1
|
Wang L, Gu Y, Shen C. Transcriptome analysis and lncRNA expression profile in brain tissues of neonatal hypoxic-ischemic brain damage rat model. Gene 2025; 952:149363. [PMID: 40064305 DOI: 10.1016/j.gene.2025.149363] [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: 11/08/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025]
Abstract
BACKGROUND AND OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) remains a critical challenge in perinatal medicine. This study aimed to elucidate the transcriptomic landscape, focusing on long non-coding RNAs (lncRNAs) expression patterns in the brain tissues of a neonatal rat model of HIE. METHODOLOGY We employed a modified Rice-Vannucci model to induce HIE in postnatal day 4 (P4) rats. The experimental groups were subjected to either 5 or 7 min of hypoxia (0 % O2, 100 % N2), while control animals were exposed to normoxic conditions. RESULTS RNA sequencing revealed a complex transcriptomic landscape in HIE brains, with approximately 80 million differentially expressed lncRNAs compared to controls. ELISA results demonstrated a significant upregulation of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and a concomitant decrease in anti-inflammatory IL-10 levels in brain tissue of HIE rats. qRT-PCR analysis revealed aberrant expression of several miRNAs. Biochemical assays indicated a marked reduction in superoxide dismutase (SOD) activity and an increase in malondialdehyde (MDA) content in HIE brain tissues. CONCLUSIONS This study highlights the potential regulatory roles of lncRNAs in HIE brains. The intricate interplay between lncRNAs, miRNAs, and mRNAs and alterations in inflammatory and oxidative stress markers suggests a complex regulatory network governing HIE pathogenesis.
Collapse
Affiliation(s)
- Limin Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Yanni Gu
- Department of Fever Emergency, Shanghai TCM-Integrated Hospital, Shanghai 200082, China
| | - Chaobin Shen
- Department of Pediatrics, Shanghai TCM-Integrated Hospital, Shanghai 200082, China.
| |
Collapse
|
2
|
Zhao L, Tian G, Wang X, Li L, Gao Y, Gao Y, Wang J. Clinical significance of a new early diagnostic model for bladder cancer based on genome-wide microarray profiling of serum exosomal lncRNAs. Int Urol Nephrol 2025; 57:1771-1783. [PMID: 39776008 DOI: 10.1007/s11255-024-04360-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The aim of our report was to recognize bladder cancer (BC)-specific serum exosome-derived long non-coding RNAs (lncRNAs) profile for early diagnosis of BC. METHODS Potential BC-specific exosomal lncRNA indicators were discerned by genome-wide microarray profiling analysis of serum exosomes from 10 healthy participants and 10 early stage BC patients (Ta and T1), followed by multi-stage validation through quantitative real-time PCR (qRT-PCR) in BC cells, culture solution as well as 200 serum specimens and 50 tissue specimens from non-muscle-invasive bladder cancer (NMIBC) patients. The diagnostic panel was established using logistic regression and evaluated by receiver-operating characteristic (ROC) curve. RESULTS In the training stage, a diagnostic panel was constructed based on three up-regulated exosomal lncRNAs (G023016, RP11-553N19.1, and LINC0087) in NMIBC patients compared with healthy controls, yielding an area under ROC curve (AUC) of 0.827. We verified tumor-derived origin of these three lncRNAs which existed steadily in serum because of being enclosed in exosomes. The three-lncRNA panel was demonstrated to perform well in terms of NMIBC diagnosis, revealing AUC values of 0.809 and 0.812, respectively, in the following expanded validation stage and double-blind stage which was demonstrated to be significantly superior to that of urine cytology in double-blind stage (AUC = 0.630) (P < 0.0001). Moreover, serum exosome-derived G023016 significantly associated with tumor grade and TNM stage (P = 0.006 and P < 0.001, respectively), and LINC0087 significantly associated with TNM stage (P = 0.023). CONCLUSION The three-exosomal lncRNA signature could function as qualified blood-based non-invasive indicator for early diagnosis of BC.
Collapse
Affiliation(s)
- Liming Zhao
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, 27 Jiefang Road, Linyi, 276003, Shandong, China
| | - Guang Tian
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, 27 Jiefang Road, Linyi, 276003, Shandong, China
| | - Xiaohua Wang
- Department of General Internal Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong, China
| | - Luning Li
- Department of Gastroenterology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong, China
| | - Yongli Gao
- Department of Oncology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong, China
| | - Yisheng Gao
- Department of Urology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong, China
| | - Jinfeng Wang
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, 27 Jiefang Road, Linyi, 276003, Shandong, China.
| |
Collapse
|
3
|
Zhang H, Gong L, Yu L, Xian C, Ma Z, Wang X, Xia R. Emerging roles of non-coding RNA derived from extracellular vesicles in regulating PD-1/PD-L1 pathway: insights into cancer immunotherapy and clinical applications. Cancer Cell Int 2025; 25:188. [PMID: 40410719 PMCID: PMC12103061 DOI: 10.1186/s12935-025-03809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
Numerous studies have demonstrated that extracellular vesicles (EVs) carry a variety of noncoding RNAs (ncRNAs), which can be taken up by neighboring cells or transported to distant sites via bodily fluids, thereby facilitating intercellular communication and regulating multiple cellular functions. Within the tumor microenvironment, EV-ncRNA, on the one hand, regulate the expression of PD-L1, thereby influencing tumor immune evasion, promoting tumor cell proliferation, and enhancing tumor growth, invasion, and metastasis in vivo. On the other hand, these specific EV-ncRNAs can also modulate the functions of immune cells (such as CD8 + T cells, macrophages, and NK cells) through various molecular mechanisms, inducing an immunosuppressive microenvironment and promoting resistance to anti-PD-1 therapy. Therefore, delving into the molecular mechanisms underlying EV-ncRNA regulation of immune checkpoints presents compelling therapeutic prospects for strategies that selectively target EV-ncRNAs. In this review, we elaborate on the cutting-edge research progress related to EV-ncRNAs in the context of cancer and dissect their pivotal roles in the PD-1/PD-L1 immune checkpoint pathway. We also highlight the promising clinical applications of EV-ncRNAs in anti-PD-1/PD-L1 immunotherapy, bridging basic research with practical clinical applications.
Collapse
Affiliation(s)
- Haixia Zhang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Lianfeng Gong
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Li Yu
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
- Department of Urology, General Hospital of The Yangtze River Shipping, Wuhan, 430010, China
| | - Chenge Xian
- Naidong District People's Hospital, Shannan, 856004, Tibet Autonomous Region, China
| | - Zhaowu Ma
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| | - Xianwang Wang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
- Shannan Maternal and Child Health Hospital, Shannan, 856099, Tibet Autonomous Region, China.
| | - Ruohan Xia
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| |
Collapse
|
4
|
Liu Y, Li X, Wang F, Cai J, Li Z, Huang Y, Duan X, Liu X, He Y, Xu G, Lu Q. MTA1-DT promotes endometrial cancer growth by modulating G2/M-related gene transcription via PURα. Int J Biol Macromol 2025; 309:142943. [PMID: 40210047 DOI: 10.1016/j.ijbiomac.2025.142943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
In recent years, patients with early endometrial cancer (EC) can achieve a good prognosis through surgery. However, advanced and recurrent cases have still posed significant therapeutic challenges. This study aimed to investigate the biological function of long non-coding RNAs (lncRNAs) in EC and elucidate its underlying molecular mechanism. Through quantitative reverse transcription polymerase chain reaction (RT-qPCR) analysis, functional assays in cell lines, and bioinformatics approaches, we identified lncRNA MTA1-DT as a novel oncogenic factor in EC progression. RNA-seq and RT-qPCR analysis demonstrated that MTA1-DT was significantly upregulated with a 5-fold increase in EC cell lines compared to normal controls. Functional studies revealed that MTA1-DT promoted cell proliferation and migration. Mechanistically, we demonstrated that MTA1-DT physically interacted with purine-rich element binding protein-alpha (PURα) and facilitated its nuclear translocation, thereby enhancing its transcription factor activity. This nuclear accumulation of PURα promoted the transcription of downstream G2/M related genes, particularly EGF, leading to accelerated tumor growth. Thus, these results indicate that MTA1-DT exerts its oncogenic effects in EC through regulation of the cell cycle. Our findings establish MTA1-DT as a promising therapeutic target for EC treatment and provide new insights into the molecular mechanisms underlying EC progression.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xin Li
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fanchen Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jinhui Cai
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zhouqi Li
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Yanchun Huang
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xiaoling Duan
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Xinyi Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Yuxin He
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Qi Lu
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai 201508, China.
| |
Collapse
|
5
|
Feng H, Nie Q, Yang S. SORFPP: Enhancing rich sequence-driven information to identify SEPs based on fused framework on validation datasets. PLoS One 2025; 20:e0320314. [PMID: 40294059 PMCID: PMC12036913 DOI: 10.1371/journal.pone.0320314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Genome sequencing has enabled us to find functional peptides encoded by short open read frames (sORFs) in long non-coding RNAs (lncRNAs). sORFs-encoded peptides (SEPs) regulate gene expression, signaling, and so on and have significant roles, unlike common peptides. Various computational methods have been proposed. However, there is a lack of contributive features and effective models. Therefore, a high-throughput computational method to predict SEPs is needed. RESULTS We propose a computational method, SORFPP, to predict SEPs by mining feature information from multiple perspectives in an experimentally validated dataset from TranLnc. SORFPP fully extracts SEP sequence information using the protein language model ESM-2 and curated traditional encoding, including QSOrder, k-mer, etc. SORFPP uses CatBoost to solve the sparsity problem of traditional encoding. SORFPP also analyzes ESM-2 pre-training characterization information with the Self-attention model. Finally, an ensemble learning framework combines the two models and their results are fed into Logistic Regression model for accurate and robust predictions. For comparison, SORFPP outperforms other state-of-the-art models in Matthew correlation coefficient by 12.2%-24.2% on three benchmark datasets. CONCLUSION Integrating the ensemble learning strategy with contributive traditional features and the protein language encoding methods shows better performance. Datasets and codes are accessible at https://doi.org/10.6084/m9.figshare.28079897 and http://111.229.198.94:5000/.
Collapse
Affiliation(s)
- Hongqi Feng
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
| | - Qi Nie
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
| | - Sen Yang
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
6
|
Song Z, Suo C, Liu Y, Jin L, Xie X, Liu J, Yu B, Wang Y, Zhang Z, Xie D. Comprehensive evaluation of non-coding RNA-mediated autophagy regulation in myocardial ischemia-reperfusion injury. Front Pharmacol 2025; 16:1581341. [PMID: 40351430 PMCID: PMC12062134 DOI: 10.3389/fphar.2025.1581341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Ischemic heart disease remains a major global health challenge, with myocardial ischemia-reperfusion injury (MIRI) being one of its most common and severe pathophysiological complications. The pathogenesis of MIRI is multifaceted, involving oxidative stress, inflammatory responses, apoptotic pathways, and autophagic regulation. Notably, autophagy exerts a dual regulatory effect, where maintaining optimal autophagic flux is essential for cardiac homeostasis. Emerging evidence underscores the crucial role of non-coding RNAs (ncRNAs) in modulating these pathological processes. In particular, long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) have been identified as key regulators of autophagy-mediated MIRI progression through complex molecular networks. This review provides a systematic analysis of the molecular pathways through which ncRNAs influence MIRI pathogenesis, with a specific focus on their autophagy-regulatory mechanisms. These insights may enhance our understanding of MIRI pathobiology and facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Gansu Institute of Cardiovascular Diseases, Lanzhou, Gansu, China
| | - Chang Suo
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yongqi Liu
- Key Laboratory of Dunhuang Medicine and Transformation of Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Ling Jin
- Longyao Industry Innovation Institute, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaodong Xie
- Institute of Genetics, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Jian Liu
- Department of Critical Care Medicine, Maternal and Child Healthcare Hospital of Gansu Province, Lanzhou, China
| | - Bo Yu
- Department of General Surgery, The Second People’s Hospital of Lanzhou City, Lanzhou, China
| | - Yanzhen Wang
- Gansu Institute of Cardiovascular Diseases, Lanzhou, Gansu, China
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Dingxiong Xie
- Gansu Institute of Cardiovascular Diseases, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Miao Z, Sha Z, He J, Liang Y, Tan L, Zhao Y, Cui X, Zhong J, Zhong R, Liang H, Yue W, Qiu B, Gao Y, Zhang L, Teng Z, He Z, Chen L, Xiao R, Pei X, He C. Long non-coding RNA LRTOR drives osimertinib resistance in non-small cell lung cancer by boosting YAP positive feedback loop. Drug Resist Updat 2025:101245. [PMID: 40316465 DOI: 10.1016/j.drup.2025.101245] [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: 01/14/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025]
Abstract
The therapeutic efficacy of osimertinib (OSI) in EGFR-mutant lung cancer is ultimately limited by the onset of acquired resistance, of which the mechanisms remain poorly understood. Here, we identify a novel long non-coding RNA, LRTOR, as a key driver of OSI resistance in non-small cell lung cancer (NSCLC). Clinical data indicate that elevated LRTOR expression correlates with poor prognosis in OSI-resistant patients. Functionally, LRTOR promotes tumor growth and confers OSI resistance both in vitro and in vivo. Mechanistically, LRTOR shields YAP from LATS-mediated phosphorylation at Ser127 and Ser381, preventing its proteasomal degradation. Furthermore, LRTOR facilitates the interaction between YAP and KCMF1, promoting K63-linked ubiquitination, nuclear translocation of YAP, and formation of the YAP/TEAD1 transcriptional complex, which in turn triggers the transcription of LRTOR, establishing a positive feedback loop that amplifies oncogenic signaling of YAP and consequently induces OSI resistance. LRTOR depletion by siRNA restores OSI sensitivity in resistant tumors, as demonstrated in patient-derived organoid xenograft models. Our findings unveil LRTOR as a central regulator of OSI resistance in NSCLC and propose it as a promising therapeutic and prognostic target for overcoming acquired OSI resistance in EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Zhimin Miao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Zhou Sha
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yongkai Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Lihua Tan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Yuxin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Xiaobing Cui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Jinmiao Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Ruting Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Huijun Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Wendi Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Boyang Qiu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Yunzhen Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Lan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Zixin Teng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Zeen He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Li Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Rufei Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Xiaofeng Pei
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China.
