101
|
Li W, Yang S, Liu H, Cao Z, Xu F, Ning C, Zhang Q, Wang D, Tang H. Identification of key LncRNAs and mRNAs associated with intramuscular fat in pig via WGCNA. BMC Genomics 2025; 26:233. [PMID: 40069611 PMCID: PMC11895181 DOI: 10.1186/s12864-025-11427-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/02/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Intramuscular fat (IMF) not only directly affects the tenderness, juiciness, and overall flavour of meat but also plays a significant role in influencing consumer preferences for pork. Therefore, exploring key biomarkers that influence IMF deposition is highly important for breeding high-quality pork. IMF is a typical quantitative trait that is regulated by the interaction of multiple coding and noncoding RNAs. Traditional differential analysis methods typically focus on individual genes, making it difficult to identify key genes and their underlying mechanisms accurately. Weighted gene coexpression network analysis (WGCNA) is an efficient and accurate method for identifying and characterizing key pathways and genes associated with complex traits. Therefore, the aim of this study was to construct an mRNA‒lncRNA coexpression network related to IMF using WGCNA to explore and identify potential candidate genes that influence IMF in pigs. RESULTS Full-length transcriptome sequencing was performed on 31 220-day-old Jiangquan black pigs raised in the same environment, and a gene expression matrix comprising 25,609 genes was constructed. Nine coexpression modules were identified through WGCNA, with the number of genes in these modules ranging from 33 to 3648. The magenta module (corr = 0.7, P < 0.01) and the turquoise module (corr = -0.77, P < 0.01) were significantly associated with IMF deposition. Hub genes in each module were identified on the basis of the screening criteria of GS > 0.4 and MM > 0.8. Combined with enrichment analysis and protein‒protein interaction (PPI) analysis, 18 key mRNAs potentially related to IMF were selected: CRKL, CBL, PDGFRB, DOCK1, YWHAH, HSP90AB1, LOC100524873, NDUFA1, NDUFA11, NDUFA12, NDUFA2, NDUFAB1, NDUFB10, NDUFB3, NDUFB7, NDUFS5, NDUFS6, and UQCR10. To explore the regulatory role of lncRNAs in the process of IMF deposition, we constructed an lncRNA‒mRNA‒pathway network on the basis of the relationships between lncRNAs and key mRNAs, as well as the results of Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. This network includes four key lncRNAs (TGOLN2, LOC100521518, LOC100524915, and LOC100622481) and predicts the potential mechanisms by which lncRNAs regulate IMF deposition. CONCLUSIONS Through WGCNA, enrichment analysis, and PPI analysis, 18 mRNAs and four lncRNAs potentially involved in IMF deposition were identified, and the lncRNA regulatory pathways were preliminarily explored. Our findings provide new insights into the regulatory mechanisms of pig IMF deposition and lay the foundation for further exploration of the molecular mechanisms underlying pig fat deposition.
Collapse
Affiliation(s)
- Wenqiang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Suozhou Yang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Huixin Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Zhi Cao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Fei Xu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Chao Ning
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China
| | - Dan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China.
| | - Hui Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China.
| |
Collapse
|
102
|
Li J, Ju Y, Zou Q, Ni F. lncRNA localization and feature interpretability analysis. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102425. [PMID: 39926317 PMCID: PMC11803160 DOI: 10.1016/j.omtn.2024.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 02/11/2025]
Abstract
Subcellular localization is crucial for understanding the functions and regulatory mechanisms of biomolecules. Long non-coding RNAs (lncRNAs) have diverse roles in cellular processes, and their localization within specific subcellular compartments provides insights into their biological functions and implications in health and disease. The nucleolus and nucleoplasm are key hubs for RNA metabolism and cellular regulation. We developed a model, LncDNN, for identifying the localization of lncRNAs in the nucleolus and nucleoplasm. LncDNN uses three different encoding schemes and employs Shapley Additive Explanations for feature analysis and selection. The results show that LncDNN is more accurate than other models. Additionally, an interpretable analysis of the features influencing the model was conducted. LncDNN is applicable for identifying the localization of lncRNA in the nucleolus and nucleoplasm, aiding in the understanding and in-depth study of related biological processes and functions.
