (qRT-PCR), Sanger sequencing and fluorescence in situ hybridization were undertaken to detect the expression, localization, and clinical significance of circHIPK2 in LSCC tissues and TU686 and TU212 cells. The functions of circHIPK2 in LSCC were explored through proliferation analysis, EdU staining, colony formation assay, wound healing assay, and Transwell assay. The regulatory mechanisms underpinning circHIPK2, miR-889-3p, and MCTS1 were investigated using luciferase assay, Western blotting, and qRT-PCR. We found that LSCC tissues and cells demonstrated high expression of circHIPK2 that was closely associated with the malignant progression and poor prognosis of LSCC. Knockdown of circHIPK2 inhibited the proliferation and migration of LSCC cells in vitro. Mechanistic studies showed that circHIPK2 competitively bound to miR-889-3p, elevated MCTS1 level, promoted IL-6 secretion, and ultimately accelerated the malignant progression of LSCC. In conclusion, an axis involving circHIPK2, miR-889-3p, MCTS1 and IL-6 regulates the malignant progression of LSCC. circHIPK2 expression may serve as a novel diagnostic and prognostic biomarker for LSCC.
Collapse
Affiliation(s)
- Yang-Guang Sun
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Zhen-Kun Yu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Xi Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Si-Yao Zhang
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Wan-Juan Wu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Kai Liu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Lei Cheng
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of Allergology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
7
|
Spinello Z, Besharat ZM, Mainiero F, Rughetti A, Masuelli L, Ferretti E, Catanzaro G. MiR-326: Role and significance in brain cancers. Noncoding RNA Res 2025; 12:56-64. [PMID: 40115178 PMCID: PMC11925037 DOI: 10.1016/j.ncrna.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/23/2025] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that act as critical regulators of gene expression by repressing mRNA translation. The role of miRNAs in cell physiology spans from cell cycle control to cell proliferation and differentiation, both during development and in adult tissues. Accordingly, dysregulated expression of miRNAs has been reported in several diseases, including cancer, where miRNAs can act as oncogenes or oncosuppressors. Of note, miRNA signatures are also under investigation for classification, diagnosis, and prognosis of cancer patients. Brain tumours are primarily associated with poor prognosis and high mortality, highlighting an urgent need for novel diagnostic, prognostic, and therapeutic tools. Among miRNAs investigated in brain tumours, miR-326 has been shown to act as a tumour suppressor in adult and paediatric brain cancers. In this review, we describe the role of miR-326 in malignant as well as benign cancers originating from brain tissue. In addition, since miR-326 expression can be regulated by other non-coding RNA species, adding a further layer of regulation in the cancer-promoting axis, we discuss this miRNA's role in targeted therapy for brain cancers.
Collapse
Affiliation(s)
- Zaira Spinello
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Fabrizio Mainiero
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Giuseppina Catanzaro
- Department of Life Science, Health, and Health Professions, Link Campus University, 00165, Rome, Italy
| |
Collapse
|
8
|
Wang X, He X, Li Z, Mu T, Pang L, Ma W, Hu X. Insight into dysregulated VEGF-related genes in diabetic retinopathy through bioinformatic analyses. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7199-7217. [PMID: 39725717 DOI: 10.1007/s00210-024-03638-y] [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: 05/23/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
Diabetic retinopathy (DR) is a prevalent microvascular complication of diabetes mellitus. VEGF plays a pivotal role in the pathogenesis of DR. To characterize the VEGF-related genes in DR patients, the RNAseq dataset of DR and normal control were downloaded from the GEO database and analyzed using R package limma. The differentially expressed VEGFGs between DR and NC were identified, and their expression levels were verified through qRT-PCR and Western blotting. Enrichment analyses were performed to understand the key functions and involved pathways of DE-VEGFGs. A two-sample MR analysis was carried out to study the causal link between prostate cancer and DR. Next, we built a nomogram model to predict the risk of DR using the expression level of DE-VEGFGs. Additionally, we estimated the immune cell infiltration between clusters and calculated the correlation between DE-VEGFGs expression and immune cell infiltration in DR. The DGIdb database was used to identify potential target drug for DE-VEGFGs. Finally, we constructed a ceRNA regulation network with predictions from miRNA-mRNA interaction databases and miRNA-lncRNA interaction database. We identified six DE-VEGFGs that are involved in the regulation of the VEGF pathway. The two-sample MR analysis revealed a positive correlation between prostate cancer and the risk of DR. The nomogram which uses the DE-VEGFGs expression to predict the DR risk shows good performance based on the calibration curve and AUC value. Monocytes and T cells CD4 memory activated show different expression between DR and NC; meanwhile, these cell types were correlated with DE-VEGFGs. The drug-gene interaction network provides candidates for DR treatment, and the ceRNA regulation network suggests a potential biomarker for DR. Our study identified dysregulated VEGF-related genes in DR and emphasized their significance in the pathogenesis of DR. Additionally, our findings offer insights into their potential clinical predictive value, immune implications, targeting drug candidates, and regulatory network dynamics.
Collapse
Affiliation(s)
- Xiaoguang Wang
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Xianglian He
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Zhen Li
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Tao Mu
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Lin Pang
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Weiguo Ma
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China
| | - Xuejun Hu
- Ningxia Hui Autonomous Region People's Hospital, Ningxia Eye Hospital, No. 301 Zhengyuan North Street, Jinfeng District, Yinchuan City, 750004, Ningxia Hui Autonomous, China.
| |
Collapse
|
9
|
Wang H, Bian C, Zhang Y, Zhang L, Wang F. Circular RNAs in glioma progression: Fundamental mechanisms and therapeutic potential: A review. Int J Biol Macromol 2025; 313:144360. [PMID: 40388873 DOI: 10.1016/j.ijbiomac.2025.144360] [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: 04/28/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Gliomas are the most common primary malignant brain tumors, characterized by aggressive invasion, limited therapeutic options, and poor prognosis. Despite advances in surgery, radiotherapy, and chemotherapy, the median survival of glioma patients remains disappointingly low. Therefore, identifying glioma-associated therapeutic targets and biomarkers is of significant clinical importance. Circular RNAs (circRNAs) are a class of naturally occurring long non-coding RNAs (lncRNAs), notable for their stability and evolutionary conservation. Increasing evidence indicates that circRNA expression is dysregulated in gliomas compared to adjacent non-tumor tissues and contributes to the regulation of glioma-related biological processes. Furthermore, numerous circRNAs function as oncogenes or tumor suppressors, mediating glioma initiation, progression, and resistance to temozolomide (TMZ). Mechanistically, circRNAs regulate glioma biology through diverse pathways, including acting as miRNA sponges, binding RNA-binding proteins (RBPs), modulating transcription, and even encoding functional peptides. These features highlight the potential of circRNAs as diagnostic and prognostic biomarkers, as well as therapeutic targets for glioma. This review summarizes the dysregulation and functions of circRNAs in glioma and explores key mechanisms through which they mediate tumor progression, including DNA damage repair, programmed cell death (PCD), angiogenesis, and metabolic reprogramming. Our aim is to provide a comprehensive perspective on the multifaceted roles of circRNAs in glioma and to highlight their potential for translational application in targeted therapy.
Collapse
Affiliation(s)
- Hongbin Wang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Chenbin Bian
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Yidan Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Li Zhang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Wang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [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: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
11
|
Yuan J, Liao YS, Zhang TC, Tang YQ, Yu P, Liu YN, Cai DJ, Yu SG, Zhao L. Integrating Bulk RNA and Single-Cell Sequencing Data Unveils Efferocytosis Patterns and ceRNA Network in Ischemic Stroke. Transl Stroke Res 2025; 16:733-746. [PMID: 38678526 DOI: 10.1007/s12975-024-01255-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/17/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
Excessive inflammatory response following ischemic stroke (IS) injury is a key factor affecting the functional recovery of patients. The efferocytic clearance of apoptotic cells within ischemic brain tissue is a critical mechanism for mitigating inflammation, presenting a promising avenue for the treatment of ischemic stroke. However, the cellular and molecular mechanisms underlying efferocytosis in the brain after IS and its impact on brain injury and recovery are poorly understood. This study explored the roles of inflammation and efferocytosis in IS with bioinformatics. Three Gene Expression Omnibus Series (GSE) (GSE137482-3 m, GSE137482-18 m, and GSE30655) were obtained from NCBI (National Center for Biotechnology Information) and GEO (Gene Expression Omnibus). Differentially expressed genes (DEGs) were processed for GSEA (Gene Set Enrichment Analysis), GO (Gene Ontology Functional Enrichment Analysis), and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses. Efferocytosis-related genes were identified from the existing literature, following which the relationship between Differentially Expressed Genes (DEGs) and efferocytosis-related genes was examined. The single-cell dataset GSE174574 was employed to investigate the distinct expression profiles of efferocytosis-related genes. The identified hub genes were verified using the dataset of human brain and peripheral blood sample datasets GSE56267 and GSE122709. The dataset GSE215212 was used to predict competing endogenous RNA (ceRNA) network, and GSE231431 was applied to verify the expression of differential miRNAs. At last, the middle cerebral artery (MCAO) model was established to validate the efferocytosis process and the expression of hub genes. DEGs in two datasets were significantly enriched in pathways involved in inflammatory response and immunoregulation. Based on the least absolute shrinkage and selection operator (LASSO) analyses, we identified hub efferocytosis-related genes (Abca1, C1qc, Ptx3, Irf5, and Pros1) and key transcription factors (Stat5). The scRNA-seq analysis showed that these hub genes were mainly expressed in microglia and macrophages which are the main cells with efferocytosis function in the brain. We then identified miR-125b-5p as a therapeutic target of IS based on the ceRNA network. Finally, we validated the phagocytosis and clearance of dead cells by efferocytosis and the expression of hub gene Abca1 in MCAO mice models.