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China.
| |
Collapse
|
8
|
Qi J, Jiang T, Liu B, Hu Q, Chen J, Ma N, Xu Y, Song H, Song J. LINC02167 stabilizes KSR1 mRNA in an m 5C-dependent manner to regulate the ERK/MAPK signaling pathway and promotes colorectal cancer metastasis. J Exp Clin Cancer Res 2025; 44:121. [PMID: 40234937 PMCID: PMC11998267 DOI: 10.1186/s13046-025-03368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Metastasis is a leading cause of colorectal cancer (CRC)-related mortality, yet its molecular mechanisms remain poorly understood. Long noncoding RNAs (lncRNAs) have emerged as critical regulators of CRC metastasis, but their specific roles are not fully elucidated. This study identifies and characterizes a novel lncRNA LINC02167 as a critical regulator of CRC metastasis. METHODS LINC02167 expression was analyzed in CRC tissues via real-time quantitative polymerase chain reaction and fluorescence in situ hybridization. Functional assays evaluated its role in CRC cell migration, invasion, and metastasis in vitro and in vivo. Mechanistic exploration involves a combination of techniques, including RNA sequencing, mass spectrometry, RNA pull-down, RNA immunoprecipitation, chromatin immunoprecipitation, luciferase reporter assays, RNA stability assays, and bioinformatics analysis, to uncover the molecular interactions and pathways regulated by LINC02167. RESULTS LINC02167 is markedly upregulated in CRC tissues and strongly correlates with advanced clinical features and poor prognosis. Functional analyses reveal that LINC02167 enhances CRC cell migration and invasion in vitro and promotes metastasis in vivo. Mechanistically, LINC02167 serves as a molecular scaffold, forming a complex with YBX1 and ILF3 to facilitate YBX1 binding to NSUN2-mediated m5C modification sites on KSR1 mRNA, thereby stabilizing KSR1 mRNA and activating the ERK/MAPK signaling pathway to drive CRC metastasis. Additionally, MYC-driven transcriptional activation leads to the upregulation of LINC02167 in CRC. CONCLUSIONS This study uncovers a novel mechanism through which LINC02167 promotes the ERK/MAPK pathway and CRC metastasis via m5C modification, underscoring its potential as a promising therapeutic target for metastatic CRC treatment.
Collapse
Affiliation(s)
- Junwen Qi
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Qihang Hu
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Junnan Chen
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ning Ma
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hu Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
9
|
Kharitonova A, Patel RS, Osborne B, Krause-Hauch M, Lui A, Vidyarthi G, Li S, Cai J, Patel NA. NPC86 Increases LncRNA Gas5 In Vivo to Improve Insulin Sensitivity and Metabolic Function in Diet-Induced Obese Diabetic Mouse Model. Int J Mol Sci 2025; 26:3695. [PMID: 40332318 PMCID: PMC12027414 DOI: 10.3390/ijms26083695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
In the United States, an estimated 38 million individuals (10% of the population) have type 2 diabetes mellitus (T2D), while approximately 97.6 million adults (38%) have prediabetes. Long noncoding RNAs (lncRNAs) are critical regulators of gene expression and metabolism. We were the first to demonstrate that lncRNA Growth Arrest-Specific Transcript 5 (GAS5 (human)/gas5 (mouse)) is decreased in the serum of T2D patients and established GAS5 as a biomarker for T2D diagnosis and onset prediction, now validated by other groups. We further demonstrated that GAS5 depletion impaired glucose uptake, decreased insulin receptor levels, and inhibited insulin signaling in human adipocytes, highlighting its potential as a therapeutic target in T2D. To address this, we developed NPC86, a small-molecule compound that stabilizes GAS5 by disrupting its interaction with UPF-1, an RNA helicase involved in nonsense-mediated decay (NMD) that regulates RNA stability. NPC86 increased GAS5 and insulin receptor (IR) levels, enhanced insulin signaling, and improved glucose uptake in vitro. In this study, we tested the efficacy of NPC86 in vivo in a diet-induced obese diabetic (DIOD) mouse model, and NPC86 treatment elevated gas5 levels, improved glucose tolerance, and enhanced insulin sensitivity, with no observed toxicity or weight changes. A transcriptomics analysis of adipose tissue revealed the upregulation of insulin signaling and metabolic pathways, including oxidative phosphorylation and glycolysis, while inflammatory pathways were downregulated. These findings highlight NPC86's therapeutic potential in T2D.
Collapse
MESH Headings
- Animals
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Mice
- Insulin Resistance/genetics
- Humans
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/drug therapy
- Disease Models, Animal
- Male
- Mice, Obese
- Obesity/metabolism
- Mice, Inbred C57BL
- Diet, High-Fat/adverse effects
- Signal Transduction/drug effects
- Insulin/metabolism
- Glucose/metabolism
- Receptor, Insulin/metabolism
Collapse
Affiliation(s)
- Anna Kharitonova
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Rekha S. Patel
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Brenna Osborne
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Meredith Krause-Hauch
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Ashley Lui
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Gitanjali Vidyarthi
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Sihao Li
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Niketa A. Patel
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Ni H, Ge Y, Zhuge Y, Liu X, Chen H, Liu J, Li W, Wang X, Shen G, Wang Q, Zhuang R, Feinberg MW, Wang F. LncRNA MIR181A1HG Deficiency Attenuates Vascular Inflammation and Atherosclerosis. Circ Res 2025; 136:862-883. [PMID: 40047069 PMCID: PMC11985291 DOI: 10.1161/circresaha.124.325196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Endothelial cell (EC) dysfunction and vascular inflammation are critical in the initiation and progression of atherosclerosis. Long noncoding RNAs play a critical role in vascular pathology, but relatively little is known about their involvement in controlling vascular inflammation. MIR181A1HG is a conserved long noncoding RNA located in juxtaposition with miR-181a1 and miR-181b1, both involved in vascular inflammation. The study aims to investigate the role of MIR181A1HG in regulating vascular inflammation. METHODS We examined the expression of MIR181A1HG in both human and mouse atherosclerotic lesions. Loss-of-function and gain-of-function studies, and multiple RNA-protein interaction assays were used to investigate the role and molecular mechanisms of MIR181A1HG in vascular inflammation and atherosclerosis. The atherosclerotic phenotypes of MIR181A1HG-/-ApoE-/- mice were analyzed in combination with single-cell RNA sequencing. The transcriptional regulation of MIR181A1HG was verified through luciferase reporter and chromatin immunoprecipitation assays. RESULTS MIR181A1HG expression was abundant in ECs and significantly increased in both human and mouse atherosclerotic lesions. MIR181A1HG-/-ApoE-/- mice had reduced NLRP (NLR family pyrin domain containing) 3 inflammasome signaling, EC activation, monocyte infiltration, and atherosclerotic lesion formation. Genetic deletion of MIR181A1HG in myeloid sells did not alter the progression of atherosclerosis. Single-cell RNA sequencing analysis revealed that MIR181A1HG deficiency reduced the proportion of immune cells and enriched anti-inflammation pathways in EC clusters in atherosclerotic lesions. In contrast, EC-specific MIR181A1HG overexpression promoted NLRP3 inflammasome signaling, EC activation, and atherosclerotic lesion formation, effects that were reversed by pharmacological inhibition of NLRP3 (MCC950). MIR181A1HG was transcriptionally activated via an NF-κB (nuclear factor kappa B)/p65-dependent pathway. Mechanistically, MIR181A1HG mediated these effects on regulating NLRP3 inflammasome and EC activation in part through decoying Foxp1 (forkhead box transcription factor 1) away from the promoters of target genes, which was independent of the miR-181a1/b1 cluster. Finally, EC-specific Foxp1 silencing reversed the antiatherosclerotic effect mediated by MIR181A1HG-deletion in vivo. CONCLUSIONS These findings identify MIR181A1HG as a central driver of vascular inflammation in atherosclerosis by its ability to decoy Foxp1 away from target gene promoters and activate NLRP3 inflammasome in the vascular endothelium. Our study suggests MIR181A1HG as a future therapeutic target for vascular inflammatory disease states.
Collapse
Affiliation(s)
- Huaner Ni
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yulong Ge
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ying Zhuge
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaoqiang Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Hangwei Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Junyi Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Weifeng Li
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiang Wang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Gu Shen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qiuling Wang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Rulin Zhuang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mark W. Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
11
|
Xu M, Gan D, Zhang X, He X, Wu RX, Yin Y, Jin R, Li L, Tan Y, Chen F, Li X, Tian B. SLC30A4-AS1 Mediates the Senescence of Periodontal Ligament Stem Cells in Inflammatory Environments via the Alternative Splicing of TP53BP1. Cell Prolif 2025; 58:e13778. [PMID: 39572253 PMCID: PMC11969240 DOI: 10.1111/cpr.13778] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 11/09/2024] [Indexed: 04/05/2025] Open
Abstract
Periodontal ligament stem cells (PDLSCs) are key cells that suppress periodontal damage during both the progression and recovery stages of periodontitis. Although substantial evidence has demonstrated that incubation under an inflammatory condition may accelerate senescence of PDLSCs, whether cellular senescence in response to inflammatory incubation contributes to cell dysfunction remain unexplored. In this study, we first observed inflammation-caused PDLSC senescence in periodontitis based on comparisons of matched patients, and this cellular senescence was demonstrated in healthy cells that were subjected to inflammatory conditions. We subsequently designed further experiments to investigate the possible mechanism underlying inflammation-induced PDLSC senescence with a particular focus on the role of long noncoding RNAs (lncRNAs). LncRNA microarray analysis and functional gain/loss studies revealed SLC30A4-AS1 as a regulator of inflammation-mediated PDLSC senescence. By full-length transcriptome sequencing, we found that SLC30A4-AS1 interacted with SRSF3 to affect the alternative splicing (AS) of TP53BP1 and alter the expression of TP53BP1-204. Further functional studies showed that decreased expression of TP53BP1-204 reversed PDLSC senescence, and SLC30A4-AS1 overexpression-induced PDLSC senescence was abolished by TP53BP1-204 knockdown. Our data suggest for the first time that SLC30A4-AS1 plays a key role in regulating PDLSC senescence in inflammatory environments by modulating the AS of TP53BP1.