Collapse
Affiliation(s)
- Jing Li
- Department of Microbiology, University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Ying Ju
- School of Informatics, Xiamen University, Xiamen, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, Zhejiang, China
| | - Fengming Ni
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
103
|
Yang QH, Fu YQ, Feng WL, Mao JF, Xu N, Liu Q, Yan QJ, Yang HJ, Zhang XP. LncRNA-MALAT1 promotes triple-negative breast cancer progression and function as ceRNA to target REEP5 by sponging miR-106a-5p. Eur J Med Res 2025; 30:159. [PMID: 40059235 PMCID: PMC11892299 DOI: 10.1186/s40001-025-02420-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/01/2025] [Indexed: 05/13/2025] Open
Abstract
Axillary lymph node metastasis (ALNM) in triple negative breast cancer (TNBC) will lead to poor prognosis. Recent studies have shown that long non-coding RNAs (lncRNAs) were involved in the progression of tumors. This study aimed to explore the role and mechanism of lncRNA-MALAT1 in the progression of TNBC and its relationship with ALNM. MALAT1 is highly expressed in TNBC cells lines, tumor tissues and serum, and it is positively correlated with the degree of ALNM. In addition, MALAT1 can act as a competitive endogenous RNA (ceRNA) that regulates cellular biological behavior by competitively binding to miR-106a-5p with REEP5. In conclusion, our results show that MALAT1 could function as ceRNA promote the proliferation, invasion and metastasis of TNBC cells through MALAT1/miR-106a-5p/REEP5 axis, which is expected to provide new ideas for the diagnosis of TNBC in clinic.
Collapse
Affiliation(s)
- Qiu-Hui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310006, Zhejiang, China
| | - Ye-Qin Fu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Wei-Liang Feng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jie-Fei Mao
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Ning Xu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qing Liu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310006, Zhejiang, China
| | - Qian-Jun Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310006, Zhejiang, China
| | - Hong-Jian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xi-Ping Zhang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
104
|
Hossam Abdelmonem B, Kamal LT, Wardy LW, Ragheb M, Hanna MM, Elsharkawy M, Abdelnaser A. Non-coding RNAs: emerging biomarkers and therapeutic targets in cancer and inflammatory diseases. Front Oncol 2025; 15:1534862. [PMID: 40129920 PMCID: PMC11931079 DOI: 10.3389/fonc.2025.1534862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Non-coding RNAs (ncRNAs) have a significant role in gene regulation, especially in cancer and inflammatory diseases. ncRNAs, such as microRNA, long non-coding RNAs, and circular RNAs, alter the transcriptional, post-transcriptional, and epigenetic gene expression levels. These molecules act as biomarkers and possible therapeutic targets because aberrant ncRNA expression has been directly connected to tumor progression, metastasis, and response to therapy in cancer research. ncRNAs' interactions with multiple cellular pathways, including MAPK, Wnt, and PI3K/AKT/mTOR, impact cellular processes like proliferation, apoptosis, and immune responses. The potential of RNA-based therapeutics, such as anti-microRNA and microRNA mimics, to restore normal gene expression is being actively studied. Additionally, the tissue-specific expression patterns of ncRNAs offer unique opportunities for targeted therapy. Specificity, stability, and immune responses are obstacles to the therapeutic use of ncRNAs; however, novel strategies, such as modified oligonucleotides and targeted delivery systems, are being developed. ncRNA profiling may result in more individualized and successful treatments as precision medicine advances, improving patient outcomes and creating early diagnosis and monitoring opportunities. The current review aims to investigate the roles of ncRNAs as potential biomarkers and therapeutic targets in cancer and inflammatory diseases, focusing on their mechanisms in gene regulation and their implications for non-invasive diagnostics and targeted therapies. A comprehensive literature review was conducted using PubMed and Google Scholar, focusing on research published between 2014 and 2025. Studies were selected based on rigorous inclusion criteria, including peer-reviewed status and relevance to ncRNA roles in cancer and inflammatory diseases. Non-English, non-peer-reviewed, and inconclusive studies were excluded. This approach ensures that the findings presented are based on high-quality and relevant sources.