Collapse
Affiliation(s)
- Jing Yuan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Yu-Sha Liao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Tie-Chun Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Yu-Qi Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Pei Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ya-Ning Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ding-Jun Cai
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Shu-Guang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
12
|
Wu W, Yao Z, Chen Y, Xu R, Jin C, Li X. HOTAIR, a ferroptosis-related gene, promotes malignant behavior of breast cancer via sponging miR-206. Discov Oncol 2025; 16:948. [PMID: 40442443 PMCID: PMC12122935 DOI: 10.1007/s12672-025-02791-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Ferroptosis, an iron-dependent regulated cell death modality driven by lipid peroxidation cascades, has emerged as a critical pathogenic mechanism in tumorigenesis and therapeutic resistance. The long non-coding RNA HOTAIR (HOX transcript antisense RNA), previously recognized as a key epigenetic modulator in tumor biology, orchestrates malignant phenotypes through regulation of cell cycle dynamics, proliferative signaling, and metastatic potential. Despite these advances, the mechanistic interface between HOTAIR-mediated ceRNA networks and ferroptosis regulation in breast carcinogenesis remains undefined. Bioinformatic analysis and RT-qPCR validation precisely mapped the subcellular localization and interaction networks of this regulatory axis. Key findings revealed that HOTAIR silencing functionally promotes malignant phenotypes (proliferation, migration and invasion), and mechanistically serves as a molecular sponge for miR-206, thereby de-repressing CERS2 expression to modulate ferroptosis susceptibility. Given the established correlation between HOTAIR silencing and ferroptosis in BRCA, our data suggest that pharmacological induction of ferroptosis represents a promising therapeutic paradigm for this molecular subset. This work not only deciphers a novel HOTAIR/miR-206/CERS2 axis in ferroptosis regulation but also provides translational insights for developing biomarker-driven treatment strategies in refractory breast malignancies.
Collapse
Affiliation(s)
- Wei Wu
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Zhihan Yao
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Yongxing Chen
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Rongxuan Xu
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Chenxin Jin
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Xiaofeng Li
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China.
| |
Collapse
|
13
|
Liu Y, Zeng Y, Wang S, Chen J, Wang Z, Zhao Y, Gong K, Wang G. LncRNA16 inhibits pyroptosis and promotes platinum resistance in non-small cell lung cancer by sponging miRNA1827 to regulate MBD3/GSDME expression. Cancer Cell Int 2025; 25:192. [PMID: 40413520 DOI: 10.1186/s12935-025-03812-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Platinum-based chemotherapy is the standard first-line cancer treatment. However, patients experience relapses due to chemoresistance. We found that long non-coding RNA 16 (lncRNA16) promotes platinum resistance and inhibits cell death in non-small cell lung cancer (NSCLC). However, the type of cell death inhibited by lncRNA16 remains unknown. METHODS The biological roles of lncRNA16 and microRNA 1827 (miRNA1827) in cell proliferation and colony formation were determined using functional experiments. Dual-luciferase reporter and RNA immunoprecipitation assays were performed to confirm the interactions between lncRNA16 and miRNA1827. In vivo patient-derived tumor xenograft (PDX) models were used to investigate the effects of miRNA1827 agomir on platinum resistance. RESULTS Pyroptosis was inhibited in platinum-resistant NSCLC cells. LncRNA16 contributed to the expression of methyl-CpG binding domain protein 3 (MBD3) by sponging miRNA1827, thereby inhibiting gasdermin E (GSDME) expression, which inhibited pyroptosis in platinum-resistant NSCLC. The miRNA1827 agomir repressed platinum resistance in vitro experiments and in vivo PDX models. CONCLUSION We identified a novel function of lncRNA16 in inhibiting pyroptosis and proposed an effective therapeutic drug, the miRNA1827 agomir, for chemosensitization. This study offers a potential strategy for treating patients with NSCLC, especially those with platinum resistance.
Collapse
Affiliation(s)
- Yanfang Liu
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China
| | - Yuanjun Zeng
- Department of Pathology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Sikai Wang
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China
| | - Jiangyan Chen
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China
| | - Zhouqi Wang
- Traditional Chinese Medicine, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Yang Zhao
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China
| | - Kuiyu Gong
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China.
| | - Guihua Wang
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410021, China.
| |
Collapse
|
14
|
Zheng L, Tang R, Ahmad F, Shi L, Chen X, Li J. hsa_circ_0081343 interacts with Rbm8a to inhibit NLRP3-mediated pyroptosis via the PI3K/AKT/HIF-1α pathways. Placenta 2025; 165:136-147. [PMID: 40267529 DOI: 10.1016/j.placenta.2025.04.005] [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: 12/23/2024] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION Pyroptosis at the maternal-fetal interface plays an important role in fetal growth restriction development. hsa_circ_0081343 can be an RNA-binding protein "sponge" regulating Rbm8a nuclear transportation through binding to Rbm8a. This study aimed to elucidate the regulatory mechanism underlying the interaction between hsa_circ_0081343 and Rbm8a in the FGR pyroptosis pathway. METHODS The expression levels of PI3K/AKT pathway-related components (PI3K, AKT, p-PI3K, and p-AKT), HIF-1α, NLRP3, and proinflammatory cytokines (IL-1β, IL-6, and TNF-α) were measured using RT-qPCR, Western blot, and ELISA. RNA-seq and ChIP-seq were used to identify the downstream signaling pathways of hsa_circ_0081343 and Rbm8a in HTR8-SVneo. RNA pull-down assays, Western blot, and RT-qPCR were performed to investigate the interactions between hsa_circ_0081343 and Rbm8a. RESULT The placenta of FGR exhibited considerable upregulation of NLRP3 compared to normal controls. Overexpression of hsa_circ_0081343 inhibited pyroptosis and subsequent inflammatory responses in HTR-8/SVneo cells, and these effects were reversed by Rbm8a knockdown. The integration of RNA-seq and ChIP-seq showed that the PI3K/AKT and HIF-1α pathways were the targets of hsa_circ_0081343 and Rbm8a. hsa_circ_0081343 upregulation and Rbm8a downregulation was accompanied by the inhibition of the PI3K/AKT/HIF-1α signaling pathway, whereas hsa_circ_0081343 knockdown of and Rbm8a overexpression led to the opposite effect. Moreover, Rbm8a binds to hsa_circ_0081343, flanking the intron sequence. Rbm8a overexpression significantly decreased hsa_circ_0081343 expression. CONCLUSION These results indicated that the interaction between hsa_circ_0081343 and Rbm8a regulates NLRP3-mediated pyroptosis through the PI3K/AKT/HIF-1α signaling pathway. Furthermore, Rbm8a binds to hsa_circ_0081343, flanking the intron sequence and modulating hsa_circ_0081343 formation. Our results provide a new direction for further exploration of the regulatory mechanisms of circRNA-RBPs in the pathogenesis of FGR.
Collapse
Affiliation(s)
- Linmei Zheng
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China; Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Rong Tang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Fiaz Ahmad
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University (NPU), Xi'an, 710072, Shaanxi, China
| | - Lei Shi
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Xiaoju Chen
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| | - Jing Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Wen J, Liu J, Wan L, Sun Y, Wang F. Crosstalk between N6-methyladenosine modification and ncRNAs in rheumatic diseases: therapeutic and diagnostic implications. Inflamm Res 2025; 74:79. [PMID: 40402257 DOI: 10.1007/s00011-025-02034-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND In eukaryotic cells, N6-methyladenosine (m6A) is the most prevalent RNA methylation modification and plays a fundamental role in regulating diverse biological processes through the modulation of non-coding RNA (ncRNA) expression and activity. The role of m6A modification in developing rheumatic diseases is crucial but remains inadequately studied. METHODS Characterized by pain and inflammation, rheumatic diseases like rheumatoid arthritis (RA), osteoarthritis (OA), ankylosing spondylitis (AS), and systemic lupus erythematosus (SLE) are autoimmune disorders. Recent findings emphasize the importance of m6A modifications and non-coding RNAs in the biological processes underlying rheumatic diseases. RESULTS This review elucidates the fundamental concept of m6A modification and the associated research methodologies. Subsequently, it systematically consolidates modern knowledge on the influence of m6A regulators and m6A modification-related ncRNAs on rheumatic diseases, incorporating perspectives on traditional Chinese medicine interventions. CONCLUSIONS Offering a comprehensive overview of m6A-related ncRNAs in the context of rheumatic diseases, this review proposes new therapeutic avenues by targeting m6A modification pathways.
Collapse
Affiliation(s)
- Jianting Wen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, 230031, Anhui, China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China.
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China.
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, 230031, Anhui, China.
| | - Lei Wan
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, 230031, Anhui, China
| | - Yue Sun
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, 230031, Anhui, China
| | - Fanfan Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
- Institute of Rheumatology, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui Province, China
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, 230031, Anhui, China
| |
Collapse
|
16
|
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
|
17
|
Chen D, Zhang QT, Li WJ, Han HL, Smagghe G, Yan Y, Jiang HB, Wang JJ, Wei D. The competing endogenous RNA lnc94641-miR957-3p mediates male fertility in Zeugodacus cucurbitae Coquillett. PEST MANAGEMENT SCIENCE 2025. [PMID: 40371678 DOI: 10.1002/ps.8874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Insect spermatogenesis is a complex process. Numerous genes are involved in sperm motility, which is crucial for male fertility. Few long non-coding RNAs (lncRNAs) in the testis regulate insect spermatogenesis. We previously identified 364 testis-enriched lncRNAs in the globally invasive pest Zeugodacus cucurbitae Coquillett. One of these lncRNAs, lnc94641, is abundantly expressed in the testis; however, its role in spermatogenesis remains unknown. RESULTS Suppression of lnc94641 expression led to a 60% decrease in spermatozoa count and a 29% decrease in offspring hatchability. A microRNA (miRNA), miR-957-3p, was experimentally demonstrated to bind to lnc94641 competitively. miR-957-3p overexpression recapitulated reproductive defect phenotypes similar to those caused by lnc94641 knockdown. Furthermore, target gene predictions combined with quantitative reverse transcription PCR, RNA pull-down, and dual luciferase reporter assays confirmed that miR-957-3p targets voltage-gated potassium channel 5 (VGKC5) and odorant receptor 85c (OR85c), elucidating a functional lncRNA-miRNA-mRNA competing endogenous RNA (ceRNA) regulatory axis. Fluorescence in situ hybridization (FISH) assays demonstrated the co-localization of lnc94641, miR-957-3p, and VGKC5/OR85c in the mature and transformed regions of the testes. Suppression of VGKC5/OR85c expression resulted in a 68% and 50% decrease in spermatozoa number and an 18% and 21% decrease in offspring hatchability, mirroring the phenotype observed with lnc94641-silencing, thereby reinforcing the mechanistic coherence of this regulatory network. CONCLUSION These results revealed a ceRNA axis mediated by 'lnc94641-miR957-3p-VGKC5/OR85c' involved in spermatogenesis that impairs male fertility in the melon fly. Molecular perturbations (lncRNA knockdown or miRNA overexpression) consistently impair sperm production and offspring viability by dysregulating ion channels and chemosensory genes. This mechanistically resolved pathway, centered on the core components VGKC5 and OR85c, revealed conserved reproductive vulnerabilities that could enable the targeted genetic control of this agricultural pest. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Dong Chen
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Qi-Tong Zhang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Wei-Jun Li
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- School of Agricultural Sciences, Jiangxi Agricultural University, Nanchang, China
| | - Hong-Liang Han
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Guy Smagghe
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Ying Yan
- Department of Insect Biotechnology in Plant Protection, Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Hong-Bo Jiang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Jin-Jun Wang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Dong Wei
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| |
Collapse
|
18
|
Wang M, Chen Y, Yang W, Li X, Liu G, Wang X, Liu S, Gao G, Meng F, Kong F, Sun D, Qin W, Dong B, Zhang J. Bioinformatics analysis of circular RNAs associated with atrial fibrillation and their evaluation as predictive biomarkers. Hum Genomics 2025; 19:52. [PMID: 40355900 PMCID: PMC12070608 DOI: 10.1186/s40246-025-00760-7] [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: 10/15/2024] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Circular noncoding RNAs (circRNAs) are implicated in many human diseases, but their role in atrial fibrillation (AF) is poorly understood. In this study, we performed bioinformatics analysis of circRNA sequencing data to identify AF-related circRNAs. METHODS Left atrial appendage (LAA) samples were obtained from patients with valvular heart disease and were categorised into the sinus rhythm (SR; n = 4) and AF (n = 4) groups. CircRNA sequencing analysis was performed to identify differentially expressed (DE) circRNAs in AF patients. Functional enrichment analysis of DE circRNAs was performed to identify enriched Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. RESULTS We identified 3338 DE circRNAs, including 2147 upregulated and 1191 downregulated circRNAs, in AF patients. A ceRNA network of 16 DE circRNAs, 11 DE miRNAs, and 15 DE mRNAs was constructed. Functional enrichment analyses revealed that the AF-related DE circRNAs were enriched in response to vitamin D, the potassium channel complex, delayed rectifier potassium channel activity, osteoclast differentiation, primary immunodeficiency, endocrine and other factor-regulated calcium reabsorption and other processes. ROC curve analysis identified circRNA_00324, circRNA_17225, circRNA_16305, circRNA_10233, circRNA_05499, circRNA_03183, circRNA_14211, and circRNA_18422 as potential predictive biomarkers for distinguishing AF patients from SR patients, with AUC values of 0.9138, 0.7370, 0.8526, 0.6803, 0.8163, 0.8662, 0.7664, and 0.9320, respectively. CONCLUSIONS In this study, we constructed an AF-related ceRNA network and identified eight circRNAs as potential predictive biomarkers of AF.