Collapse
Affiliation(s)
- Mei Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Dian Gan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xi‐Yu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Xin Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Lin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yu‐Jie Tan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
12
|
Huang H, Ma L, Wang X, Huang X, Wang H, Peng Y, Xiao J, Liu H, Yang Z, Cao Z. Platr3/NUDT21/NF-κB Axis Mediates P. gingivalis-Suppressed Cementoblast Mineralization. Inflammation 2025; 48:621-632. [PMID: 38961014 DOI: 10.1007/s10753-024-02069-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
Porphyromonas gingivalis (P. gingivalis) is one of the major pathogens causing periodontitis and apical periodontitis (AP). Long noncoding RNA (lncRNA) can regulate cellular mineralization and inflammatory diseases. The aim of this study was to investigate the role and mechanism of lncRNA in P. gingivalis-stimulated cementoblast mineralization. In vivo, C57BL/6 mice were divided into the healthy, the AP, and AP + P. gingivalis groups (n = six mice per group). Micro computed tomography, immunohistochemistry staining, and fluorescence in situ hybridization were used to observe periapical tissue. In vitro, cementoblasts were treated with osteogenic medium or P. gingivalis. Pluripotency associated transcript 3 (Platr3), interleukin 1 beta (IL1B), and osteogenic markers were analyzed by quantitative real-time polymerase chain reaction and western blot. RNA pull-down and RNA immunoprecipitation assays were used to detect proteins that bind to Platr3. RNA sequencing was performed in Platr3-silenced cementoblasts. In vivo, P. gingivalis promoted periapical tissue destruction and IL1B expression, but inhibited Platr3 expression. In vitro, P. gingivalis facilitated IL1B expression (P < 0.001), whereas suppressed the expression of Platr3 (P < 0.001) and osteogenic markers (P < 0.01 or 0.001). In contrast, Platr3 overexpression alleviated the repressive effect of P. gingivalis on cementoblast mineralization (P < 0.01 or 0.001). Furthermore, Platr3 bound to nudix hydrolase 21 (NUDT21) and regulated the nuclear factor-κB (NF-κB) signaling pathway. Knocking down NUDT21 suppressed osteogenic marker expression and activated the above signaling pathway. Collectively, the results elucidated that Platr3 mediated P. gingivalis-suppressed cementoblast mineralization through the NF-κB signaling pathway by binding to NUDT21.
Collapse
Affiliation(s)
- Hantao Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Li Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China
| | - Xiaoxuan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China
| | - Xin Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huiyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yan Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junhong Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengkun Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
13
|
Zhang Y, Xu Y, Zhang Y, Wang S, Zhao M. The multiple functions and mechanisms of long non-coding RNAs in regulating breast cancer progression. Front Pharmacol 2025; 16:1559408. [PMID: 40223929 PMCID: PMC11985786 DOI: 10.3389/fphar.2025.1559408] [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: 01/12/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer (BC) is a malignant tumor that has the highest morbidity and mortality rates in the female population, and its high tendency to metastasize is the main cause of poor clinical prognosis. Long non-coding RNAs (lncRNAs) have been extensively documented to exhibit aberrant expression in various cancers and influence tumor progression via multiple molecular pathways. These lncRNAs not only modulate numerous aspects of gene expression in cancer cells, such as transcription, translation, and post-translational modifications, but also play a crucial role in the reprogramming of energy metabolism by regulating metabolic regulators, which is particularly significant in advanced BC. This review examines the characteristics and mechanisms of lncRNAs in regulating BC cells, both intracellularly (e.g., cell cycle, autophagy) and extracellularly (e.g., tumor microenvironment). Furthermore, we explore the potential of specific lncRNAs and their regulatory factors as molecular markers and therapeutic targets. Lastly, we summarize the application of lncRNAs in the treatment of advanced BC, aiming to offer novel personalized therapeutic options for patients.
Collapse
Affiliation(s)
- Yongsheng Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yanjiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanping Zhang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shoushi Wang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Mingqiang Zhao
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
14
|
Hu Y, Liu Z, Qin Y, Wu N, Yang T, Cheng X, Wang C, Wang X. Identification of Pivotal ceRNA Networks Associated with Stanford-A Aortic Dissection via Integrated Bioinformatics Analysis. Int J Gen Med 2025; 18:1509-1527. [PMID: 40123810 PMCID: PMC11927577 DOI: 10.2147/ijgm.s509177] [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] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
Objective Stanford-A Aortic dissection (TAAD) is a rare and fatal disease, genetic factors remains poorly known. Study has confirmed that lncRNA play an important role in various physiological and pathological processes. This study attempts to elucidate the underlying molecular mechanisms of TAAD through lncRNA-associated competitive endogenous RNA (ceRNA) networks. Methods In this study, aortic vascular of 5 TAAD and 5 control (ischemic heart disease) were subjected to lncRNA and mRNA microarray analysis, and differentially expressed mRNAs (DEGs) and differentially expressed lncRNAs (DELs) were identified. The differentially expressed miRNAs (DEmiR) were screened by GSE98770 dataset. The ceRNA network (lncRNA-miRNA-mRNA) was constructed by bioinformatics analysis. The accuracy of hub genes as biomarkers for predicting TAAD was evaluated by receiver operating characteristic (ROC) curve. Finally, the biomarkers were verified by assessing their mRNA levels using real-time quantitative PCR (RT-qPCR). Results This study revealed 161 DELs, 87 DEmiRs and 103 DEGs between TAAD and control. We constructed ceRNA networks based on the screened 1 lncRNA, 4 miRNAs and 7 mRNAs. We identified three lncRNA-miRNA-mRNA regulatory axes, namely the VCAN axis (LINC01355 - hsa-miR-186-5p / hsa-miR-30a-5p /hsa-miR-30c-5p - VCAN), LOX axis (LINC01355-hsa-miR-145-5p/hsa-miR-186-5p/ hsa-miR-30a-5p / hsa-miR-30c-5p - LOX), and CTSS axis (LINC01355 - hsa-miR-186-5p - CTSS) based on gene ontology, pathway enrichment and protein-protein interaction (PPI) network, which may play an important role in TAAD. The clinical performance of VCAN, CTSS, and LOX in TAAD diagnosis was evaluated, and the AUCs of VCAN, CTSS, and LOX were 0.920 (p<0.001), 0.880 (p=0.002) and 0.840 (p=0.011), respectively. Furthermore, mRNA expression of VCAN in human aortic tissue significantly overexpressed in the TAAD patients (p<0.001). Conclusion This study identifies three ceRNA interaction axes, especially VCAN associated with TAAD pathogenesis, providing fundamentals of bioinformatics for understanding the molecular mechanisms of TAAD pathogenesis and developing potential therapeutic strategies for TAAD.
Collapse
Affiliation(s)
- Yuyuan Hu
- Department of Cardiac Surgery, the First Affiliated Hospital of Shandong Second Medical University, Weifang, 261000, People’s Republic of China
| | - Zhenhao Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Yan Qin
- Department of Science and Technology Education, Shanxi Center for Clinical Laboratory, Taiyuan, 030012, People’s Republic of China
| | - Nan Wu
- Department of Cardiac Surgery, Shanxi Provincial People’s Hospital, Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, People’s Republic of China
| | - Tao Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Xinmeng Cheng
- Department of Cardiovascular Surgery, the Affiliated Cardiovascular Hospital of Shanxi Medical University and Shanxi Cardiovascular Hospital (Institute), Taiyuan, Shanxi, 030000, People’s Republic of China
| | - Chunyan Wang
- Department of Clinical Laboratory, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, People’s Republic of China
| | - Xuening Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| |
Collapse
|
15
|
Xie X, Macknight HP, Lu AL, Chalfant CE. RNA splicing variants of the novel long non-coding RNA, CyKILR, possess divergent biological functions in non-small cell lung cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102412. [PMID: 39807365 PMCID: PMC11728077 DOI: 10.1016/j.omtn.2024.102412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
The CDKN2A gene, responsible for encoding the tumor suppressors p16(INK4A) and p14(ARF), is frequently inactivated in non-small cell lung cancer (NSCLC). Herein, an uncharacterized long non-coding RNA (lncRNA) (ENSG00000267053) on chromosome 19p13.12 was found to be overexpressed in NSCLC cells with an active, wild-type CDKN2A gene. This lncRNA, named cyclin-dependent kinase inhibitor 2A-regulated lncRNA (CyKILR), also correlated with an active WT STK11 gene, which encodes the tumor suppressor, liver kinase B1. CyKILR displayed two splice variants, CyKILRa (exon 3 included) and CyKILRb (exon 3 excluded), which are cooperatively regulated by CDKN2A and STK11 as knockdown of both tumor suppressor genes was required to induce a significant loss of exon 3 inclusion in mature CyKILR RNA. CyKILRa localized to the nucleus, and its downregulation using antisense RNA oligonucleotides enhanced cellular proliferation, migration, clonogenic survival, and tumor incidence. In contrast, CyKILRb localized to the cytoplasm, and its downregulation using small interfering RNA reduced cell proliferation, migration, clonogenic survival, and tumor incidence. Transcriptomics analyses revealed the enhancement of apoptotic pathways with concomitant suppression of key cell-cycle pathways by CyKILRa demonstrating its tumor-suppressive role. CyKILRb inhibited tumor suppressor miRNAs indicating an oncogenic nature. These findings elucidate the intricate roles of lncRNAs in cell signaling and tumorigenesis.
Collapse
Affiliation(s)
- Xiujie Xie
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - H. Patrick Macknight
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Amy L. Lu
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Charles E. Chalfant
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA
- Program in Cancer Biology, University of Virginia NCI Comprehensive Cancer Center, Charlottesville, VA 22903, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA 23298, USA
| |
Collapse
|
16
|
Saxena T, Quan A, Chan E, Kozlova N, Matai L, Lee JD, Rupaimoole R, Beca F, Muranen T, Slack FJ. EGFR-induced lncRNA TRIDENT promotes drug resistance in non-small cell lung cancer via phospho-TRIM28-mediated DNA damage repair. Proc Natl Acad Sci U S A 2025; 122:e2415389122. [PMID: 40030013 PMCID: PMC11912419 DOI: 10.1073/pnas.2415389122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) play numerous roles in cellular biology and alterations in lncRNA expression profiles have been implicated in a variety of cancers. Here, we identify and characterize a lncRNA, TRIM28 Interacting DNA damage repair Enhancing Noncoding Transcript (TRIDENT), whose expression is induced upon epithelial growth factor receptor (EGFR) activation, and which exerts pro-oncogenic functions in EGFR-driven non-small cell lung cancer. Knocking down TRIDENT leads to decreased tumor-cell proliferation in both in vitro and in vivo model systems and induces sensitization to chemotherapeutic drugs. Using ChIRP-MS analysis we identified TRIM28 as a protein interactor of TRIDENT. TRIDENT promotes phosphorylation of TRIM28 and knocking down TRIDENT leads to accumulation of DNA damage in cancer cells via decreased TRIM28 phosphorylation. Altogether, our results reveal a molecular pathway in which TRIDENT regulates TRIM28 phosphorylation to promote tumor cell growth and drug resistance. Our findings suggest that TRIDENT can be developed as a biomarker or therapeutic target for EGFR mutant non-small cell lung cancer.
Collapse
Affiliation(s)
- Tanvi Saxena
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Anan Quan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Erica Chan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Nina Kozlova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Latika Matai
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jonathan D. Lee
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Rajesha Rupaimoole
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Francisco Beca
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Frank J. Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
17
|
Liu Q. Role of exercise on the reduction of cancer development: a mechanistic review from the lncRNA point of view. Clin Exp Med 2025; 25:77. [PMID: 40063304 PMCID: PMC11893680 DOI: 10.1007/s10238-025-01618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
More research has been done on the correlation between exercise and cancer, which has revealed several ways that physical activity decreases the risk of developing the disease. The developing function of lncRNAs as an important molecular link between exercise and cancer suppression is the main topic of this review. According to recent research, regular physical exercise also alters the expression levels of several lncRNAs, which are generally elevated in cancer. A complex network of interactions that may provide protective effects against carcinogenesis is suggested by the contribution of these lncRNAs in various cellular processes, such as epigenetic alterations, proliferation, and apoptosis regulation. We offer a comprehensive summary of the existing information regarding specific lncRNAs that are influenced by physical activity and could potentially impact cancer-related processes. We also go over the difficulties in interpreting these alterations, taking into account the fact that several lncRNAs have a dual function in promoting and preventing cancer in various physiological settings. To understand the complex impacts of exercise-induced lncRNA regulation in cancer biology, more study is required. The critique strongly highlights the possibility of lncRNAs serving as both indicators and treatment prospects for cancer-preventive strategies.