Collapse
Affiliation(s)
- Basma Hossam Abdelmonem
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- Basic Sciences Department, Faculty of Physical Therapy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Lereen T. Kamal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Lilian Waheed Wardy
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- Research and Development Department, Eva Pharma for Pharmaceuticals Industries, Cairo, Egypt
| | - Manon Ragheb
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- School of Medicine, New Giza University (NGU), Giza, Egypt
| | - Mireille M. Hanna
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Mohamed Elsharkawy
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| |
Collapse
|
105
|
Hsu CY, Jamal A, Kamal MA, Ahmad F, Bokov DO, Mustafa YF, Saud A, Kulsum SN, Jawad MA, Gabble BC. Pathological roles of lncRNA HOTAIR in liver cancer: An updated review. Gene 2025; 940:149180. [PMID: 39708931 DOI: 10.1016/j.gene.2024.149180] [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: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Liver cancer ranks as the sixth most prevalent form of cancer and stands as the fourth leading cause of cancer-related fatalities on a global scale. The two primary types of liver cancer are hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). While ICC originates from the bile ducts, HCC develops from hepatocytes, which are the primary functional cells of the liver. In cases where liver cancer is detected in its early stages, it can be effectively treated through locoregional interventions such as surgical resection, Radiofrequency Ablation, Transarterial chemoembolization, or liver transplantation. However, HCC is typically diagnosed at advanced stages, rendering these treatment options ineffective due to the unresectable nature of the tumor. LncRNAs, a novel class of RNA molecules and epigenetic regulators, have emerged as key players in the development and advancement of different types of tumors. They exert their influence by regulating the expression of downstream genes in cancer-related signaling pathways, thereby promoting the proliferation, migration, and invasion of tumor cells. Additionally, these transcripts have the ability to modify the activity and expression of tumor suppressors and oncogenes, further contributing to tumorigenesis. Recently, growing numbers of experiments have demonstrated the elevated expression of HOX antisense intergenic RNA (HOTAIR), a spliced and poly-adenylated lncRNA, in liver cancers and its association with cancer patient's prognosis and overall survival, as well as tumor cells' growth, metastasis, and resistance to therapies. This updated review will summarize molecular pathways by which lncRNA HOTAIR promotes liver cancer development, and highlight its diagnostic and therapeutic potential, though.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ 85004, USA
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713, Saudi Arabia
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy named after A.P. Nelyubin, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow 119991, Russian Federation; Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow 109240, Russian Federation
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | | | - Syeda Nazia Kulsum
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | | | - Baneen C Gabble
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
106
|
Esmaeili A, Yazdanpanah N, Rezaei N. LncRNAs Orchestrating Neuroinflammation: A Comprehensive Review. Cell Mol Neurobiol 2025; 45:21. [PMID: 40056236 PMCID: PMC11890384 DOI: 10.1007/s10571-025-01538-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/17/2025] [Indexed: 03/10/2025]
Abstract
CNS diseases account for a major part of the comorbidity and mortality of the human population; moreover, neuroinflammation has become an indication for different CNS diseases, for instance, Parkinson's and Alzheimer's disease. Microglia and astrocytes are the two main glial cells that can be found in the CNS. Each of these plays an important role in mediating immune responses like inflammation. There are many studies suggesting the role of LncRNAs in mediating neuroinflammation. Indeed, LncRNAs orchestrate neuroinflammation through various mechanisms, namely miRNA sponge, and transcriptional activation/inhibition. In addition, LncRNAs regulate different downstream pathways like NF-κB, and PI3K/AKT. In this study, we gathered the existing studies regarding the mechanisms of action of LncRNAs in the pathogenesis of different CNS diseases like neurodegenerative diseases and traumatic injuries through regulating neuroinflammation. We aim to elaborate on the regulatory roles of LncRNAs in neuroinflammation and bring a more profound understanding of the etiology of CNS diseases in terms of neuroinflammation.