Collapse
Affiliation(s)
- Manman Wang
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuanyuan Chen
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Weiwei Yang
- Department of Medical Record, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xiangting Li
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Genli Liu
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xin Wang
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Shuai Liu
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Ge Gao
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Fanhua Meng
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Feifei Kong
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Dandan Sun
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Wei Qin
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Jinguo Zhang
- Shandong Provincial Key Medical and Health Discipline of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Jining Key Laboratory of Precise Therapeutic Research of Coronary Intervention, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
19
|
Wang M, Zheng S, Zhang Y, Zhang J, Lai F, Zhou C, Zhou Q, Li X, Li G. Transcriptome analysis reveals PTBP1 as a key regulator of circRNA biogenesis. BMC Biol 2025; 23:127. [PMID: 40350413 PMCID: PMC12067716 DOI: 10.1186/s12915-025-02233-8] [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: 05/27/2024] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are a class of non-coding RNAs generated through back splicing. High expression of circRNAs is often associated with numerous abnormal cellular biological processes. However, the regulatory factors of circRNAs are not fully understood. RESULTS In this study, we identified PTBP1 as a crucial regulator of circRNA biogenesis through a comprehensive analysis of the whole transcriptome profiles across 10 diverse cell lines. Knockdown of PTBP1 led to a significant decrease in circRNA expression, concomitant with a distinct reduction in cell proliferation. To investigate the regulatory mechanism of PTBP1 on circRNA biogenesis, we constructed a minigene reporter based on SPPL3 gene. The results showed that PTBP1 can bind to the flanking introns of circSPPL3, and the mutation of PTBP1 motif impedes the back splicing of circSPPL3. Subsequently, to demonstrate that this observation is not an exception, the comprehensive regulatory effects of PTBP1 on circRNAs were confirmed by miniGFP, reflecting the necessity of the binding site in the flanking introns. Analysis of data from clinical samples showed that both PTBP1 and circRNAs exhibited substantial upregulation in acute myeloid leukemia, further demonstrating a potential role for PTBP1 in promoting circRNA biogenesis under in vivo conditions. Competitive endogenous RNA (ceRNA) network revealed that PTBP1-associated circRNAs participated in biological processes associated with cell proliferation. CONCLUSIONS In summary, our study is the first to identify the regulatory effect of PTBP1 on circRNA biogenesis and indicates a possible link between PTBP1 and circRNA expression in leukemia.
Collapse
Affiliation(s)
- Mohan Wang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shanshan Zheng
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yan Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jingwen Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fuming Lai
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Cong Zhou
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiangwei Zhou
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xingwang Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Guoliang Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Farming Technology for Agricultural Animals, 3D Genomics Research Center, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
20
|
Wang D, Di D, Jiang B, Wang Y, Jiang Z, Jing Y, Wu H, Xin S. Revealing the multiple faces of LRG1: gene expression, structure, function, and therapeutic potential. J Adv Res 2025:S2090-1232(25)00342-X. [PMID: 40368176 DOI: 10.1016/j.jare.2025.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 05/05/2025] [Accepted: 05/11/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND As the structural characterization of the Leucine-rich alpha-2-glycoprotein 1 (LRG1) protein progresses, its functional diversity has been increasingly unveiled, highlighting its clinical relevance in elucidating disease pathogenesis and identifying potential therapeutic targets. AIM OF REVIEW Grounded in structural biology principles, this review systematically examines the regulatory mechanisms, pathological functions, and intervention strategies associated with LRG1, providing a theoretical foundation for translating these insights into clinical drug therapies. KEY SCIENTIFIC CONCEPTS OF REVIEW LRG1, distinguished by its leucine-rich repeat motifs, plays a pivotal role in various physiological and pathological processes. This review presents a comprehensive analysis of LRG1's multifaceted characteristics and its implications in disease. Initially, the regulatory mechanisms modulating LRG1 gene expression are detailed, encompassing both transcriptional and post-transcriptional controls. The structural attributes and distributions of LRG1 are subsequently outlined, with an emphasis on the functional relevance of its leucine-rich repeat motifs. Furthermore, the review elaborates on the molecular interactions through which LRG1 engages with distinct receptors, triggering downstream signaling pathways involved in pathological processes. Finally, current therapeutic approaches targeting LRG1 and its receptors are summarized, alongside prospective research avenues for innovative therapeutic development.
Collapse
Affiliation(s)
- Ding Wang
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Di Di
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China; Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110032, China
| | - Bo Jiang
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Yunlong Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Zhenqi Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China
| | - Yuchen Jing
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang City, Liaoning Province 110122, China; Scientific Experimental Center, School of Pharmacy, China Medical University, Shenyang City, Liaoning Province 110122, China.
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital, China Medical University, Shenyang City, Liaoning Province 110001, China; Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang City, Liaoning Province 110001, China; Regenerative Medicine Research Center of China Medical University, Shenyang City, Liaoning Province 110001, China.
| |
Collapse
|
21
|
Xie H, Lu Y, Pan J, Zeng H, Zhang Z, Yin J, Zhu J, Luo B, Guo D, Wu C, Zeng C, Shao Y, Bai X, Cai D, Zhang H. MiR-335-5p Escaped from CircKIAA0586 Adsorption Contributes to Mechanical Overloading-Induced Cartilage Degeneration by Targeting Lymphoid-Specific Helicase. RESEARCH (WASHINGTON, D.C.) 2025; 9:0694. [PMID: 40342810 PMCID: PMC12059312 DOI: 10.34133/research.0694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025]
Abstract
Mechanical overload is a critical contributor to cartilage degeneration in osteoarthritis (OA) pathogenesis. Circular RNA (circRNA) is expected to provide a long-lasting therapy for OA. However, the involvement of the circRNA-associated competitive endogenous RNA network in chondrocyte senescence induced by mechanical overloading remains unestablished. A mechanical overloading-induced chondrocyte senescence model in human primary chondrocytes is constructed, and differences in the expression of circRNAs and miRNAs were analyzed. The biological roles of circKIAA0586/miR-335-5p in chondrocyte senescence and OA progression under mechanical overloading and its downstream targets were determined using gain- and loss-of-function experiments in various biochemical assays in human chondrocytes. The in vivo effects of circKIAA0586 overexpression were also determined in destabilization of the medial meniscus (DMM) OA mice and aged spontaneous OA mice. The mechanical overloading-induced chondrocyte senescence was aggravated by miR-335-5p or circKIAA0586 knockdown. Accumulated DNA damage response was observed following mechanical overloading, which reduced after miR-335-5p inhibition or circKIAA0586 supplementation. MiR-335-5p was regulated by circKIA0586 adsorption. HELLS was prominently down-regulated following mechanical overloading treatment. Moreover, miR-335-5p bound to lymphoid-specific helicase (HELLS) mRNA during mechanical overloading was demonstrated to mediate the nonhomologous end joining (NHEJ) pathway, thereby inducing DNA damage and senescence. In addition, the senescence delaying and cartilage protective functions of circKIAA0586 and HELLS were validated in DMM OA mice and aged spontaneous OA mice. Our findings suggest that miR-335-5p, which escapes circKIAA0586 adsorption, facilitates mechanical overloading-induced chondrocyte senescence and OA progression by impairing the NHEJ pathway through HELLS inhibition. Overall, targeting circKIAA0586/miR-335-5p/HELLS signaling provides a novel therapeutic approach for OA.