Collapse
Affiliation(s)
- Qi Liu
- Nanchang Institute of Technology, Nanchang, 330044, China.
| |
Collapse
|
18
|
Shi M, Zhou R, Shen W, Liang Y, Zhang Y, Liu L, Shao R, Fang Y, Zhao C, Wu L. LncRNA ENST00000581911 Regulates Extraocular Muscle Remodeling by Interacting With KHSRP in Thyroid Eye Disease. Invest Ophthalmol Vis Sci 2025; 66:46. [PMID: 40116677 PMCID: PMC11935560 DOI: 10.1167/iovs.66.3.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Purpose Thyroid eye disease (TED) is a visually debilitating and cosmetically disfiguring orbital disorder, characterized by the remodeling of extraocular muscles (EOMs). This study aimed to investigate the role of long non-coding RNA (lncRNA) ENST00000581911 in the EOMs of TED. Methods LncRNA microarray analysis was performed on EOM tissues sampled from patients with TED and patients with concomitant esotropia. LncRNA ENST00000581911 was identified and subjected to bioinformatics analysis. High-throughput RNA sequencing, CCK-8 assay, CFSE staining, and ELISA were used to investigate the regulatory function of ENST00000581911 in vitro. Furthermore, RNA pull-down, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and western blot (WB) analyses were applied to identify the RNA-binding protein (RBP) interacting with ENST00000581911. Results A total of 1261 lncRNAs were found to be differentially expressed in the EOMs of TED, with 648 upregulated and 613 downregulated lncRNAs. Among these, the upregulated lncRNA ENST00000581911 exhibited the highest expression level, as validated by quantitative real-time PCR (qRT-PCR). Functional analysis demonstrated that ENST00000581911 might be involved in inflammatory response, regulation of muscle contraction, and amino sugar and nucleotide sugar metabolism. RNA sequencing of ENST00000581911-overexpressing and control orbital fibroblasts (OFs) showed that ENST00000581911 might play a regulatory role in DNA replication, extracellular matrix, and cell cycle. Furthermore, KHSRP was identified as the RBP of ENST00000581911. Overexpression of ENST00000581911 promoted cell proliferation and hyaluronic acid secretion in OFs, whereas silencing KHSRP attenuated these effects. Conclusions This study provides novel insights into the role of lncRNA ENST00000581911 in the pathogenesis of EOM remodeling in TED. ENST00000581911 may serve as a potential therapeutic target of TED.
Collapse
Affiliation(s)
- Mingsu Shi
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Rongmei Zhou
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Weiai Shen
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yu Liang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yihan Zhang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Lingyun Liu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Runyi Shao
- School of Basic Medical Science, Fudan University, Shanghai, China
| | - Yanxi Fang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Chen Zhao
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Lianqun Wu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, National Health Commission (NHC), Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
19
|
Li J, Zhao L, Li L, Wang X, Gao Y, Gao Y, Wang J. Urine exosomal lncRNAs as novel biomarkers for early diagnosis of bladder cancer based on microarray differential expression profiling. Int J Biol Markers 2025; 40:24-34. [PMID: 39943913 DOI: 10.1177/03936155251317551] [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] [Indexed: 03/15/2025]
Abstract
PurposeWe aimed to exploit a urine exosomal long non-coding RNAs (lncRNAs) fingerprint to facilitate the early diagnosis of bladder cancer.MethodsMicroarray differential expression profiling of lncRNAs was for the first time employed in urine exosomes from 10 non-muscle-invasive bladder cancer (NMIBC) patients and 10 healthy controls to screen out candidate exosomal lncRNA biomarkers, which were then verified by quantitative real-time polymerase chain reaction in three independent phases including bladder cancer cells, culture fluid and 200 NMIBC participants. Logistic regression was performed to construct a diagnostic model-the diagnostic potency of which was assessed.ResultsThe profile of three exosome-derived lncRNAs (CCDC148-AS1, XLOC_006419, and RP5-1148A21.3) was screened and further verified to be notably over-expressed in NMIBC patients and bladder cancer cell lines, and exhibited area under the receiver-operating characteristic curve values of 0.873, 0.825, and 0.834, respectively, in training, validation, and double-blind validation phases. The profile was superior to urinary cytology in discriminating NMIBC from healthy controls (P < 0.0001). A significant correlation existed between a higher level of CCDC148-AS1 and a higher tumor grade (P < 0.001), and up-regulated CCDC148-AS1 as well as XLOC_006419 were statistically related with tumor node metastasis stage (P = 0.004 and P = 0.031, respectively). These three identified lncRNAs were confirmed to originate from bladder cancer cells and be packaged within exosomes, thus staying sufficiently stable in urine.ConclusionsTumor-originated urine exosomal lncRNAs, as fingerprint in NMIBC, exhibited satisfying clinical significance in early diagnosis of bladder cancer.
Collapse
Affiliation(s)
- Jun Li
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Liming Zhao
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Luning Li
- Department of Gastroenterology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Xiaohua Wang
- Department of General Internal Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Yisheng Gao
- Department of Urology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Yongli Gao
- Department of Oncology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Jinfeng Wang
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| |
Collapse
|
20
|
Jin M, Lu Q, Xia N, Fan X, Zhang Z, Huang X, Sun L, Zhang L, Jiang Z, Yu Q. LncRNA Gm35585 transcriptionally activates the peroxidase EHHADH against diet-induced fatty liver. Exp Mol Med 2025; 57:652-666. [PMID: 40082671 PMCID: PMC11958773 DOI: 10.1038/s12276-025-01420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/13/2024] [Accepted: 01/01/2025] [Indexed: 03/16/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease is one of the most common chronic liver diseases worldwide and has no approved treatment thus far. Here we report that the hepatic overexpression of Gm35585, a novel lncRNA downregulated in the livers of mice fed a high-fat diet, is functionally important in alleviating hepatic lipid accumulation pathologies. Gm35585 activates the peroxisome proliferator-activated receptor α (PPARα) signaling pathway and promotes the expression of downstream PPARα-target gene, enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH), which is one of the four enzymes of the peroxisomal β-oxidation pathway. Activation of EHHADH promotes the oxidation of long-chain fatty acids (LCFAs), and the increased levels of hepatic LCFAs contribute to metabolic-dysfunction-associated steatotic liver disease. Mechanistically, Gm35585 binds to retinoid X receptor α (RXRα) and then forms a PPARα/RXRα heterodimer with PPARα and guides the heterodimer to recognize the promoter of EHHADH, which is called peroxisome proliferator-activated receptor response element, causing transcriptional activation of EHHADH. Taken together, Gm35585 is a hepatic lipid metabolism regulator that activates EHHADH transcription, promoting peroxisomal β-oxidation of LCFAs and ultimately ameliorating diet-induced fatty liver.
Collapse
Affiliation(s)
- Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qian Lu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ninglin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xue Fan
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ziling Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaofei Huang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Li Sun
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China.
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
21
|
Yan D, He Q, Wang C, Li T, Yi X, Yu H, Wu W, Yang H, Wang W, Ma L. miR-135b: A Potential Biomarker for Pathological Diagnosis and Biological Therapy. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70002. [PMID: 40034060 DOI: 10.1002/wrna.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 03/05/2025]
Abstract
MicroRNAs (miRNAs) are a class of endogenous non-coding RNAs found in eukaryotes with post-transcriptional regulatory functions. A variety of miRNAs is differentially expressed in cancer tissues and thus can be used as biomarkers. microRNA-135b-5p (miR-135b) has been shown to be involved in the pathological processes of a variety of neoplastic and non-neoplastic diseases. Under different conditions, miR-135b has different tumor suppressive and carcinogenic effects. miR-135b regulates the development of cancer, including metabolism, proliferation, apoptosis, invasion, fibrosis, angiogenesis, immunomodulation, and drug resistance. miR-135b can be used as a new biomarker for tumor diagnosis and prognosis, which has the potential for clinical guidance. This article reviews the relevant research on miR-135B in the field of tumors, including the biogenesis background of miR-135b, the expression of miR-135b in tumors, and the related targets and signaling pathways of miR-135b mediating tumor progression in order to sort out and explore the clinical transformation value of miR-135b.
Collapse
Affiliation(s)
- Dezhi Yan
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingliu He
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chunjian Wang
- Department of Hematology, Peking University International Hospital, Beijing, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xueping Yi
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Haisheng Yu
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Wu
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hanyun Yang
- Faculty of Health Sciences for Occupational Therapy, Curtin University, West Australia, Australia
| | - Wenzhao Wang
- Department of Orthopedic, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| | - Liang Ma
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
22
|
Cao C, Li A, Xu C, Wu B, Yao L, Liu Y. Engineering artificial non-coding RNAs for targeted protein degradation. Nat Chem Biol 2025; 21:393-401. [PMID: 39215101 DOI: 10.1038/s41589-024-01719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Targeted protein degradation has become a notable drug development strategy, but its application has been limited by the dependence on protein-based chimeras with restricted genetic manipulation capabilities. The use of long non-coding RNAs (lncRNAs) has emerged as a viable alternative, offering interactions with cellular proteins to modulate pathways and enhance degradation capabilities. Here we introduce a strategy employing artificial lncRNAs (alncRNAs) for precise targeted protein degradation. By integrating RNA aptamers and sequences from the lncRNA HOTAIR, our alncRNAs specifically target and facilitate the ubiquitination and degradation of oncogenic transcription factors and tumor-related proteins, such as c-MYC, NF-κB, ETS-1, KRAS and EGFR. These alncRNAs show potential in reducing malignant phenotypes in cells, both in vitro and in vivo, offering advantages in efficiency, adaptability and versatility. This research enhances knowledge of lncRNA-driven protein degradation and presents an effective method for targeted therapies.
Collapse
Affiliation(s)
- Congcong Cao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China
| | - Aolin Li
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Chaojie Xu
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Baorui Wu
- Department of Urology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Lin Yao
- Department of Urology, Peking University First Hospital, Beijing, China.
| | - Yuchen Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
23
|
An Z, Ding W. LncRNA-Gm17586 inhibits S.typhimurium mediated pyroptosis by promoting NLRP3 and Tnip1 binding. Sci Rep 2025; 15:6713. [PMID: 40000898 PMCID: PMC11861967 DOI: 10.1038/s41598-025-91296-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
The aim of this study is to identify the lncRNA that directly interacts with NLRP3 molecules in RAW264.7 cells infected with S. typhimurium and its role in S. typhimurium-mediated pyroptosis. We have identified LncRNA-Gm17586, which directly interacts with NLRP3 in RAW264.7 cells infected with S. typhimurium. LncRNA-Gm17586 inhibits S. typhimurium-mediated pyroptosis in RAW264.7 cells. Overexpression of LncRNA-Gm17586 not only directly suppresses NLRP3 inflammasome activation but also enhances the binding affinity between Tnip1 and NLRP3, thereby indirectly facilitating Tnip1-mediated inhibition of the NLRP3 inflammasome. Consequently, this dual mechanism effectively inhibits S. typhimurium-induced pyroptosis. Our results demonstrate that LncRNA-Gm17586 directly interacts with NLRP3 during S. typhimurium-mediated pyroptosis and inhibits this process by enhancing the interaction between Tnip1 and NLRP3.
Collapse
Affiliation(s)
- Zhiyuan An
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Wenyi Ding
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
24
|
Gu X, Zou Y, Huang Z, Wei M, Ji L. Biochemical biomarkers for the toxicity induced by Traditional Chinese Medicine: A review update. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119315. [PMID: 39755183 DOI: 10.1016/j.jep.2024.119315] [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: 07/10/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) is widely used in China for disease treatment and has become a valuable resource for drug development due to its high efficacy and low risk of side-effects. However, growing toxicity reports has garnered significant global attention. A major challenge in addressing TCM-induced toxicity is lack of specific and sensitive biomarkers for diagnosing and predicting its toxicity. Identifying toxicological biomarkers reflecting TCM-induced toxicity is crucial for timely detection and intervention, and provides significant clues for elucidating the underlying toxic mechanism and key target. AIM OF THE STUDY This article aims to summarize and classify some potential toxicological biomarkers for side-effects induced by TCM and its contained phytochemical ingredients. METHODS The keywords "biomarkers", "traditional Chinese medicine", "Chinese herb", "phytochemical ingredient", "natural product", "toxicity", "hepatotoxicity", "nephrotoxicity", "cardiotoxicity" were used to collect relevant information from literature databases (including PubMed, Web of Science) up to October 2024. RESULTS Research has indicated that more sensitive and specific biomarkers are needed for reflecting TCM's side-effects. PA-protein adducts and AA-DNA adducts could be served as diagnostic biomarkers for hepatotoxicity and nephrotoxicity induced by TCM containing PA and AA, respectively. Multiple miRNAs like miRNA-122-3p, miRNA-5099, and miRNA-21-3p, as well as some endogenous metabolites such as hypoxanthine, choline, and L-valine could be potential biomarkers associated with TCM-induced hepatotoxicity, nephrotoxicity, and cardiotoxicity. CONCLUSION In this review, different research demonstrates that DNA/protein-adducts, noncoding RNAs, endogenous metabolites and so on show the potential to be new early-warning biomarkers for TCM-induced toxicity with high specificity and sensitivity.