Collapse
Affiliation(s)
- Haoyu Xie
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Yuheng Lu
- Department of Rehabilitation Medicine, Xijing Hospital,
Fourth Military Medical University, Xi ’an 710032, China
| | - Jianying Pan
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Hua Zeng
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Zhicheng Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Jianbin Yin
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Jinjian Zhu
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Bingsheng Luo
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Dong Guo
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Chunyu Wu
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Chun Zeng
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Yan Shao
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences,
Southern Medical University, Guangzhou 510515, China
| | - Daozhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| | - Haiyan Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- The Third School of Clinical Medicine,
Southern Medical University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou 510630, China
| |
Collapse
|
22
|
Periyasamy P, Singh S, Oladapo A, Kannan M, Buch S. Role of lncRNA Xist-miR-124-CCL2 axis in HIV Tat-mediated microglial activation and neuroinflammation. Front Immunol 2025; 16:1558842. [PMID: 40406114 PMCID: PMC12094947 DOI: 10.3389/fimmu.2025.1558842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/11/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction HIV proteins, such as the Transactivator of transcription (Tat), mediate neuroinflammation in the central nervous system by promoting the release of pro-inflammatory cytokines and chemokines. Long noncoding RNAs (lncRNAs) regulate gene expression by sponging microRNAs (miRs), but their role in HIV Tat-mediated microglial activation remains poorly understood. This study aimed to investigate the involvement of the lncRNA Xist-miR-124-CCL2 axis in HIV Tat-exposed microglial cells. Methods Mouse primary microglial cells were exposed to HIV Tat, and the expression of lncRNA Xist, miR-124, and CCL2 was evaluated using qPCR, Western blotting, and ELISA. Dual-luciferase reporter and Argonaute immunoprecipitation assays were used to confirm molecular interactions. Functional experiments involved lncRNA Xist silencing and miR-124 overexpression. In vivo validation was performed using doxycycline-inducible HIV Tat transgenic mice. Results HIV Tat significantly upregulated lncRNA Xist and downregulated miR-124 expression in mouse primary microglial cells. miR-124 was identified as a direct target of lncRNA Xist and the 3'-UTR of CCL2. Silencing lncRNA Xist or overexpressing miR-124 reduced HIV Tat-induced CCL2 expression and microglial activation. In vivo studies corroborated these findings, with doxycycline-fed iTat mice showing elevated lncRNA Xist and CCL2 levels and reduced miR-124 expression in the frontal cortex. Discussion Our findings identify a novel regulatory axis whereby HIV Tat-induced upregulation of lncRNA Xist sponges miR-124, leading to CCL2 overexpression and microglial activation. Targeting the lncRNA Xist-miR-124-CCL2 pathway may represent a promising therapeutic strategy to mitigate neuroinflammation associated with NeuroHIV.
Collapse
|
23
|
Shang Z, Jin S, He Y, Zhu Y, Zhang H, Wu J, Hong Z, Ye D. Comprehensive analysis of the LINC01122/TPD52 axis as a predictive biomarker in prostate adenocarcinoma. Sci Rep 2025; 15:16122. [PMID: 40341647 PMCID: PMC12062280 DOI: 10.1038/s41598-025-98219-1] [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: 09/27/2024] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Prostate cancer (PCa) ranks among the most prevalent malignant tumors worldwide. The pivotal role of competitive endogenous RNA (ceRNA) regulatory networks in numerous cancer types has been underscored. However, the specific characteristics of the ceRNA network in PCa remained unknown. This study aims to elucidate the ceRNA regulatory network associated with phosphatase and tensin homolog (PTEN) and to identify potential prognostic markers for PCa. The Cancer Genome Atlas (TCGA) database was employed to extract the expression patterns of long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs). LINC01122-hsa-miR-34c-5p/hsa-miR-449a-TPD52 ceRNA network regarding the prognosis of PCa was explored via bioinformatics analysis. Through correlation analysis, we investigated the LINC01122/TPD52 axis within the ceRNA network, identifying it as a significant clinical prognostic marker for PCa. Subsequent analyses indicated that hypomethylation was responsible for the abnormal upregulation of the LINC01122/TPD52 axis. Furthermore, immune infiltration analysis revealed the impact of the LINC01122/TPD52 axis on the tumor immune microenvironment and the progression of PCa. Finally, a nomogram was constructed to forecast the 1-year, 3-year, and 5-year survival probabilities of PCa patients. In summary, our study demonstrates the significant role of the ceRNA-based LINC01122/TPD52 axis in the progression of PCa and its correlation with prognosis.
Collapse
Affiliation(s)
- Zhi Shang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yandong He
- Department of Urology, South Hospital, The Sixth People's Hospital of Shanghai, Shanghai, 201499, China
| | - Yiping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Zhe Hong
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
24
|
Li P, Zhang C, Yin W, Tao M, Niu Z, Cui Y, Wu D, Gao F. From bone marrow mesenchymal stem cells to diseases: the crucial role of m 6A methylation in orthopedics. Stem Cell Res Ther 2025; 16:228. [PMID: 40329380 PMCID: PMC12057228 DOI: 10.1186/s13287-025-04364-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Elucidating the molecular mechanisms underlying orthopedic diseases is crucial for guiding therapeutic strategies and developing innovative interventions. N6-methyladenosine (m6A)-an epitranscriptomic modification-has emerged as a key regulator of cellular fate and tissue homeostasis. Specifically, m6A plays a pivotal role in several RNA biological processes such as precursor RNA splicing, 3'-end processing, nuclear export, translation, and stability. Recent advancements indicate that m6A methylation regulates stem cell proliferation and osteogenic differentiation by modulating various signaling pathways. Extensive research has shown that abnormalities in m6A methylation contribute significantly to the onset and progression of various orthopedic diseases such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), and bone tumors. This review aims to summarize the key proteases involved in m6A methylation and their functions. The detailed mechanisms by which m6A methylation regulates osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) through direct and indirect ways are also discussed, with a focus on specific molecular pathways. Finally, this review analyzes the roles and mechanisms of m6A modification in the development and progression of multiple orthopedic diseases, offering a comprehensive understanding of the pathophysiology of these conditions and proposing new directions and molecular targets for innovative treatment strategies.
Collapse
Affiliation(s)
- Peng Li
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Chu Zhang
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Wen Yin
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Mijia Tao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Zhipeng Niu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yutao Cui
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Dankai Wu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Feng Gao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| |
Collapse
|
25
|
Guo Y, Gong Y, Wu M, Ji M, Xie F, Chen H, Niu H, Tang C. CircRNAs in the tumor microenvironment: new frontiers in cancer progression and therapy. Crit Rev Oncol Hematol 2025; 212:104754. [PMID: 40320223 DOI: 10.1016/j.critrevonc.2025.104754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
The tumor microenvironment (TME), a dynamic ecosystem which including immune cells, cancer-associated fibroblasts (CAFs), endothelial cells, pericytes and acellular components, is orchestrating cancer progression through crosstalk between malignant cells and stromal components and increasingly recognized as a therapeutic frontier. Within this intricate network, circular RNAs (circRNAs) have emerged as pivotal regulators due to their unique covalently closed structures, which confer exceptional stability and multifunctional capabilities. This regulation is mediated through multiple mechanisms, such as acting as microRNA (miRNA) sponges, interacting with proteins, and, in certain instances, encoding functional peptides. The interaction between circRNAs and the TME not only affects cancer growth and metastasis but also influences immune evasion and therapeutic resistance. Elucidating the mechanisms by which circRNAs orchestrate these interactions is essential for identifying novel diagnostic biomarkers and developing effective therapeutic strategies. Such insights are expected to bridge gaps in current cancer biology, offering promising avenues for precision oncology and ultimately improving clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Yipei Guo
- School of Elderly Care Services and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanxun Gong
- Guangxi Key Laboratory for Preclinical and Translational Research on Bone and Joint Degenerative Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Man Wu
- School of Clinical Medicine, Wannan Medical College, Wuhu 241002, China
| | - Mengjia Ji
- School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Fei Xie
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao 266013, China.
| | - Hao Chen
- Department of Pathology, Wannan Medical College, Wuhu 241002, China; Postdoctoral Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China.
| | - Haitao Niu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao 266013, China.
| | - Chao Tang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
26
|
Zhang B, Matsumoto Y. Integrated Network Analysis Decipher ZNF384-Related miR-20b-5p and miR-424-5p in Colon Adenocarcinoma. Cancer Rep (Hoboken) 2025; 8:e70233. [PMID: 40405535 PMCID: PMC12098962 DOI: 10.1002/cnr2.70233] [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: 05/20/2024] [Revised: 04/09/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND ZNF384 is a C2H2-type zinc finger protein (ZNF) which is implicated in DNA double-strand break (DSB) repair through the classical non-homologous end-joining (cNHEJ) pathway. AIMS To clarify the regulatory mechanisms involving ZNF384 in colon adenocarcinoma (COAD). METHODS AND RESULTS First, we conducted a differential expression gene (DEG) analysis of mRNA and lncRNA using TCGA-COAD RNA-Seq data. We also identified ZNF384-related mRNAs through Pearson's correlation coefficient calculation and conducted weighted gene co-expression network analysis (WGCNA) for these genes, leading to the identification of a cluster of 331 genes with strongly positive correlation to tumor, 84 of which overlapped with DEGs. Gene functional analysis showed enrichment of genes in DNA repair, replication fork, and cell cycle checkpoint signaling pathways. Protein-protein interaction (PPI) network analysis of these 84 genes led to the identification of the top 20 key mRNAs. Then we employed three machine learning methods to refine our selection of candidate genes from these intersecting mRNAs. We constructed a competitive endogenous RNA (ceRNA) network and identified two significant intersecting miRNAs, miR-20b-5p and miR-424-5p, which have been shown to act as a tumor suppressor gene and an oncogene, respectively. Additionally, we found that KIF14 and KIF18B are regulated by these two miRNAs in this ceRNA network, particularly in DNA damage repair and cell cycle. Finally, validation using an external dataset from the GEO database confirmed their expression patterns. CONCLUSION The current study clarifies the mechanisms of how miR-20b-5p and miR-424-5p work in colon cancer and underscores their predictive capabilities in colon cancer.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Transdisciplinary Science and EngineeringSchool of Environment and Society, Institute of Science TokyoTokyoJapan
- Laboratory for Zero‐Carbon Energy, Institute of Integrated ResearchInstitute of Science TokyoTokyoJapan
| | - Yoshihisa Matsumoto
- Department of Transdisciplinary Science and EngineeringSchool of Environment and Society, Institute of Science TokyoTokyoJapan
- Laboratory for Zero‐Carbon Energy, Institute of Integrated ResearchInstitute of Science TokyoTokyoJapan
| |
Collapse
|
27
|
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
|
28
|
Guan H, Hu Q, Wan L, Wang C, Xue Y, Feng N, Zhao C, Chen M, You Z. LINC00908 Inactivates Wnt/β-Catenin Signaling Pathway to Inhibit Prostate Cancer Cell Stemness via Upregulating GSK3B and FBXW2. Cancer Med 2025; 14:e70887. [PMID: 40344383 PMCID: PMC12061849 DOI: 10.1002/cam4.70887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/31/2024] [Accepted: 09/10/2024] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Chemotherapy and androgen-deprivation treatment are the curative approaches utilized to suppress prostate cancer (PCa) progression. However, drug resistance and metastasis are extensive and hard to overcome even though remarkable progress has been made in recent decades. The cancer stem cell-related theoretical model explains the distinct molecular characteristics of cancer, its relapse, metastasis, and drug resistance. Meanwhile, noncoding RNA functions in the formation of drug resistance and metastasis in most cancers. The long intergenic nonprotein coding RNA 908 (LINC00908) has been reported to restrain cell proliferation, migration, and invasion of some cancers like triple-negative breast cancer, diffuse large B-cell lymphoma, PCa, and so on. However, its role in stemness for PCa remains unclear. METHODS We delved into the impact of LINC00908 in PCa cell stemness and the principal molecular mechanism. Then, the impact of LINC00908 on PCa cell stemness and its corresponding mechanism was explored by using functional assays and bioinformatics evaluation. RESULTS We found that LINC00908 was low-expressed in PCa cells, and it exerted suppressive functions in PCa cell stemness and tumor growth. Additionally, we revealed that LINC00908 down-regulation was mediated by the HDAC2-p300-YY1 transcription complex. Moreover, LINC00908 up-regulated glycogen synthase kinase 3 beta (GSK3B) via sponging miR-3179. Meanwhile, LINC00908 deployed DEAD-box helicase 3 X-linked (DDX3X) to facilitate the stabilization of F-box and WD repeat domain containing 2 (FBXW2) mRNA. Importantly, LINC00908 enhanced GSK3B and FBXW2 expression to induce the ubiquitination of β-catenin protein, leading to Wnt pathway inactivation. CONCLUSION These results reveal that LINC00908 inhibits PCa cell stemness via inactivating the GSK3B/FBXW2-regulated Wnt pathway, which might enrich people's knowledge of PCa stemness and provide some new potential biomarkers for PCa.