Collapse
Affiliation(s)
- Xinnan Gu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Zou
- School of Basic Medical Science of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengjuan Wei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Academy of International Standardization for Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
25
|
Ding C, Chen G, Luan S, Gao R, Fan Y, Zhang Y, Wang X, Li G, Foda MF, Yan J, Li X. Simultaneous profiling of chromatin-associated RNA at targeted DNA loci and RNA-RNA Interactions through TaDRIM-seq. Nat Commun 2025; 16:1500. [PMID: 39929795 PMCID: PMC11811046 DOI: 10.1038/s41467-024-53534-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/09/2024] [Indexed: 02/13/2025] Open
Abstract
Eukaryotic genomes are extensively transcribed into various types of RNAs, many of which are physically associated with chromatin in cis at their transcription sites or in trans to other genomic loci. Emerging roles have been uncovered for these chromatin-associated RNAs (caRNAs) in gene regulation and genome organization, yet they remain challenging to interrogate. Here, we present TaDRIM-seq, a technique employing Protein G (PG)-Tn5-targeted DNA elements and in situ proximity ligation to concurrently probe caRNAs across diverse genomic regions as well as global RNA-RNA interactions within intact nuclei. Notably, this approach diminishes required cell inputs, minimizes hands-on time compared to established methodologies, and is compatible in both mammalian cells and plants. Using this technique, we identify extensive caRNAs at DNA anchor regions associated with chromatin loops and reveal diurnal variation in RNA-DNA and RNA-RNA connectivity networks within rice.
Collapse
Affiliation(s)
- Cheng Ding
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Guoting Chen
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics and Hubei Engineering Technology Research Center of Agricultural Big Data, 3D Genomics Research Center, Huazhong Agricultural University, Wuhan, China
| | - Shiping Luan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Runxin Gao
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yudong Fan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Ying Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Xiaoting Wang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Guoliang Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics and Hubei Engineering Technology Research Center of Agricultural Big Data, 3D Genomics Research Center, Huazhong Agricultural University, Wuhan, China
| | - Mohamed F Foda
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Department of Biochemistry, Faculty of Agriculture, Benha University, Moshtohor, Toukh13736, Egypt
| | - Jiapei Yan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
| | - Xingwang Li
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
26
|
Wang X, Zhang Q, Zhao D, Li X, Yi L, Li S, Wang X, Gu M, Gao J, Jia X. Identification of regulatory genes associated with POAG by integrating expression and sequencing data. Ophthalmic Genet 2025; 46:56-64. [PMID: 39568137 DOI: 10.1080/13816810.2024.2431103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/24/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Primary open-angle glaucoma (POAG) is a subtype of glaucoma that accounts for 60%~70% of all cases. Its pathogenic mechanism is intricate and its pathogenic process is concealed. Numerous significant biological processes associated with POAG continue to be elucidated. METHODS In this study, by exploring the expression data of POAG tissues and normal tissues, we mined the regulatory lncRNAs and mRNAs closely associated with the pathogenesis and progression of POAG by exploring a regulatory network of competing endogenous RNA (ceRNA), established by integrating gene expression data with the known lncRNA-miRNA and miRNA-mRNA-regulatory interactions. The key regulatory pathways and regulatory elements of POAG were identified by topological analysis. Simultaneously, the exome data of 28 cases with POAG and healthy controls were analyzed to identify high-frequency mutations and genes. RESULTS A total of 2712 differentially expressed genes were identified, including 1828 mRNAs and 884 lncRNAs. Network analysis suggested that lncRNAs such as HAGLR, HOTAIR and MIR29B2CHG, and mRNAs such as PPP6R3, BMPR2 and CFL2, may be involved in the onset and progression of POAG. In addition, 55 mutations with potential pathogenicity were identified. CONCLUSION These genes and mutations provide novel potential genetic heterogeneity and genetic susceptibility of POAG, as well as fresh suggestions for elucidating the molecular mechanism underlying the pathogenesis of POAG.
Collapse
Affiliation(s)
- Xizi Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Qiang Zhang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Dongdong Zhao
- Department of Ophthalmology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Xiaofen Li
- Liao Cheng 120 Medical Emergency Command and Dispatch Center, Liaocheng, Shandong, P.R. China
| | - Lili Yi
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Siyuan Li
- Department of Ophthalmology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Xin Wang
- Department of Ophthalmology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Jianlu Gao
- Department of Ophthalmology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| |
Collapse
|
27
|
Chen Y, Ye X, Hu M, Hu Y, Ding J. Long non-coding RNAs in pancreatic cancer. Clin Chim Acta 2025; 566:120040. [PMID: 39536894 DOI: 10.1016/j.cca.2024.120040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
This article reviews the recent advances in pathogenesis, diagnosis and treatment of pancreatic cancer, as well as the relationship between long non-coding RNA (lncRNA) in disease progression. Unfortunately, pancreatic cancer has no early symptoms and quickly invades surrounding tissue and organs, making it one of the deadliest. Accordingly, we urgently need to identify high-risk individuals with precancerous lesions through screening methods to identify early disease, provide better prevention strategies and improve overall survival. LncRNAs have a variety of biological functions in both physiologic and pathophysiologic states including tumor growth, differentiation and proliferation. Herein we review the biological functions, expression patterns, clinical significance and targeted therapy potential of lncRNAs to provide new approaches for diagnosis and treatment in pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Xiaohua Ye
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Minli Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Yibing Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China.
| |
Collapse
|
28
|
Hamamoto K, Zhu G, Lai Q, Lesperance J, Luo H, Li Y, Nigam N, Sharma A, Yang FC, Claxton D, Qiu Y, Aplan PD, Xu M, Huang S. HoxBlinc lncRNA reprograms CTCF-independent TADs to drive leukemic transcription and HSC dysregulation in NUP98-rearranged leukemia. J Clin Invest 2025; 135:e184743. [PMID: 39883527 PMCID: PMC11957699 DOI: 10.1172/jci184743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Although nucleoporin 98 (NUP98) fusion oncogenes often drive aggressive pediatric leukemia by altering chromatin structure and expression of homeobox (HOX) genes, underlying mechanisms remain elusive. Here, we report that the Hoxb-associated lncRNA HoxBlinc was aberrantly activated in NUP98-PHF23 fusion-driven leukemias. HoxBlinc chromatin occupancies led to elevated mixed-lineage leukemia 1 (MLL1) recruitment and aberrant homeotic topologically associated domains (TADs) that enhanced chromatin accessibilities and activated homeotic/hematopoietic oncogenes. HoxBlinc depletion in NUP98 fusion-driven leukemia impaired HoxBlinc binding, TAD integrity, MLL1 recruitment, and the MLL1-driven chromatin signature within HoxBlinc-defined TADs in a CCCTC-binding factor-independent (CTCF-independent) manner, leading to inhibited homeotic/leukemic oncogenes that mitigated NUP98 fusion-driven leukemogenesis in xenografted mouse models. Mechanistically, HoxBlinc overexpression in the mouse hematopoietic compartment induced leukemias resembling those in NUP98-PHF23-knockin (KI) mice via enhancement of HoxBlinc chromatin binding, TAD formation, and Hox gene aberration, leading to expansion of hematopoietic stem and progenitor cell and myeloid/lymphoid cell subpopulations. Thus, our studies reveal a CTCF-independent role of HoxBlinc in leukemic TAD organization and oncogene-regulatory networks.
Collapse
Affiliation(s)
- Karina Hamamoto
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ganqian Zhu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Qian Lai
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Julia Lesperance
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Huacheng Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ying Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nupur Nigam
- Genetics Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Arati Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Feng-Chun Yang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Claxton
- Division of Hematology/Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Yi Qiu
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Peter D. Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
- Penn State Cancer Institute, Hershey, Pennsylvania, USA
| |
Collapse
|
29
|
Yan Q, Wang Q. Exploring the Characters of Non-Coding RNAs in Spermatogenesis and Male Infertility. Int J Mol Sci 2025; 26:1128. [PMID: 39940895 PMCID: PMC11817410 DOI: 10.3390/ijms26031128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Infertility is a widespread clinical problem that affects human reproduction and species persistence worldwide. Around 40-70% of cases are due to male reproductive defects. Functional spermatogenesis (sperm production through several coordinated events) is at the heart of male fertility. Non-coding RNAs (ncRNAs) are the primary regulators of gene expression, controlling extensive critical cellular processes, for example proliferation, differentiation, apoptosis, and reproduction. Due to advancements in high-throughput sequencing tools, many studies have revealed that ncRNAs are widely expressed in germ cells, meiosis, spermatogenesis, sperm fertility, early post-fertilization development, and male infertility. The present review examines the biology and function of ncRNAs, including microRNAs, circular RNAs, and long ncRNAs, in spermatogenesis, their correlation with infertility, and their potential as biomarkers for sperm quality and fertility. The function of ncRNA in Sertoli cells (SCs) and Leydig cells (LCs) is also outlined throughout this study, because spermatogenesis requires testicular somatic cells to be involved in testicular development and male fertility. Meanwhile, the future development of ncRNAs for the clinical treatment of male infertility is also anticipated and discussed.
Collapse
Affiliation(s)
- Qiu Yan
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China;
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Qi Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China;
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
30
|
Liu LH, Chen J, Lai S, Zhao X, Yang M, Wu YR, Zhang Z, Jiang A. Functional RNA mining using random high-throughput screening. Nucleic Acids Res 2025; 53:gkae1173. [PMID: 39673274 PMCID: PMC11754670 DOI: 10.1093/nar/gkae1173] [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: 07/29/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 12/16/2024] Open
Abstract
Functional RNA participates in various life processes in cells. However, there is currently a lack of effective methods to screen for functional RNA. Here, we developed a technology named random high-throughput screening (rHTS). rHTS uses a random library of ∼250-nt synthesized RNA fragments, with high uniformity and abundance. These fragments are circularized into circular RNA by an auto-cyclizing ribozyme to improve their stability. Using rHTS, we successfully screened and identified three RNA fragments contributing significantly to the growth of Escherichia coli, one of which possesses coding potential. Moreover, we found that two noncoding RNAs (ncRNAs) effectively inhibited the growth of E. coli, in vivo rather than in vitro. Subsequently, we applied the rHTS to a coenzyme-dependent screening platform. In this context, two ncRNAs were identified that could effectively promote the conversion from NADPH to NADP+. Exogenous expression of these two ncRNAs was able to increase the conversion rate of glycerol dehydrogenase from glycerol to 1,3-dihydroxyacetone from 18.3% to 21.8% and 23.2%, respectively. These results suggest that rHTS is a powerful technology for functional RNA mining.
Collapse
Affiliation(s)
- Li-Hua Liu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Jinde Chen
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Shijing Lai
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Xuemei Zhao
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Min Yang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Yi-Rui Wu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Zhiqian Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| | - Ao Jiang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Tongchaunghui South District, No. 40, Shangchong South, Haizhu District, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
31
|
Wu L, Yang B, Sun Y, Fan G, Ma L, Ma Y, Xiong X, Zhou H, Wang H, Zhang L, Yang B. Isoprenaline Inhibits Histone Demethylase LSD1 to Induce Cardiac Hypertrophy. Cardiovasc Toxicol 2025; 25:34-47. [PMID: 39521734 DOI: 10.1007/s12012-024-09937-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Histone demethylation in cardiac hypertrophy is poorly understood. This study aims to determine the role of the histone demethylase LSD1 in pathological cardiac hypertrophy. Both isoprenaline (ISO)-treated and transverse aortic constriction (TAC)-treated rats developed hypertrophic hearts. LSD1 was significantly decreased; the histone marks mono- and dimethyl H3K4 and H3K9 (H3K4me1/2 and H3K9me1/2) were significantly up-regulated in the hypertrophic heart tissue, as well as the expression of the ANP, α-HMC and MLV-2v genes. An LSD1 inhibitor, OG-L002 could also induce cardiac hypertrophy and enhance the induction of cardiac hypertrophy by ISO. Overexpressed LSD1 abolished ISO-induced cardiac hypertrophy and downregulated H3K4me1/2 and H3K9me1/2 expression. Overexpression of LSD1 also reduced the expression of ANP, α-HMC and MLV-2v. In addition, we have reported isoprenaline (ISO) as one of the histone demethylase LSD1 inhibitors. This was confirmed by molecular docking, molecular dynamic studies and a histone demethylation assay. The H3K4me1/2 expression increases with the incubation of ISO in HEK 293T and HELA cells. CaMKII could be significantly activated by the LSD1 inhibitor OG-L002 as well as by ISO in rats. In summary, we have identified a novel role for LSD1 in initiating and maintaining cardiac hypertrophy.
Collapse
Affiliation(s)
- Lili Wu
- Department Pathology and Pathophysiology, Logistics University of People's Armed Police Force, Tianjin, 300309, China
| | - Bo Yang
- Department of Cardiology, Tianjin Beichen Hospital, Tianjin, 300134, China
| | - Yingze Sun
- Department of Communication Engineering, College of Electronic Information and Optical Engineering, Nankai University, Tianjin, 300350, China
| | - Guanwei Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lina Ma
- Department of Health Management, Tianjin Hospital, Tianjin, 300211, China
| | - Ying Ma
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xianjia Xiong
- College of Basic Medical Sciences, Tianjin Medical University, 22# Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Hui Zhou
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Huiping Wang
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ling Zhang
- College of Basic Medical Sciences, Tianjin Medical University, 22# Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.
| | - Bing Yang
- College of Basic Medical Sciences, Tianjin Medical University, 22# Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.