Collapse
Affiliation(s)
- Han Guan
- Department of UrologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Qiang Hu
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Lilin Wan
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Can Wang
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Yifeng Xue
- Department of UrologyChangzhou JinTan First People's HospitalChangzhouChina
| | - Ninghan Feng
- Department of UrologyWuxi No. 2 Hospital, Nanjing Medical UniversityWuxiChina
| | - Chenggui Zhao
- Department of LaboratoryAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Ming Chen
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| | - Zonghao You
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
- Institute of Medical Phenomics ResearchAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
| |
Collapse
|
29
|
Wu C, Gao Y, Jin Z, Huang Z, Wang H, Lu S, Guo S, Zhang F, Zhang J, Huang J, Tao X, Liu X, Zhang X, You L, Li Q, Wu J. PTPRG-AS1 regulates the KITLG/KIT pathway through the ceRNA axis to promote the malignant progression of gastric cancer and the intervention effect of Compound Kushen injection on it. Pharmacol Res 2025; 215:107743. [PMID: 40250508 DOI: 10.1016/j.phrs.2025.107743] [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: 03/06/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Gastric cancer (GC) is a common malignant tumor with high mortality, recurrence, and metastasis rates. Compound Kushen injection (CKI) combination chemotherapy has been clinically used for the treatment of GC in China for many years, but its underlying mechanisms of action remain unclear. Recent reports have highlighted the important role of the competing endogenous RNA (ceRNA) mechanism of noncoding RNA (ncRNA) and messenger RNA (mRNA) formation in GC and other tumors. This study aimed to investigate the effects of CKI on GC from the ceRNA perspective. We confirmed the inhibitory effect of CKI on GC in mouse models and cell lines. By examining the GC cell lines sensitive to CKI treatment, we developed the CNScore method to analyze the ceRNA network, revealing that the CKI-GC ceRNA network promotes GC proliferation and metastasis through the PTPRG-AS1/hsa-miR-421/KITLG axis. Finally, we constructed GC cell models with PTPRG-AS1 overexpression or knockdown and GC liver metastasis models and found that PTPRG-AS1 can sponge hsa-miR-421, releasing KITLG and promoting GC proliferation and metastasis through the KITLG/KIT pathway. Taken together, CKI can suppress these malignant phenotypes by regulating the PTPRG-AS1/hsa-miR-421/KITLG axis.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yifei Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhengsen Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Haojia Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoyu Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Leiming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang Province 310022, China.
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
30
|
Wei Z, Li X, Zhou J, Zhou Y, Xiao Z, Yang Q, Liu X, Peng Y, Yang Y, Ding Y, Ru Z, Wang Y, Yang M, Yang X. Inhibition of miRNA-365-2-5p Targeting SIRT1 Regulates Functions of Keratinocytes to Enhance Wound Healing. FASEB J 2025; 39:e70560. [PMID: 40261275 DOI: 10.1096/fj.202401124rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
The development of drugs to accelerate wound healing is an important area of clinical research. Recent advancements have highlighted the prospects of microRNAs as therapeutic targets for various disorders, although their involvement in mice wound healing remains unclear. Peptides have been proved to be unique and irreplaceable molecules in the elucidation of competing endogenous RNAs mechanisms (ceRNA) involved with skin wound healing. In the present work, CyRL-QN15, a peptide characterized by its minimal length and maximal wound healing efficacy, was applied as a probe to explore the ceRNA mechanism in regard to accelerated wound healing. Results showed that the use of CyRL-QN15 significantly reduced the expression of miRNA-365-2-5p at the wound in mice. In mouse keratinocytes, miRNA-365-2-5p inhibition increased SIRT1 and FOXO1 protein expression and decreased STAT2 protein expression, promoting cell proliferation, migration, and reducing inflammatory factors. Similarly, inhibiting miRNA-365-2-5p at mouse wounds promoted Full-thickness injured skin wounds healing, increased SIRT1 and FOXO1 protein expression, decreased STAT2 protein expression, and reduced inflammatory factors. Overall, these findings demonstrate that miRNA-365-2-5p serves a crucial function in the biological processes underlying cutaneous wound healing in mice, offering a novel target for future therapeutic interventions in wound healing.
Collapse
Affiliation(s)
- Ziqi Wei
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xingguo Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyi Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxuan Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoxun Xiao
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Qian Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xin Liu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Peng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuliu Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yujing Ding
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zeqiong Ru
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, Yunnan, China
| | - Meifeng Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
31
|
Lin W, Huang L, Ou Z, Xuan Y, Zhu D, Zhang Q, Xu E. N6-methyladenosine-modified circ_0000517 promotes non-small cell lung cancer metastasis via miR-1233-3p/CDH6 axis. J Mol Histol 2025; 56:139. [PMID: 40278968 DOI: 10.1007/s10735-025-10421-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Circular RNAs (circRNAs) exhibit dysregulation in non-small cell lung cancer (NSCLC) and regulate the malignant biological behavior of NSCLC. The N6-methyladenosine (m6A) modification of circRNAs plays a critical role in multiple malignant tumors, and their biological relevance in NSCLC is unclear. Herein, this study was conducted to investigate the novel functional mechanism of highly expressed circ_0000517 in NSCLC by developing in vitro experiments. We found that circ_0000517 was upregulated in NSCLC tissues and cells, and that increased circ_0000517 expression was associated with m6A modification. Biologically, silenced circ_0000517 hindered the proliferation, colony formation, migration and invasion of NSCLC cells in vitro, and also suppressed the EMT-related process. Mechanistically, highly expressed circ_0000517 activated CDH6 expression and EMT evolution through sponging miR-1233-3p. Notably, miR-1233-3p had the opposite effect and reversed the promotion effect of circ_0000517 on the malignant biological behavior of NSCLC cells. Our study revealed a promising novel endogenous regulatory network that m6A-modified circ_0000517 accelerated malignant evolution of NSCLC by targeting the miR-1233-3p/CDH6 axis.
Collapse
Affiliation(s)
- Weixian Lin
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510510, Guangdong, China
| | - Lifang Huang
- Department of Surgery, Huizhou Hospital, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510510, Guangdong, China
| | - Zhu'an Ou
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, PLA, 510010, Guangzhou, China
| | - Yiwen Xuan
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, PLA, 510010, Guangzhou, China
| | - Daoqi Zhu
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, PLA, 510010, Guangzhou, China
| | - Qipeng Zhang
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, PLA, 510010, Guangzhou, China
| | - Enwu Xu
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, PLA, 510010, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China.
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, No.111 Liuhua Road, 510010, Guangzhou, China.
| |
Collapse
|
32
|
Li Santi A, Alfieri M, Meo L, Ragno P. Let-7 Family microRNAs Regulate the Expression of the Urokinase-Receptor in Acute Myeloid Leukemia Cells. Cells 2025; 14:623. [PMID: 40358147 PMCID: PMC12071396 DOI: 10.3390/cells14090623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/31/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The urokinase-receptor (uPAR) exerts multiple functions supporting most cancer hallmarks. Increased uPAR expression is associated with an unfavorable prognosis in several cancer types, including hematologic malignancies. We previously reported that three oncosuppressor microRNAs (miRNAs) can target the 3'untranslated region (3'UTR) of the uPAR mRNA and that uPAR mRNA is a competitive endogenous RNA (ceRNA) able to recruit oncosuppressor miRs, thus impairing their activity. We now show that uPAR mRNA can also be targeted by oncosuppressor members of the let-7 miRNA family in acute myeloid leukemia (AML) cell lines. Indeed, let-7a, let7d and let-7g directly target the 3'UTR of uPAR mRNA, thus down-regulating uPAR expression. These let-7 miRNAs are expressed in KG1 and U937 AML cells; their levels are high in KG1 cells, which express low uPAR levels, and low in the U937 cell line, expressing high levels of uPAR. Overexpression of these miRNAs down-regulates uPAR expression and impairs the adhesion to fibronectin and migration of U937 cells, without affecting their proliferation. Accordingly, the overexpression of specific inhibitors targeting these let-7 miRNAs efficiently increases uPAR expression in KG1 cells. These results indicate that selected let-7 miRNAs regulate uPAR expression and impair the adhesion and migration of AML cells.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Receptors, Urokinase Plasminogen Activator/genetics
- Receptors, Urokinase Plasminogen Activator/metabolism
- Cell Line, Tumor
- 3' Untranslated Regions/genetics
- Gene Expression Regulation, Leukemic
- Cell Movement/genetics
- Cell Proliferation/genetics
- U937 Cells
- Cell Adhesion/genetics
- Gene Expression Regulation, Neoplastic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Down-Regulation/genetics
Collapse
Affiliation(s)
- Anna Li Santi
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| | - Mariaevelina Alfieri
- Clinical Pathology, Pausilipon Hospital, A.O.R.N Santobono-Pausilipon, 80129 Naples, Italy;
| | - Luigia Meo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (Salerno), Italy
| |
Collapse
|
33
|
He P, Jiang H, Zhu J, Hu M, Song P. Identification and validation of the inflammatory response-related LncRNAs as diagnostic biomarkers for acute ischemic stroke. Sci Rep 2025; 15:13818. [PMID: 40258919 PMCID: PMC12012103 DOI: 10.1038/s41598-025-98101-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
Ischemic stroke is one of the leading causes of deaths and disability, which is linked to inflammation. In this study, we aimed to identify inflammation-related lncRNAs as diagnostic biomarkers of acute ischemic stroke (AIS). A competing endogenous RNAs (ceRNA) network was established through whole transcriptome analysis. Gene expression datasets from the GEO database were analyzed to identify differentially expressed genes (DEGs), miRNAs and lncRNAs. Inflammation-related DEGs were determined through the intersection of the DEGs of the inflammation-related gene set from Genecards. Multiple databases like lncBase and Targetscan were analyzed to establish a ceRNA network. Several hub genes and sub-networks were obtained from a protein to protein (PPI) network. In addition, the candidate lncRNAs derived from the subnetwork were validated using mice MCAO model and clinical samples. Finally, a network comprising 20 lncRNAs, 26 miRNAs, and 43 inflammatory genes was analyzed, leading to the identification of MALAT1, SNHG8, and GAS5 as potential diagnostic biomarkers. Knockdown of MALAT1 and GAS5 resulted in decreased neurological severity score and inflammation response in mice MCAO model, indicating that these genes were significant diagnostic biomarkers for distinguishing AIS from healthy controls. These findings show that circulating MALAT1 and GAS5 have the potential to serve as clinical diagnostic biomarkers of AIS associated with inflammation.