- Department of Public Health, International School, Krirk University, 3# Soi Ramintra 1 Anusawari Subdistrict, Bang Khen District, Bangkok, 10220, Thailand.
| |
Collapse
|
32
|
Qin Y, Shirakawa J, Xu C, Chen R, Yang X, Ng C, Nakano S, Elguindy M, Deng Z, Prasanth KV, Eissmann MF, Nakagawa S, Ricci WM, Zhao B. Long non-coding RNA Malat1 fine-tunes bone homeostasis and repair by orchestrating cellular crosstalk and β-catenin-OPG/Jagged1 pathway. eLife 2024; 13:RP98900. [PMID: 39714456 DOI: 10.7554/elife.98900] [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] [Indexed: 12/24/2024] Open
Abstract
The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts, and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.
Collapse
Affiliation(s)
- Yongli Qin
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Jumpei Shirakawa
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Ruge Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, United States
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, United States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Shinichi Nakano
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Mahmoud Elguindy
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer center at Illinois, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Moritz F Eissmann
- Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - William M Ricci
- Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, NewYork, United States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, United States
| |
Collapse
|
33
|
Liu C, Chen J, Huang X, Xia Q, Yang L, Guo J, Tian J, Wang J, Niu Y, Li L, Gou D. lncRNA VELRP Modulates Pulmonary Arterial Smooth Muscle Cell Proliferation and Promotes Vascular Remodeling in Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:2560-2576. [PMID: 39360410 DOI: 10.1161/atvbaha.124.321416] [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: 06/20/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Pulmonary hypertension is a devastating vascular disorder characterized by extensive pulmonary vascular remodeling, ultimately leading to right ventricular failure and death. Activation of PDGF (platelet-derived growth factor) signaling promotes the hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs), thus contributing to the pulmonary vascular remodeling. However, the molecular mechanisms that govern hyperproliferation of PASMCs induced by PDGF remain largely unknown, including the contribution of long noncoding RNAs (lncRNAs). In this study, we aimed to identify a novel lncRNA regulated by PDGF implicated in PASMC proliferation in pulmonary vascular remodeling. METHODS RNA-sequencing analysis was conducted to identify a novel lncRNA named vessel-enriched lncRNA regulated by PDGF-BB (platelet-derived growth factor-BB; VELRP). Functional investigations of VELRP were performed using knockdown and overexpression strategies along with RNA sequencing. Validation of the function and potential mechanisms of VELRP was performed through Western blot, RNA immunoprecipitation, and chromatin immunoprecipitation assays. RESULTS We identified a novel vessel-enriched lncRNA with an increased response to PDGF-BB stimulus. VELRP was identified as an evolutionarily conserved RNA molecule. Modulation of VELRP in PASMCs significantly altered cell proliferation. Mechanistically, VELRP enhances trimethylation of H3K4 (histone H3 lysine 4) by interacting with WDR5 (WD repeat-containing protein 5), leading to increased expression of CDK (cyclin-dependent kinase) 1, CDK2, and CDK4 and consequent hyperproliferation of PASMCs. The pathological relevance of VELRP upregulation in pulmonary artery was confirmed using rat pulmonary hypertension models in vivo, as well as in PASMCs from patients with idiopathic pulmonary arterial hypertension. Specific knockdown of VELRP in smooth muscle cells using adeno-associated virus type 9 SM22α (smooth muscle protein 22α) promoter-shRNA-mediated silencing of VELRP resulted in a significant decrease in right ventricular systolic pressure and vascular remodeling in rat pulmonary hypertension model. CONCLUSIONS VELRP, as an lncRNA upregulated by PDGF-BB, mediates PASMC proliferation via WDR5/CDK signaling. In vivo studies demonstrate that targeted intervention of VELRP in smooth muscle cells can prevent the development of pulmonary hypertension.
Collapse
MESH Headings
- RNA, Long Noncoding/metabolism
- RNA, Long Noncoding/genetics
- Cell Proliferation
- Animals
- Vascular Remodeling
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Becaplermin/pharmacology
- Becaplermin/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Male
- Rats, Sprague-Dawley
- Cells, Cultured
- Disease Models, Animal
- Signal Transduction
- Rats
- Humans
- Histones/metabolism
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/pathology
Collapse
Affiliation(s)
- Cuilian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Xingtao Huang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Qinyi Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jiao Guo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jinglin Tian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Shenzhen University, China
| |
Collapse
|
34
|
Sun Y, Zhen F, Wang H, Liang X, Wang Y, Wang F, Hu J. Exosomal long non-coding RNA-LINC00839 promotes lung adenocarcinoma progression by activating NF-κB signaling pathway. Ann Med 2024; 56:2430029. [PMID: 39582330 PMCID: PMC11590188 DOI: 10.1080/07853890.2024.2430029] [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: 02/09/2024] [Revised: 07/13/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma is the most common type of lung cancer, accounting for approximately 40% of all lung cancer cases, and has the highest incidence among lung cancer subtypes. Recent studies have suggested that long non-coding RNAs (lncRNAs) play a crucial role in the initiation and progression of lung adenocarcinoma. METHODS Based on integrative analysis through databases, we screened Long intergenic non-protein coding RNA 00839 (LINC00839) as one of the most highly upregulated lncRNAs in lung adenocarcinoma. In vitro and in vivo experiments demonstrated that LINC00839 promotes lung adenocarcinoma proliferation, migration, and invasion and that it is present in exosomes secreted by lung adenocarcinoma cells. RESULTS In the cytoplasm, LINC00839 regulates the Toll-like receptor 4 (TLR4)/NF-κB signaling pathway by acting as a molecular sponge of miR-17-5p, thereby influencing the biological behavior of lung adenocarcinoma cells. LINC00839 binds to Polypyrimidine tract binding protein 1 (PTBP1) in the nucleus to regulate the nuclear translocation of NF-κB p65 molecules and, consequently, the transcription of downstream molecules. CONCLUSIONS Our study confirmed that LINC00839 promotes the biological progression of lung adenocarcinoma by performing dual roles in the cytoplasm and nucleus to co-regulate the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yue Sun
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fang Zhen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongyi Wang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiao Liang
- Key laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yaru Wang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Feiran Wang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jing Hu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
35
|
Zolfaghari Dehkharghani M, Mousavi S, Kianifard N, Fazlzadeh A, Parsa H, Tavakoli Pirzaman A, Fazlollahpour-Naghibi A. Importance of long non-coding RNAs in the pathogenesis, diagnosis, and treatment of myocardial infarction. IJC HEART & VASCULATURE 2024; 55:101529. [PMID: 39498345 PMCID: PMC11532444 DOI: 10.1016/j.ijcha.2024.101529] [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: 08/15/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024]
Abstract
Myocardial infarction (MI), a major global cause of mortality and morbidity, continues to pose a significant burden on public health. Despite advances in understanding its pathogenesis, there remains a need to elucidate the intricate molecular mechanisms underlying MI progression. Long non-coding RNAs (lncRNAs) have emerged as key regulators in diverse biological processes, yet their specific roles in MI pathophysiology remain elusive. Conducting a thorough review of literature using PubMed and Google Scholar databases, we investigated the involvement of lncRNAs in MI, focusing on their regulatory functions and downstream signaling pathways. Our analysis revealed extensive dysregulation of lncRNAs in MI, impacting various biological processes through diverse mechanisms. Notably, lncRNAs act as crucial modulators of gene expression and signaling cascades, functioning as decoys, regulators, and scaffolds. Furthermore, studies identified the multifaceted roles of lncRNAs in modulating inflammation, apoptosis, autophagy, necrosis, fibrosis, remodeling, and ischemia-reperfusion injury during MI progression. Recent research highlights the pivotal contribution of lncRNAs to MI pathogenesis, offering novel insights into potential therapeutic interventions. Moreover, the identification of circulating lncRNA signatures holds promise for the development of non-invasive diagnostic biomarkers. In summary, findings underscore the significance of lncRNAs in MI pathophysiology, emphasizing their potential as therapeutic targets and diagnostic tools for improved patient management and outcomes.
Collapse
Affiliation(s)
| | - Safa Mousavi
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazanin Kianifard
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Fazlzadeh
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Parsa
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
36
|
Dakal TC, Kakde GS, Maurya PK. Genomic, epigenomic and transcriptomic landscape of glioblastoma. Metab Brain Dis 2024; 39:1591-1611. [PMID: 39180605 DOI: 10.1007/s11011-024-01414-8] [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: 05/23/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
The mostly aggressive and extremely malignant type of central nervous system is Glioblastoma (GBM), which is characterized by an extremely short average survival time of lesser than 16 months. The primary cause of this phenomenon can be attributed to the extensively altered genome of GBM, which is characterized by the dysregulation of numerous critical signaling pathways and epigenetics regulations associated with proliferation, cellular growth, survival, and apoptosis. In light of this, different genetic alterations in critical signaling pathways and various epigenetics regulation mechanisms are associated with GBM and identified as distinguishing markers. Such GBM prognostic alterations are identified in PI3K/AKT, p53, RTK, RAS, RB, STAT3 and ZIP4 signaling pathways, metabolic pathway (IDH1/2), as well as alterations in epigenetic regulation genes (MGMT, CDKN2A-p16INK4aCDKN2B-p15INK4b). The exploration of innovative diagnostic and therapeutic approaches that specifically target these pathways is utmost importance to enhance the future medication for GBM. This study provides a comprehensive overview of dysregulated epigenetic mechanisms and signaling pathways due to mutations, methylation, and copy number alterations of in critical genes in GBM with prevalence and emphasizing their significance.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, Rajasthan, 313001, India.
| | - Ganesh S Kakde
- Department of Biochemistry, Central University of Haryana, Mahendergarh, 123031, Haryana, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh, 123031, Haryana, India.
| |
Collapse
|
37
|
Alissa M, Aldurayhim M, Abdulaziz O, Alsalmi O, Awad A, Algopishi UB, Alharbi S, Safhi AY, Khan KH, Uffar C. From molecules to heart regeneration: Understanding the complex and profound role of non-coding RNAs in stimulating cardiomyocyte proliferation for cardiac repair. Curr Probl Cardiol 2024; 49:102857. [PMID: 39306148 DOI: 10.1016/j.cpcardiol.2024.102857] [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: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Recent studies of noncoding genomes have shown important implications for regulating gene expression and genetic programs during development and their association with health, including cardiovascular disease. There are nearly 2,500 microRNAs (miRNAs), 12,000 long-chain non-coding RNAs (lncRNA), and nearly 4,000 circular RNAs (circles). Even though they do not code for proteins, they make up nearly 99% of the human genome. Non-coding RNA families (ncRNAs) have recently been discovered and established as novel and necessary controllers of cardiovascular risk factors and cellular processes and, therefore, have the potential to improve the diagnosis and prediction of cardiovascular disease. The increase in the prevalence of cardiovascular disease can be explained by the shortcomings of existing therapies, which focus only on the non-coding RNAs that protein codes for. On the other hand, recent studies point to the possibility of using ncRNAs in the early detection and intervention of CVD. These findings suggest that developing diagnostic tools and therapies based on miRNAs, lncRNAs, and circRNAs will potentially enhance the clinical management of patients with cardiovascular disease. Cardiovascular diseases include CH, HF, RHD, ACS, MI, AS, MF, ARR, and PAH, of which CH is the most common cardiovascular disease, followed by HF and RHD. This paper aims to elucidate the biological and clinical significance of miRNAs, increase, and circles, as well as their expression profiles and the possibility of regulating non-coding transcripts in cardiovascular diseases to improve the application of ncRNAs in diagnosis and treatment.
Collapse
Affiliation(s)
- Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mohammed Aldurayhim
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Ohud Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Alsamghan Awad
- King Khalid University, College of Medicine, Family Medicine department, Saudi Arabia
| | | | - Sarah Alharbi
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khadijah Hassan Khan
- Department of Laboratory, King Faisal Medical Complex, Ministry of Health, Taif 26514, Saudi Arabia
| | - Christin Uffar
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
38
|
Fan L, Zhang L, Zhang X, Wei W, Liu Z. Long Noncoding RNA EMX2-AS Facilitates Osteoblast Differentiation and Bone Formation by Inhibiting EMX2 Protein Translation and Activating Wnt/ β-Catenin Pathway. Stem Cells Int 2024; 2024:4397807. [PMID: 39628661 PMCID: PMC11614513 DOI: 10.1155/sci/4397807] [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: 02/22/2024] [Revised: 06/24/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
Long noncoding RNAs (lncRNAs), as a potentially new and crucial element of biological regulation, have gained widespread attention in recent years. Our previous work identified lncRNA empty spiracles homeobox 2 antisence (EMX2-AS) was significantly increased during the osteoblast differentiation of mesenchymal stem cells (MSCs). Overexpression of lncRNA EMX2-AS promoted osteogenesis in vitro and enhanced heterotopic bone formation in vivo, whereas lncRNA EMX2-AS knockdown had the opposite effect. EMX2 could negatively regulate the osteoblast differentiation of MSCs. lncRNA EMX2-AS was 80% expressed in the cytoplasm during osteoblast differentiation in MSCs. Mechanistic analysis revealed that lncRNA EMX2-AS acts as a positive regulator of osteogenic differentiation through interaction with EMX2 and suppression of its expression at the translational level and Wnt/β-catenin pathway is involved in lncRNA EMX2-AS/EMX2 regulated osteogenic differentiation. Our findings not only provide new targets for the treatment of diseases related to osteoblast differentiation disruption but also enrich the understanding of the regulation mechanisms of lncRNA during stem cell differentiation.
Collapse
Affiliation(s)
- Linyuan Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Li Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Xin Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Wei Wei
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Zhaohui Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| |
Collapse
|
39
|
Xie X, Macknight HP, Lu AL, Chalfant CE. RNA splicing variants of the novel long non-coding RNA, CyKILR, possess divergent biological functions in non-small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602494. [PMID: 39026815 PMCID: PMC11257467 DOI: 10.1101/2024.07.08.602494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The CDKN2A gene, responsible for encoding the tumor suppressors p16(INK4A) and p14(ARF), is frequently inactivated in non-small cell lung cancer (NSCLC). Herein, an uncharacterized long non-coding RNA (lncRNA) (ENSG00000267053) on chromosome 19p13.12 was found to be overexpressed in NSCLC cells with an active, wild-type CDKN2A gene. This lncRNA, named Cy clin-Dependent K inase I nhibitor 2A-regulated l nc R NA (CyKILR), also correlated with an active WT STK11 gene, which encodes the tumor suppressor, Liver kinase B1. CyKILR displayed two splice variants, CyKILRa (exon 3 included) and CyKILRb (exon 3 excluded), which are cooperatively regulated by CDKN2A and STK11 as knockdown of both tumor suppressor genes was required to induce a significant loss of exon 3 inclusion in mature CyKILR RNA. CyKILRa localized to the nucleus, and its downregulation using antisense RNA oligonucleotides enhanced cellular proliferation, migration, clonogenic survival, and tumor incidence. In contrast, CyKILRb localized to the cytoplasm, and its downregulation using siRNA reduced cell proliferation, migration, clonogenic survival, and tumor incidence. Transcriptomics analyses revealed enhancement of apoptotic pathways with concomitant suppression of key cell cycle pathways by CyKILRa demonstrating its tumor-suppressive role. CyKILRb inhibited tumor suppressor microRNAs indicating an oncogenic nature. These findings elucidate the intricate roles of lncRNAs in cell signaling and tumorigenesis.
Collapse
|
40
|
Pei M, Zhang J, Yu Z, Peng Y, Chen Y, Peng S, Wei X, Wu J, Huang X, Xie Y, Yang P, Hong L, Huang X, Wu X, Tang W, Chen Y, Liu S, Lin J, Xiang L, Wang J. LINC02139 interacts with and stabilizes XIAP to regulate cell proliferation and apoptosis in gastric cancer. Commun Biol 2024; 7:1497. [PMID: 39533104 PMCID: PMC11557945 DOI: 10.1038/s42003-024-07202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Previous reports showed that long non-coding RNA (lncRNA) participates in the development and progression of tumors. Nevertheless, the effect of LINC02139 and its mechanism on gastric cancer (GC) is still unknown. We revealed that LINC02139 is upregulated in GC cell lines and tissues and high LINC02139 expression was correlated with the advancement of GC in patients. Functionally, overexpression of LINC02139 promoted, while knockdown of LINC02139 impaired GC cell proliferation, migration, and invasion in vitro and impeded tumorigenesis in a tumor xenograft model in vivo. Mechanistically, LINC02139 directly bound to XIAP and increased the protein level by maintaining its protein stability through inhibition of the ubiquitination and proteasome-dependent degradation pathway. Importantly, the regulatory function of XIAP in LINC02139-mediated oncogenic effects was demonstrated. Both in vitro and in vivo experiments showed that LINC02139 and XIAP collaboratively modulate GC cell growth and apoptosis. Analysis of clinical GC tissues further confirmed the upregulation of XIAP and the positive association between LINC02139 and XIAP expression. These findings established LINC02139 as a driver of tumorigenesis and highlighted the crucial involvement of the LINC02139-XIAP axis in GC progression, suggesting its potential as a promising therapeutic target for combating GC advancement.
Collapse
Affiliation(s)
- Miaomiao Pei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Jieming Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhen Yu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ying Peng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yidong Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Siyang Peng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiangyang Wei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jieke Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaodong Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanci Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ping Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Linjie Hong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoting Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510515, China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weimei Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ye Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Jianjiao Lin
- Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| | - Li Xiang
- Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| |
Collapse
|
41
|
Feng N, Mandal A, Jambhale A, Narnur P, Chen G, Akula N, Kramer R, Kolachana B, Xu Q, McMahon FJ, Lipska BK, Auluck PK, Marenco S. Schizophrenia risk-associated SNPs affect expression of microRNA 137 host gene: a postmortem study. Hum Mol Genet 2024; 33:1939-1947. [PMID: 39239979 PMCID: PMC12102068 DOI: 10.1093/hmg/ddae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Common variants in the MicroRNA 137 host gene MIR137HG and its adjacent gene DPYD have been associated with schizophrenia risk and the latest Psychiatric Genomics Consortium (PGC). Genome-Wide Association Study on schizophrenia has confirmed and extended these findings. To elucidate the association of schizophrenia risk-associated SNPs in this genomic region, we examined the expression of both mature and immature transcripts of the miR-137 host gene (MIR137HG) in the dorsolateral prefrontal cortex (DLPFC) and subgenual anterior cingulate cortex (sgACC) of postmortem brain samples of donors with schizophrenia and psychiatrically-unaffected controls using qPCR and RNA-Seq approaches. No differential expression of miR-137, MIR137HG, or its transcripts was observed. Two schizophrenia risk-associated SNPs identified in the PGC study, rs11165917 (DLPFC: P = 2.0e-16; sgACC: P = 6.4e-10) and rs4274102 (DLPFC: P = 0.036; sgACC: P = 0.002), were associated with expression of the MIR137HG long non-coding RNA transcript MIR137HG-203 (ENST00000602672.2) in individuals of European ancestry. Carriers of the minor (risk) allele of rs11165917 had significantly lower expression of MIR137HG-203 compared with those carrying the major allele. However, we were unable to validate this result by short-read sequencing of RNA extracted from DLPFC or sgACC tissue. This finding suggests that immature transcripts of MIR137HG may contribute to genetic risk for schizophrenia.
Collapse
Affiliation(s)
- Ningping Feng
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Ajeet Mandal
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Ananya Jambhale
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Pranav Narnur
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Gang Chen
- Scientific and Statistical Computing Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, bldg 10, room 1D73, Bethesda, MD 20892, United States
| | - Nirmala Akula
- Human Genetics Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 35 Convent Dr. Bldg. 35, RM 1A202, MSC 3719, Bethesda, MD 20892, United States
| | - Robin Kramer
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Bhaskar Kolachana
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Qing Xu
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 35 Convent Dr. Bldg. 35, RM 1A202, MSC 3719, Bethesda, MD 20892, United States
| | - Barbara K Lipska
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Pavan K Auluck
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Stefano Marenco
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| |
Collapse
|
42
|
Liu R, Wang X, Zhou M, Zhai J, Sun J. PSF-lncRNA interaction as a target for novel targeted anticancer therapies. Biomed Pharmacother 2024; 180:117491. [PMID: 39332189 DOI: 10.1016/j.biopha.2024.117491] [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: 05/19/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
The Polypyrimidine Tract-Binding Protein-Associated Splicing Factor (PSF), a component of the Drosophila Behavior/Human Splicing (DBHS) complex, plays a pivotal role in cancer pathogenesis. The epigenetic regulation mediated by PSF and long noncoding RNA (lncRNA), along with PSF's alternative splicing activity, has been implicated in promoting cancer cell proliferation, migration, invasion, metastasis, and drug resistance in various human cancers. Recent research highlights the therapeutic promise of targeting the PSF-lncRNA interaction to combat aggressive malignancies, making it a compelling target for cancer therapy. This review offers a detailed synthesis of the current understanding of PSF's role in oncogenic pathways and recent progress in identifying inhibitors of PSF-lncRNA interactions. Furthermore, it discusses the potential of using these inhibitors in cancer treatment strategies, especially as adjuncts to immune checkpoint blockade therapies to improve the efficacy of anti-PD-(L)1 treatments in Glioblastoma Multiforme (GBM). By outlining the interaction patterns of existing PSF-lncRNA inhibitors, this article aims to guide the development and refinement of future pharmacological interventions.
Collapse
Affiliation(s)
- Ren Liu
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Min Zhou
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Jingfang Zhai
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
43
|
Wang Z, Tan W, Li B, Chen J, Zhu J, Xu F, Tang F, Yoshida S, Zhou Y. LncRNA-MM2P regulates retinal neovascularization through M2 macrophage polarization. Exp Eye Res 2024; 248:110072. [PMID: 39241859 DOI: 10.1016/j.exer.2024.110072] [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: 04/13/2024] [Revised: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The study aims to investigate the effects and potential mechanisms of lncRNA-MM2P on retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). The OIR model was established in C57BL/6J mice. RAW264.7 cell line and bone marrow-derived macrophages (BMDMs) from mice were used for in vitro studies. RT-qPCR was used to analyze the expressions of lncRNA and mRNAs. The protein expression levels were determined by western blotting. The size of avascular areas and neovascular tufts were assessed based on isolectin B4 immunofluorescence staining images. The human retinal endothelial cells (HRECs) were used to evaluate the proliferation, migration, and tube formation of endothelial cells. The expression of lncRNA-MM2P was significantly upregulated from P17 to P25 in OIR retinas. Knockdown of lncRNA-MM2P levels in vivo led to a significant reduction in the neovascular tufts and avascular areas in the retinas of OIR mice. Knockdown of lncRNA-MM2P levels in vitro suppressed the expression of M2 markers in macrophages. Moreover, we found a significant inhibition of avascular areas and neovascular tufts in OIR mice injected intravitreally with M2 macrophages treated by shRNA-MM2P. The cellular functions of proliferation, migration, and tube formation were significantly attenuated in HRECs cultured with a supernatant of shRNA-MM2P-treated M2 macrophages. Our results indicate that lncRNA-MM2P regulates retinal neovascularization by inducing M2 polarization of macrophages in OIR mice. Therefore, lncRNA-MM2P may be a potential molecular target for immunoregulation of retinal neovascularization.
Collapse
Affiliation(s)
- Zicong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Fan Xu
- Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health, Nanning, Guangxi, 530021, China
| | - Fen Tang
- Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health, Nanning, Guangxi, 530021, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
44
|
Xu Q, Wang L, Song Q, Chen S, Du K, Teng X, Zou C. Distinct Hippocampal Expression Profiles of lncRNAs in Obese Type 2 Diabetes Mice Exhibiting Cognitive Impairment. Neuromolecular Med 2024; 26:42. [PMID: 39470862 DOI: 10.1007/s12017-024-08811-3] [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: 05/08/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
Cognitive dysfunction has been accepted as a possible complication of type 2 diabetes (T2D), but few studies revealed the potential roles of Long non‑coding RNAs (lncRNAs) in cognitive dysfunction in T2D. The current research aims to demonstrate the specific expression patterns of lncRNA-mRNA in the hippocampi of T2D db/db mice exhibiting cognitive impairment. In this study, the results from behavioral tests showed that T2D db/db mice displayed short-term and spatial working memory deficits compared to db/m mice. Furthermore, western blot analysis demonstrated that compared with db/m mice, p-GSK3β (ser9) protein levels were markedly elevated in T2D db/db mice (P < 0.01). In addition, though not statistically significant, the ratio of p-Tau (Ser396) to Tau 46, α-Synuclein expression, and p-GSK3α (ser21) expression were also relatively higher in T2D db/db mice than in db/m mice. The microarray profiling revealed that 75 lncRNAs and 26 mRNAs were dysregulated in T2D db/db mice (> 2.0 fold change, P < 0.05). GO analysis demonstrated that the differentially expressed mRNAs participated in immune response, extracellular membrane-bounded organelle, and extracellular region. KEGG analysis revealed that the differentially expressed mRNAs were mainly involved in one carbon pool by folate, glyoxylate and dicarboxylate metabolism, autophagy, glycine, serine and threonine metabolism, and B cell receptor signaling pathway. A lncRNA‑mRNA coexpression network containing 71 lncRNAs and 26 mRNAs was built to investigate the interaction between lncRNA and mRNA. Collectively, these results revealed the differential hippocampal expression profiles of lncRNAs in T2D mice with cognitive dysfunction, and the findings from this study provide new clues for exploring the potential roles of lncRNAs in the pathogenesis of cognitive dysfunction in T2D.