Collapse
Affiliation(s)
- Peidong He
- Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hongxiang Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang Distict, Wuhan, 430060, Hubei Province, China
| | - Jiangrui Zhu
- Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Ping Song
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang Distict, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
34
|
Xu S, He L, Chen Y, Lin T, Tang L, Wu Y, He Y, Sun X. Clinical implications of miR-195 in cancer: mechanisms, potential applications, and therapeutic strategies. J Cancer Res Clin Oncol 2025; 151:148. [PMID: 40261408 PMCID: PMC12014848 DOI: 10.1007/s00432-025-06195-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: 02/04/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
This review explores the dual role of miR-195 in cancer, acting as both a tumor suppressor and, in specific contexts, a tumor promoter. It highlights its molecular mechanisms, focusing on key signaling pathways such as Wnt-1/β-catenin, VEGF/VEGFR, and PI3K/AKT/mTOR, as well as its involvement in competitive gene regulation. The clinical potential of miR-195 in cancer screening, diagnosis, prognosis, and therapy is examined, particularly its ability to enhance therapeutic efficacy and reduce recurrence risk when combined with chemotherapy or immunotherapy. Despite these promising aspects, challenges such as precise regulation, efficient delivery systems, and clinical translation remain. Future research should prioritize advancing miR-195's integration into personalized medicine, immunotherapy, and novel delivery technologies, aiming to establish it as a reliable biomarker and therapeutic target for improved cancer care.
Collapse
Affiliation(s)
- Shuli Xu
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lan He
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Yan Chen
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ting Lin
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Le Tang
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yonghui Wu
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yingchun He
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, 410208, China.
- Hunan Provincial Key Lab for the Prevention, Treatment of Ophthalmology and Otolaryngology Diseases With Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Xiaofeng Sun
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
35
|
Li S, Cui Z, Gao M, Shan Y, Ren Y, Zhao Y, Wang D, Meng T, Liu H, Yin Z. Hsa_circ_0072088 promotes non-small cell lung cancer progression through modulating miR-1270/TOP2A axis. Cancer Cell Int 2025; 25:156. [PMID: 40259294 PMCID: PMC12010575 DOI: 10.1186/s12935-025-03749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
According to the data released by the International Agency for Research on Cancer (IARC) in 2020, lung cancer ranks second among newly diagnosed malignant tumors globally. As a special class of non-coding RNA, circRNA has become a new hotspot in the field of biomarker research. With the continuous deepening of molecular-level investigations, the underlying mechanisms of circRNA are being gradually unveiled. The more widely studied mechanism is the competitive endogenous RNA mechanism of circRNA. Studies related to circRNA expression were searched in GEO database and statistically analyzed using the "limma" package and weighted gene co-expression network analysis. The expression of circRNA, microRNA and mRNA in cells and tissues were examined via qRT-PCR. MTS assay was used to measure cell proliferation, Transwell assay was used to measure cell migration, and apoptosis assay was carried out to detect cell apoptosis. Additionally, a dual-luciferase reporter assay was further executed to explore the targeted binding relationships between circRNA-microRNA and microRNA-mRNA. It was discovered that hsa_circRNA_103809 was differentially highly expressed in non-small cell lung cancer cells, whereas miR-1270 was differentially lowly expressed. The knockdown of circ_0072088 inhibited the cell proliferation and migration, while promoting cell apoptosis. The same biological function was found with the overexpression of miR-1270. The rescue experiment further validated that circ_0072088 could regulate the biological function of cells by influencing miR-1270. Finally, the targeted binding relationship was verified by dual luciferase reporting experiment. In conclusion, circ_0072088 is differentially highly expressed in non-small cell lung cancer and can affect the progression of non-small cell lung cancer through the circ_0072088/miR-1270/TOP2A axis.
Collapse
Affiliation(s)
- Sixuan Li
- Postdoctoral Research Station, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Zhigang Cui
- School of Nursing, China Medical University, Shenyang, 110122, Liaoning, China
| | - Min Gao
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yanan Shan
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yihong Ren
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yuxin Zhao
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Di Wang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Tingyu Meng
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| | - Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
36
|
Xu R, Sun Y, Tian F, Zhao M. LncRNA NEAT1 sponges miR-214-3p to promote osteoblast differentiation through regulating the PI3K/AKT/mTOR pathway in aortic valve calcification. Sci Rep 2025; 15:13665. [PMID: 40258988 PMCID: PMC12012154 DOI: 10.1038/s41598-025-98578-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Calcific aortic valve disease (CAVD) is the common valvular disease associated with significant morbidity and mortality. Dysregulation of long non-coding RNA (lncRNA) has been implicated in the pathogenesis of CAVD. This study aims to investigate the role of NEAT1 in CAVD pathogenesis. NEAT1, miR-214-3p and mRNA expressions were determined by qRT-PCR. Protein expressions were detected by Western blotting. Mineralized bone matrix formation was assessed by Alizarin Red staining. The osteogenic phenotype was evaluated by the alkaline phosphatase activity assay. Dual-luciferase assays were employed to confirm the binding interactions between NEAT1 and miR-214-3p, miR-214-3p and PTEN. NEAT1 was up-regulated in calcific aortic valves and after osteogenic induction of valve interstitial cells (VICs). NEAT1 could act as a positive regulator of osteogenic differentiation by repressing miR-214-3p and thereby promote expression of osteoblast-specific markers. Mechanistically, we identified PTEN as a direct target of miR-214-3p. PTEN could regulate the PI3K/AKT/mTOR pathway and participate in osteogenic differentiation. Importantly, NEAT1 could directly interact with miR-214-3p and change of miR-214-3p expression could efficiently reverse PTEN expression and osteogenic differentiation induced by NEAT1. Thus, NEAT1 positively regulated PTEN expression and activated autophagy through sponging miR-214-3p, and promoted osteogenic differentiation through the PI3K/AKT/mTOR pathway. In conclusion, we elucidates the vital function of NEAT1 as a miRNA sponge in CAVD pathogenesis, and sheds new light on lncRNA-directed diagnostic and therapeutic strategies for CAVD.
Collapse
Affiliation(s)
- Rongjian Xu
- Department of Medical Microbiology, College of Basic Medicine, Qingdao University, Qingdao, China
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuhui Sun
- Department of Cardiothoracic Surgery, Qingdao Eighth People's Hospital, Qingdao, China
| | - Fangfei Tian
- Department of Clinical Laboratory, Qingdao Eighth People's Hospital, Qingdao, China
| | - Min Zhao
- Center of Laboratory Medicine, Qilu Hospital of Shandong University (Qingdao), No. 758 Hefei Road, Qingdao, 266035, China.
| |
Collapse
|
37
|
Tani H. Biomolecules Interacting with Long Noncoding RNAs. BIOLOGY 2025; 14:442. [PMID: 40282307 PMCID: PMC12025117 DOI: 10.3390/biology14040442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
This review explores the complex interactions between long noncoding RNAs (lncRNAs) and other biomolecules, highlighting their pivotal roles in gene regulation and cellular function. LncRNAs, defined as RNA transcripts exceeding 200 nucleotides without encoding proteins, are involved in diverse biological processes, from embryogenesis to pathogenesis. They interact with DNA through mechanisms like triplex structure formation, influencing chromatin organization and gene expression. LncRNAs also modulate RNA-mediated processes, including mRNA stability, translational control, and splicing regulation. Their versatility stems from their forming of complex structures that enable interactions with various biomolecules. This review synthesizes current knowledge on lncRNA functions, discusses emerging roles in development and disease, and evaluates potential applications in diagnostics and therapeutics. By examining lncRNA interactions, it provides insights into the intricate regulatory networks governing cellular processes, underscoring the importance of lncRNAs in molecular biology. Unlike the majority of previous reviews that primarily focused on individual aspects of lncRNA biology, this comprehensive review uniquely integrates structural, functional, and mechanistic perspectives on lncRNA interactions across diverse biomolecules. Additionally, this review critically evaluates cutting-edge methodologies for studying lncRNA interactions, bridges fundamental molecular mechanisms with potential clinical applications, and highlights their potential.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Health Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| |
Collapse
|
38
|
Li X, Wang J, Mai J, Sun Y, Li W, Cai Z, Xu W, Chen Z, Chen S, Wang N. Role of the cell cycle-related gene cdk2 and its associated ceRNA network in sexual size dimorphism of Cynoglossus semilaevis. Comp Biochem Physiol A Mol Integr Physiol 2025; 305:111867. [PMID: 40250729 DOI: 10.1016/j.cbpa.2025.111867] [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: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Sexual size dimorphism (SSD) in Cynoglossus semilaevis affects its annual production and restricts aquaculture development. Our previous multi-omics data analysis showed that cell cycle genes and the relevant non-coding RNAs (ncRNAs) were closely involved in SSD regulation. In this study, we analyzed cyclin-dependent kinase 2 (cdk2) gene together with its associated microRNA (miRNA) and long ncRNA (lncRNA) in C. semilaevis, predicting a competing endogenous RNA (ceRNA) regulatory network (MSTRG.24810.1-miR-460-cdk2) and verifying the targeting relationship using dual luciferase reporter assays. Expression profile analysis showed that cdk2 and the lncRNA MSTRG.24810.1 were highly expressed in female gonad and muscle, and their expression levels increased from 3-month-old (3M) to 8M. On the other hand, their negative regulator miR-460-x displayed lower expression in female than in male. After miR-460-x mimic transfection in C. semilaevis ovarian cells, the expressions of cdk2, cyclin E, and MSTRG.24810.1 were significantly decreased and cell cycle transition through G1 to S phase was obviously blocked. In vitro and in vivo experiments also indicated that RNAi-mediated knock-down of cdk2 caused down-regulation of MSTRG.24810.1 and other cell cycle related genes like cyclin E, cyclin A, e2f1, and h2b. Taken together, these results suggested that cdk2 gene and its associated ceRNA network may affect sex growth difference and differentiation of C. semilaevis individuals via regulating cell division and proliferation. The study will not only expand our knowledge on SSD regulatory mechanism, but also help to make an application on promoting growth and development of the fish.
Collapse
Affiliation(s)
- Xihong Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Jiacheng Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Jiaqi Mai
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Yuqi Sun
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Wenjie Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zhenyu Cai
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Wenteng Xu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Zhangfan Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Songlin Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Na Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
39
|
Alimohammadi M, Abolghasemi H, Cho WC, Reiter RJ, Mafi A, Aghagolzadeh M, Hushmandi K. Interplay between LncRNAs and autophagy-related pathways in leukemia: mechanisms and clinical implications. Med Oncol 2025; 42:154. [PMID: 40202565 DOI: 10.1007/s12032-025-02710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Autophagy is a conserved catabolic process that removes protein clumps and defective organelles, thereby promoting cell equilibrium. Growing data suggest that dysregulation of the autophagic pathway is linked to several cancer hallmarks. Long non-coding RNAs (lncRNAs), which are key parts of gene transcription, are increasingly recognized for their significant roles in various biological processes. Recent studies have uncovered a strong connection between the mutational landscape and altered expression of lncRNAs in the tumor formation and development, including leukemia. Research over the past few years has emphasized the role of lncRNAs as important regulators of autophagy-related gene expression. These RNAs can influence key leukemia characteristics, such as apoptosis, proliferation, epithelial-mesenchymal transition (EMT), migration, and angiogenesis, by modulating autophagy-associated signaling pathways. With altered lncRNA expression observed in leukemia cells and tissues, they hold promise as diagnostic biomarkers and therapeutic targets. The current review focuses on the regulatory function of lncRNAs in autophagy and their involvement in leukemia, potentially uncovering valuable therapeutic targets for leukemia treatment.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Abolghasemi
- Department of Pediatrics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahboobeh Aghagolzadeh
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Zhang S, Lan X, Lei L. LINC01559: roles, mechanisms, and clinical implications in human cancers. Hum Cell 2025; 38:83. [PMID: 40205068 DOI: 10.1007/s13577-025-01218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Long intergenic non-protein coding RNA 1559 (LINC01559), a long non-coding RNA (lncRNA) located on chromosome 12p13.1, plays a critical role in the progression of various cancers. The aberrant expression of LINC01559 significantly impacts multiple biological processes in tumor cells, including cell proliferation, epithelial-mesenchymal transition (EMT), migration, invasion, angiogenesis, and cellular stemness. Notably, the expression levels of LINC01559 correlate with the pathological features and prognosis of several cancers, such as pancreatic, breast, and gastric cancers, and it may serve as a diagnostic marker for non-small cell lung cancer. Moreover, the expression of LINC01559 is regulated by various mechanisms and can influence cancer initiation and progression through a competing endogenous RNA (ceRNA) network, where it interacts with a cohort of eight different microRNAs (miRNAs). Additionally, LINC01559 may directly interact with downstream proteins, thereby promoting their functions or enhancing their stability. LINC01559 is also implicated in key signaling pathways associated with cancer development, including the PI3 K/AKT, RAS, and autophagy signaling pathways. Furthermore, it has been linked to drug resistance in breast cancer and hepatocellular carcinoma. This review provides a comprehensive assessment of the clinical implications of dysregulated LINC01559 expression across various cancer types, highlighting its crucial functions and underlying molecular mechanisms in tumorigenesis. Additionally, we present in-depth discussions and propose hypotheses regarding the functional roles of LINC01559 in cancer pathogenesis, while outlining potential research avenues for future exploration of this molecular target.
Collapse
Affiliation(s)
- Shuwen Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xin Lan
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ling Lei
- Prevention and Treatment Center, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
41
|
Wen B, Chang W, Yang L, Lv D, Wang L, Wang L, Xu Y, Hu J, Ding K, Xue Q, Qi X, Yang B, Wang J. The long noncoding RNA APR attenuates PPRV infection-induced accumulation of intracellular iron to inhibit membrane lipid peroxidation and viral replication. mBio 2025; 16:e0012725. [PMID: 40126010 PMCID: PMC11980570 DOI: 10.1128/mbio.00127-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that has long been a significant threat to small ruminant productivity worldwide. Iron metabolism is vital to the host and the pathogen. However, the mechanism underlying host-PPRV interactions from the perspective of iron metabolism and iron-mediated membrane lipid peroxidation has not been reported thus far. In this study, we identified a novel host long-noncoding RNA (lncRNA), APR, that impairs PPRV infectivity by sponging miR-3955-5p, a negative microRNA (miRNA) that directly targets the gene encoding the ferritin-heavy chain 1 (FTH1) protein. Importantly, we demonstrated that PPRV infection causes aberrant cellular iron accumulation by increasing transferrin receptor (TFRC) expression and that iron accumulation induces reticulophagy and ferroptosis, which benefits PPRV replication. Moreover, PPRV infection enhanced the localization of cellular iron on the endoplasmic reticulum (ER) and caused ER membrane damage by promoting excess lipid peroxidation to induce reticulophagy. Interestingly, APR decreased PPRV infection-induced accumulation of intracellular Fe2+ via miR-3955-5p/FTH1 axis and ultimately inhibited reticulophagy and ferroptosis. Additionally, our results indicate that interferon regulatory factor 1 promotes APR transcription by positively regulating APR promoter activity after PPRV infection. Taken together, our findings revealed a new pattern of PPRV-host interactions, involving noncoding RNA regulation, iron metabolism, and iron-related membrane lipid peroxidation, which is critical for understanding the host defense against PPRV infection and the pathogenesis of PPRV.IMPORTANCEMany viruses have been demonstrated to engage in iron metabolism to facilitate their replication and pathogenesis. However, the mechanism by which PPRV interacts with host cells from the perspective of iron metabolism, or iron-mediated membrane lipid peroxidation, has not yet been reported. Our data provide the first direct evidence that PPRV infection induces aberrant iron accumulation to promote viral replication and reveal a novel host lncRNA, APR, as a regulator of iron accumulation by promoting FTH1 protein expression. In this study, PPRV infection increased cellular iron accumulation by increasing TFRC expression, and more importantly, iron overload increased viral infectivity as well as promoted ER membrane lipid peroxidation by enhancing the localization of cellular iron on the ER and ultimately induced ferroptosis and reticulophagy. Furthermore, a host factor, the lncRNA APR, was found to decrease cellular iron accumulation by sponging miR-3955-5p, which directly targets the gene encoding the FTH1 protein, thereby attenuating PPRV infection-induced ferroptosis and reticulophagy and inhibiting PPRV infection. Taken together, the results of the present study provide new insight into our understanding of host-PPRV interaction and pathogenesis from the perspective of iron metabolism and reveal potential targets for therapeutics against PPRV infection.
Collapse
Affiliation(s)
- Bo Wen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Wenchi Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Lulu Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
- China Institute of Veterinary Drug Control, Beijing, Beijing, China
| | - Lizhen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Yanzhao Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Ke Ding
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, Beijing, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| |
Collapse
|
42
|
Tang Z, Xue Z, Liu X, Zhang Y, Zhao J, Liu J, Zhang L, Guo Q, Feng B, Wang J, Zhang D, Li X. Inhibition of hypoxic exosomal miR-423-3p decreases glioma progression by restricting autophagy in astrocytes. Cell Death Dis 2025; 16:265. [PMID: 40199864 PMCID: PMC11978802 DOI: 10.1038/s41419-025-07576-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) of gliomas comprises glioma cells and surrounding cells, such as astrocytes, macrophages, T cells, and neurons. In the TME, glioma cells can activate normal human astrocytes (NHAs) through the secretion of exosomes and the activation of astrocytes can further improve the progression of glioma, leading to a poor prognosis for patients. However, the molecular mechanisms underlying NHAs activation by gliomas remain largely unknown. It this study, glioma-derived exosomes (GDEs) play an important role in the modulation of autophagy and activation of NHAs. Compared with normoxic GDEs, hypoxic glioma-derived exosomes (H-GDEs) further improved autophagy and activation of astrocytes, which strongly promoted the progression of glioma cells. In an miRNA array between two types of exosomes from gliomas, miR-423-3p was highly expressed in H-GDEs and played an important role in autophagy, resulting in the activation of NHAs. The mechanism by which hypoxic glioma cells react with NHAs to create an immunosuppressive microenvironment was identified and 15d-PGJ2 was established as an effective inhibitor of miR-423-3p to suppress NHAs activation. These findings provide new insights into the diagnosis and treatment of gliomas by targeting autophagy and miR-423-3p expression.
Collapse
Affiliation(s)
- Ziyi Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Xuchen Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiangli Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Junzhi Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Lin Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Bowen Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| |
Collapse
|
43
|
Zhou RT, Luo XJ, Zhang XXR, Wu JF, Ni YR. The potential of miR-29 in modulating tumor angiogenesis: a comprehensive review. Discov Oncol 2025; 16:474. [PMID: 40189720 PMCID: PMC11973036 DOI: 10.1007/s12672-025-02246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of short non-coding RNAs that play a crucial role in the post-transcriptional regulation of gene expression. They are associated with various biological processes related to tumors. Among the numerous miRNAs, miR-29 has garnered attention for its role in regulating tumor angiogenesis. In numerous human tumors, miR-29 has been demonstrated to negatively correlate with the capacity for angiogenesis and the degree of malignancy, as well as with the expression levels of pro-angiogenic factors such as vascular endothelial growth factor vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and matrix metalloproteinase (MMP)-2. Multiple studies, utilizing techniques like dual-luciferase reporter assays, have confirmed that miR-29 directly targets the 3'-untranslated region (UTR) of mRNAs for VEGF, PDGF, and MMP-2. Extensive investigations involving tumor cell lines and animal models have shown that the overexpression of miR-29, achieved through miRNA transfection or the introduction of miRNA mimics, effectively inhibits angiogenesis by upregulating these pro-angiogenic factors. Conversely, downregulation of miR-29 using specific inhibitors promotes angiogenesis. While small molecule inhibitors and antibodies targeting VEGF constitute a primary strategy in anti-angiogenesis therapies, miR-29's ability to target multiple pro-angiogenic molecules positions it as a promising candidate for future therapeutic interventions, especially with ongoing advancements in nucleic acid drug design and delivery systems.