Collapse
Affiliation(s)
- Qianqian Xu
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lihui Wang
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qiong Song
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shuai Chen
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Kechen Du
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiahong Teng
- School of International Education, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Chunlin Zou
- Key Laboratory of Longevity and Aging-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, Nanning, Guangxi, China.
- Department of Human Anatomy, Institute of Neuroscience and Guangxi Key Laboratory of Brain Science, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
45
|
Song J, Cui Q, Gao J. Roles of lncRNAs related to the p53 network in breast cancer progression. Front Oncol 2024; 14:1453807. [PMID: 39479021 PMCID: PMC11521785 DOI: 10.3389/fonc.2024.1453807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
The p53 is a crucial tumor suppressor and transcription factor that participates in apoptosis and senescence. It can be activated upon DNA damage to regulate the expression of a series of genes. Previous studies have demonstrated that some specific lncRNAs are part of the TP53 regulatory network. To enhance our understanding of the relationship between lncRNAs and P53 in cancers, we review the localization, structure, and function of some lncRNAs that are related to the mechanisms of the p53 pathway or serve as p53 transcriptional targets.
Collapse
Affiliation(s)
| | - Qiuxia Cui
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
46
|
Qin Y, Shirakawa J, Xu C, Chen R, Yang X, Ng C, Nakano S, Elguindy M, Deng Z, Prasanth KV, Eissmann MF, Nakagawa S, Ricci WM, Zhao B. Long non-coding RNA Malat1 fine-tunes bone homeostasis and repair by orchestrating cellular crosstalk and the β-catenin-OPG/Jagged1 pathway. RESEARCH SQUARE 2024:rs.3.rs-3793919. [PMID: 38234849 PMCID: PMC10793491 DOI: 10.21203/rs.3.rs-3793919/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.
Collapse
Affiliation(s)
- Yongli Qin
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jumpei Shirakawa
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Ruge Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Shinichi Nakano
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Mahmoud Elguindy
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Moritz F. Eissmann
- Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, 60596 Frankfurt, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - William M. Ricci
- Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| |
Collapse
|
47
|
Hong P, Wang D, Wu Y, Zhang Q, Liu P, Pan J, Yu M, Tian W. A novel long noncoding RNA AK029592 contributes to thermogenic adipocyte differentiation. Stem Cells Transl Med 2024; 13:985-1000. [PMID: 39115701 PMCID: PMC11465168 DOI: 10.1093/stcltm/szae056] [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: 03/16/2024] [Accepted: 06/29/2024] [Indexed: 10/11/2024] Open
Abstract
Exploration of factors originating from brown adipose tissue that govern the thermogenic adipocyte differentiation is imperative for comprehending the regulatory framework underlying brown fat biogenesis and for devising therapeutic approaches for metabolic disorders associated with obesity. Prior evidence has illuminated the pivotal role of long noncoding RNAs (lncRNAs) in orchestrating thermogenesis within adipose tissue. Here, we aimed to explore and identify the critical lncRNA that could promote thermogenic adipocyte differentiation and to provide a novel strategy to treat obesity-related metabolic diseases in the future. In this study, through amalgamation with our previous lncRNA microarray data from small extracellular vesicles derived from BAT (sEV-BAT), we have identified sEV-BAT-enriched lncRNA AK029592 as a critical constituent of the thermogenic program, which actively fostered beige adipocyte differentiation and enhanced the thermogenic capacities of adipose tissue. Moreover, lncRNA AK029592 could sponge miR-199a-5p in adipocytes to stimulate thermogenic gene expression. Consequently, we concluded lncRNA AK029592 as a crucial lncRNA component of the thermogenic program that regulated beige adipocyte differentiation and white adipose tissue browning, thereby providing a novel therapeutic target and strategy in combating obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Pengyu Hong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Dianri Wang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, People’s Republic of China
| | - Yue Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Qi Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Pan Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Jian Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Mei Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| |
Collapse
|
48
|
Zheng H, Cheng J, Zhuang Z, Li D, Yang J, Yuan F, Fan X, Liu X. A disulfidptosis-related lncRNA signature for analyzing tumor microenvironment and clinical prognosis in hepatocellular carcinoma. Front Immunol 2024; 15:1412277. [PMID: 39434887 PMCID: PMC11491388 DOI: 10.3389/fimmu.2024.1412277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Disulfidptosis is a recently identified form of non-apoptotic programmed cell death which distinguishes itself from classical cell death pathways. However, the prognostic implications of disulfidptosis-related long non-coding RNAs (DRLs) and their underlying mechanisms in hepatocellular carcinoma (HCC) remain largely unexplored. Methods In this study, we leveraged RNA-sequencing data and clinical information of HCC patients from the TCGA database. Through expression correlation and prognostic correlation analyses, we identified a set of top-performing long non-coding RNAs. Subsequently, a 5-DRLs predictive signature was established by conducting a Lasso regression analysis. Results This signature effectively stratified patients into high- and low-risk groups, revealing notable differences in survival outcomes. Further validation through univariate and multivariate Cox regression analyses confirmed that the risk score derived from our signature independently predicted the prognosis of HCC patients. Moreover, we observed significant disparities in immune cell infiltration and tumor mutation burden (TMB) between the two risk groups, shedding light on the potential connection between immune-related mechanisms and disulfidptosis. Notably, the signature also exhibited predictive value in the context of chemotherapeutic drug sensitivity and immunotherapy efficacy for HCC patients. Finally, we performed experimental validation at both cellular and patient levels and successfully induced a disulfidptosis phenotype in HCC cells. Discussion In general, this multifaceted approach provides a comprehensive overview of DRLs profiles in HCC, culminating in the establishment of a novel risk signature that holds promise for predicting prognosis and therapy outcomes of HCC patients.
Collapse
Affiliation(s)
- Haishui Zheng
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jigan Cheng
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziyun Zhuang
- Shantou University Medical College, Shantou, China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital.Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Yuan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaolong Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
49
|
Fu Z, Chen Y, Cai G, Peng H, Wang X, Li P, Gu A, Li Y, Ma D. An Antisense Long Non-Coding RNA, LncRsn, Is Involved in Sexual Reproduction and Full Virulence in Fusarium graminearum. J Fungi (Basel) 2024; 10:692. [PMID: 39452644 PMCID: PMC11508260 DOI: 10.3390/jof10100692] [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: 08/13/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Fusarium head blight (FHB), primarily caused by Fusarium graminearum, is a devastating crop disease that leads to significant declines in wheat yield and quality worldwide. Long non-coding RNAs (lncRNAs) are found to play significant functions in various biological processes, but their regulatory functions in the sexual reproduction and pathogenicity of F. graminearum have not been studied extensively. This study identified an antisense lncRNA, named lncRsn, located in the transcription initiation site region between the 5'-flanking gene FgSna and the 3'-flanking gene FgPta. A deletion mutant of lncRsn (ΔlncRsn) was constructed through homologous recombination. ΔlncRsn exhibited huge reductions in pathogen and sexual reproduction. Additionally, the deletion of lncRsn disrupted the biosynthesis of deoxynivalenol (DON) and impaired the formation of infection structures. RT-qPCR analysis reveals that lncRsn may negatively regulate the transcription of the target gene FgSna. This study found that lncRsn plays an important role in sexual and asexual reproduction, pathogenicity, virulence, osmotic stress, and cell wall integrity (CWI) in F. graminearum. Further characterization of pathogenesis-related genes and the reaction between lncRsn and protein-coding genes will aid in developing novel approaches for controlling F. graminearum diseases.
Collapse
Affiliation(s)
- Zhizhen Fu
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Yanjie Chen
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Gaolei Cai
- Shiyan Academy of Agricultural Sciences, Shiyan 442000, China;
| | - Huijuan Peng
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Xiaoyu Wang
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Ping Li
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Aiguo Gu
- Jiangsu Product Quality Testing & Inspection Institute, 5 Guanghua Street, Nanjing 210007, China;
| | - Yanli Li
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| | - Dongfang Ma
- Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River, College of Agriculture, Yangtze University, Jingzhou 434025, China; (Z.F.); (Y.C.); (H.P.); (X.W.); (P.L.)
| |
Collapse
|
50
|
Lluch A, Latorre J, Oliveras-Cañellas N, Fernández-Sánchez A, Moreno-Navarrete JM, Castells-Nobau A, Comas F, Buxò M, Rodríguez-Hermosa JI, Ballester M, Espadas I, Martín-Montalvo A, Zhang B, Zhou Y, Burkhardt R, Höring M, Liebisch G, Castellanos-Rubio A, Santin I, Kar A, Laakso M, Pajukanta P, Olkkonen VM, Fernández-Real JM, Ortega FJ. A novel long non-coding RNA connects obesity to impaired adipocyte function. Mol Metab 2024; 90:102040. [PMID: 39362599 PMCID: PMC11544081 DOI: 10.1016/j.molmet.2024.102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) can perform tasks of key relevance in fat cells, contributing, when defective, to the burden of obesity and its sequelae. Here, scrutiny of adipose tissue transcriptomes before and after bariatric surgery (GSE53378) granted identification of 496 lncRNAs linked to the obese phenotype. Only expression of linc-GALNTL6-4 displayed an average recovery over 2-fold and FDR-adjusted p-value <0.0001 after weight loss. The aim of the present study was to investigate the impact on adipocyte function and potential clinical value of impaired adipose linc-GALNTL6-4 in obese subjects. METHODS We employed transcriptomic analysis of public dataset GSE199063, and cross validations in two large transversal cohorts to report evidence of a previously unknown association of adipose linc-GALNTL6-4 with obesity. We then performed functional analyses in human adipocyte cultures, genome-wide transcriptomics, and untargeted lipidomics in cell models of loss and gain of function to explore the molecular implications of its associations with obesity and weight loss. RESULTS The expression of linc-GALNTL6-4 in human adipose tissue is adipocyte-specific and co-segregates with obesity, being normalized upon weight loss. This co-segregation is demonstrated in two longitudinal weight loss studies and two cross-sectional samples. While compromised expression of linc-GALNTL6-4 in obese subjects is primarily due to the inflammatory component in the context of obesity, adipogenesis requires the transcriptional upregulation of linc-GALNTL6-4, the expression of which reaches an apex in terminally differentiated adipocytes. Functionally, we demonstrated that the knockdown of linc-GALNTL6-4 impairs adipogenesis, induces alterations in the lipidome, and leads to the downregulation of genes related to cell cycle, while propelling in adipocytes inflammation, impaired fatty acid metabolism, and altered gene expression patterns, including that of apolipoprotein C1 (APOC1). Conversely, the genetic gain of linc-GALNTL6-4 ameliorated differentiation and adipocyte phenotype, putatively by constraining APOC1, also contributing to the metabolism of triglycerides in adipose. CONCLUSIONS Current data unveil the unforeseen connection of adipocyte-specific linc-GALNTL6-4 as a modulator of lipid homeostasis challenged by excessive body weight and meta-inflammation.
Collapse
Affiliation(s)
- Aina Lluch
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Jèssica Latorre
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.
| | - Núria Oliveras-Cañellas
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | | | - José M Moreno-Navarrete
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Anna Castells-Nobau
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Ferran Comas
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain
| | - Maria Buxò
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain
| | - José I Rodríguez-Hermosa
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; School of Medicine, University of Girona (UdG), Girona, Spain
| | - María Ballester
- Animal Breeding and Genetics Programme, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, Caldes de Montbui, Spain
| | - Isabel Espadas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), University Pablo de Olavide, Seville, Spain
| | - Alejandro Martín-Montalvo
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), University Pablo de Olavide, Seville, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Birong Zhang
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - You Zhou
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ainara Castellanos-Rubio
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Izortze Santin
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Bizkaia, Spain; Instituto de Investigación Sanitaria Biocruces Bizkaia, Bizkaia, Spain
| | - Asha Kar
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles (CA), USA; Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles (CA), USA; Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, University of Helsinki, Helsinki, Finland
| | - José M Fernández-Real
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain; School of Medicine, University of Girona (UdG), Girona, Spain.
| | - Francisco J Ortega
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.
| |
Collapse
|