Collapse
Affiliation(s)
- Rui-Ting Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Department of Gastroenterology, The First College of Clinical Medical Science and Yichang Central People's Hospital, China Three Gorges University, Yichang, 443003, China
- Division of Gastroenterology and Hepatology, Renmin Hospital, Wuhan University, Wuhan, 430060, China
| | - Xiao-Jie Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China
| | - Xiao-Xin-Ran Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China
| | - Jiang-Feng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China.
| | - Yi-Ran Ni
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Daxue Road 8#, Yichang, 443002, Hubei, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, 443002, Yichang, China.
| |
Collapse
|
44
|
Le LTT. Long non coding RNA function in epigenetic memory with a particular emphasis on genomic imprinting and X chromosome inactivation. Gene 2025; 943:149290. [PMID: 39880342 DOI: 10.1016/j.gene.2025.149290] [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: 01/14/2024] [Revised: 12/13/2024] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Cells preserve and convey certain gene expression patterns to their progeny through the mechanism called epigenetic memory. Epigenetic memory, encoded by epigenetic markers and components, determines germline inheritance, genomic imprinting, and X chromosome inactivation. First discovered long non coding RNAs were implicated in genomic imprinting and X-inactivation and these two phenomena clearly demonstrate the role of lncRNAs in epigenetic memory regulation. Undoubtedly, lncRNAs are well-suited for regulating genes in close proximity at imprinted loci. Due to prolonged association with the transcription site, lncRNAs are able to guide chromatin modifiers to certain locations, thereby enabling accurate temporal and spatial regulation. Nevertheless, the current state of knowledge regarding lncRNA biology and imprinting processes is still in its nascent phase. Herein, we provide a synopsis of recent scientific advancements to enhance our comprehension of lncRNAs and their functions in epigenetic memory, with a particular emphasis on genomic imprinting and X chromosome inactivation, thus gaining a deeper understanding of the role of lncRNAs in epigenetic regulatory networks.
Collapse
Affiliation(s)
- Linh T T Le
- Faculty of Biotechnology, Ho Chi Minh City Open University, Ho Chi Minh City 700000 Viet Nam
| |
Collapse
|
45
|
Piergentili R, Sechi S. Targeting Regulatory Noncoding RNAs in Human Cancer: The State of the Art in Clinical Trials. Pharmaceutics 2025; 17:471. [PMID: 40284466 PMCID: PMC12030637 DOI: 10.3390/pharmaceutics17040471] [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: 01/22/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Noncoding RNAs (ncRNAs) are a heterogeneous group of RNA molecules whose classification is mainly based on arbitrary criteria such as the molecule length, secondary structures, and cellular functions. A large fraction of these ncRNAs play a regulatory role regarding messenger RNAs (mRNAs) or other ncRNAs, creating an intracellular network of cross-interactions that allow the fine and complex regulation of gene expression. Altering the balance between these interactions may be sufficient to cause a transition from health to disease and vice versa. This leads to the possibility of intervening in these mechanisms to re-establish health in patients. The regulatory role of ncRNAs is associated with all cancer hallmarks, such as proliferation, apoptosis, invasion, metastasis, and genomic instability. Based on the function performed in carcinogenesis, ncRNAs may behave either as oncogenes or tumor suppressors. However, this distinction is not rigid; some ncRNAs can fall into both classes depending on the tissue considered or the target molecule. Furthermore, some of them are also involved in regulating the response to traditional cancer-therapeutic approaches. In general, the regulation of molecular mechanisms by ncRNAs is very complex and still largely unclear, but it has enormous potential both for the development of new therapies, especially in cases where traditional methods fail, and for their use as novel and more efficient biomarkers. Overall, this review will provide a brief overview of ncRNAs in human cancer biology, with a specific focus on describing the most recent ongoing clinical trials (CT) in which ncRNAs have been tested for their potential as therapeutic agents or evaluated as biomarkers.
Collapse
|
46
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [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: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
47
|
Frediani E, Anceschi C, Ruzzolini J, Ristori S, Nerini A, Laurenzana A, Chillà A, Germiniani CEZ, Fibbi G, Del Rosso M, Mocali A, Venturin M, Battaglia C, Giovannelli L, Margheri F. Divergent regulation of long non-coding RNAs H19 and PURPL affects cell senescence in human dermal fibroblasts. GeroScience 2025; 47:2079-2097. [PMID: 39438391 PMCID: PMC11979041 DOI: 10.1007/s11357-024-01399-3] [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: 03/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Cellular senescence is a permanent cell growth arrest that occurs in response to various intrinsic and extrinsic stimuli and is associated with cellular and molecular changes. Long non-coding RNAs (lncRNAs) are key regulators of cellular senescence by affecting the expression of many important genes involved in senescence-associated pathways and processes. Here, we evaluated a panel of lncRNAs associated with senescence for their differential expression between young and senescent human dermal fibroblasts (NHDFs) and studied the effect of a known senomorphic compound, resveratrol, on the expression of lncRNAs in senescent NHDFs. As markers of senescence, we evaluated cell growth, senescence-associated (SA)-β-Gal staining, and the expression of p21, Lamin B1 and γH2AX. We found that H19 and PURPL were the most altered lncRNAs in replicative, in doxorubicin (DOXO) and ionising radiation (IR)-induced senescence models. We then investigated the function of H19 and PURPL in cell senescence by siRNA-mediated silencing in young and senescent fibroblasts, respectively. Our results showed that H19 knockdown reduced cell viability and induced cell senescence and autophagy of NHDFs through the regulation of the PI3K/AKT/mTOR pathway; conversely, PURPL silencing reversed senescence by reducing (SA)-β-Gal staining, recovering cell proliferation with an increase of S-phase cells, and reducing the p53-dependent DNA damage response. Overall, our data highlighted the role of H19 and PURPL in the senescent phenotype and suggested that these lncRNAs may have important implications in senescence-related diseases.
Collapse
Affiliation(s)
- Elena Frediani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Cecilia Anceschi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Sara Ristori
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Alice Nerini
- Department of Neurofarba (Department of Neurosciences, Drug Research and Child Health), University of Florence, Viale Pieraccini, 6 - 50139, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Claudia Elena Zoe Germiniani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Marco Venturin
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Lisa Giovannelli
- Department of Neurofarba (Department of Neurosciences, Drug Research and Child Health), University of Florence, Viale Pieraccini, 6 - 50139, Florence, Italy.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy.
| |
Collapse
|
48
|
Pan Y, Tang X, Xie Y, Zhang H, Huang Z, Huang C. Long non-coding RNA BCAR4 regulates osteosarcoma progression by targeting microRNA-1260a. Bull Cancer 2025; 112:375-386. [PMID: 40087067 DOI: 10.1016/j.bulcan.2024.09.003] [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: 04/20/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 03/16/2025]
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in modulating cancer progression and metastasis. This study investigates the tumor-promoting function of long non-coding RNA BCAR4 in osteosarcoma and elucidates its regulatory mechanism. Although BCAR4 acts as a tumor promoter in osteosarcoma, its regulatory mechanism remains unclear. Bioinformatic analysis revealed a specific interaction between BCAR4 and miR-1260a, with osteosarcoma exhibiting elevated miR-1260a expression inversely correlated with BCAR4 expression. Overexpression of BCAR4 significantly suppressed miR-1260a expression, indicating regulation between BCAR4 and miR-1260a. Luciferase reporter assays confirmed a direct association between miR-1260a and BCAR4 at the sequence level. Silencing of BCAR4 inhibited osteosarcoma cell proliferation and migration while promoting cellular apoptosis, primarily mediated by miR-1260a. Our findings demonstrate that BCAR4 functions as a tumor promotor in osteosarcoma, and that its activity is regulated by miR-1260a. This study also proposes a potential therapeutic approach for treating osteosarcoma by targeting the BCAR4/miR-1260a axis. These different insights shed light on the intricate regulatory network underlying osteosarcoma pathogenesis and offer promising avenues for developing targeted therapies against this aggressive cancer.
Collapse
Affiliation(s)
- Yixin Pan
- Department of Pathology, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, China
| | - Xiaolei Tang
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| | - Yadong Xie
- Department of Orthopedics, the First Hospital of Lanzhou University, Lanzhou, Gansu 730030, China
| | - Huamin Zhang
- Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Ziyu Huang
- Medical Laboratory Technology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Changjia Huang
- Spine Orthopaedics, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China.
| |
Collapse
|
49
|
Vivek AT, Sahu N, Kalakoti G, Kumar S. ANNInter: A platform to explore ncRNA-ncRNA interactome of Arabidopsis thaliana. Comput Biol Chem 2025; 115:108328. [PMID: 39754835 DOI: 10.1016/j.compbiolchem.2024.108328] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Eukaryotic transcriptomes are remarkably complex, encompassing not only protein-coding RNAs but also an expanding repertoire of noncoding RNAs (ncRNAs). In plants, ncRNA-ncRNA interactions (NNIs) have emerged as pivotal regulators of gene expression, orchestrating development and adaptive responses to stress. Despite their critical roles, the functional significance of NNIs remains poorly understood, largely due to a lack of comprehensive resources. Here, we present ANNInter, a comprehensive platform that integrates computational predictions with experimental datasets to systematically identify and analyze NNIs. The current version catalogs over 90,000 interactions spanning eight categories of sRNA-to-longer ncRNAs, each extensively annotated with interaction types, identification methods, and functional descriptions. The integrated schema and advanced visualization framework in ANNInter enable users to explore intricate interaction networks, providing system-wide insights into ncRNA-mediated regulation. These interaction data provide unparalleled opportunities to uncover the regulatory roles of NNIs in key biological processes such as growth regulation, stress adaptation, and cellular signaling. By providing an extensive, curated repository of computational and degradome-based interaction data, ANNInter will provide a platform to the study of ncRNA biology, elucidating the complex mechanisms of NNIs and supporting the concept of competing endogenous RNAs (ceRNAs) in gene regulation. The platform is freely accessible at https://www.nipgr.ac.in/ANNInter/.
Collapse
Affiliation(s)
- A T Vivek
- Bioinformatics Lab, National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Namrata Sahu
- Bioinformatics Lab, National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Garima Kalakoti
- Bioinformatics Lab, National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shailesh Kumar
- Bioinformatics Lab, National Institute of Plant Genome Research (NIPGR), Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
50
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [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: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|