Collapse
Affiliation(s)
- Arash Esmaeili
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
107
|
Zhao X, Zhang H, Liu Y, Li L, Wei H. Study on the metastatic mechanism of LINC00115 in adenocarcinoma of the Esophagogastric junction. Hum Mol Genet 2025; 34:492-511. [PMID: 39807637 DOI: 10.1093/hmg/ddae193] [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: 06/15/2024] [Revised: 10/16/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Adenocarcinoma of the esophagogastric junction (AEG) is a common and deadly cancer, and an in-depth investigation of its molecular mechanisms of metastasis is crucial for discovering new therapeutic targets. This study explores the role of the long non-coding RNA (lncRNA) LINC00115 in AEG metastasis and its underlying mechanisms. Through the analysis of 108 pairs of AEG cancer tissues and matched adjacent tissues, we found a significant upregulation of LINC00115 in AEG tissues, closely associated with TNM staging and lymph node metastasis. Utilizing cell counting kit-8 (CCK-8) assays, colony formation experiments, wound healing assays, flow cytometry for apoptosis and cell cycle analysis, and Transwell assays, we have confirmed that LINC00115 significantly promotes proliferation, migration, and invasion of AEG cells in vitro. Animal experiments further validate the role of LINC00115 in promoting tumor growth and metastasis in vivo. Additionally, our nuclear-cytoplasmic fractionation experiments and RNA fluorescence in situ hybridization (FISH) reveal that LINC00115, along with its interacting protein KH-Type splicing regulatory protein (KHSRP), predominantly localizes to the cell nucleus. By conducting RNA pull-down assays and mass spectrometry (MS) analysis, we have identified a direct interaction between LINC00115 and KHSRP protein and further determined their binding sites through catRAPID and ENCORI databases. This study provides evidence of LINC00115 as a novel biomarker and potential therapeutic target for AEG and offers a fresh perspective on understanding the molecular mechanisms of AEG metastasis.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Gastroenterology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | - Haifeng Zhang
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | | | - Li Li
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
- Department of Thoracic Surgery, Huaihe Hospital of Henan University/Henan University School of Nursing and Health, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | - Haitao Wei
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| |
Collapse
|
108
|
Qin T, Lei C, Xiao H, Yang J, Luo Q, Hu L, Chen F, Long M, Zhang H. lncRNA ACVR2B-AS1 modulates thyroid cancer progression by regulating miR-195-5p. Discov Oncol 2025; 16:267. [PMID: 40045085 PMCID: PMC11883067 DOI: 10.1007/s12672-025-01993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND lncRNAs are key regulators in thyroid cancer (TC). While lncRNA ACVR2B-AS1 has been proposed as a potential TC biomarker, its role remains underexplored. This study aims to clarify its clinical significance in TC and investigate its molecular mechanism. MATERIALS AND METHODS qRT-PCR was used to assess the expression of ACVR2B-AS1 in TC tissues and cell lines. Kaplan-Meier survival curves and Cox regression were utilized to assess the prognostic value of ACVR2B-AS1 expression. The interaction between ACVR2B-AS1 and miR-195-5p, as well as their effects on cell viability, migration, and invasion, were evaluated using dual-luciferase reporter assays, CCK-8 assays, and Transwell assays. RESULTS ACVR2B-AS1 was significantly upregulated in TC tissues and cell lines, and its expression correlated with TNM stage and lymph node metastasis. Elevated ACVR2B-AS1 levels were associated with poor survival outcomes, and it was identified as an independent risk factor for TC progression. A direct regulatory relationship was established between ACVR2B-AS1 and miR-195-5p, with ACVR2B-AS1 negatively regulating miR-195-5p, thereby promoting TC cell proliferation, migration, and invasion. FGF2 was predicted and validated as a target gene of miR-195-5p. CONCLUSION lncRNA ACVR2B-AS1 shows potential as a prognostic marker in TC and may regulate tumor progression through the miR-195-5p/FGF2 axis, offering new insights for TC diagnosis and treatment.
Collapse
Affiliation(s)
- Tianshi Qin
- Department of General Surgery, Ningbo Hangzhou Bay Hospital, Ningbo, 315336, China
| | - Chengqiang Lei
- Department of Thoracic and Cardiac Surgery, The People's Hospital of Dazu, Chongqing/The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Henghua Xiao
- Department of Ultrasound, Central Hospital of Hengyang, No.12, Yancheng Road, Yanfeng District, Hengyang, 421000, Hunan, China
| | - Jun Yang
- Department of Ultrasound, Central Hospital of Hengyang, No.12, Yancheng Road, Yanfeng District, Hengyang, 421000, Hunan, China
| | - Qiong Luo
- General Surgery III, Affiliated Hengyang Hospital of Hunan Normal University (Central Hospital of Hengyang), Hengyang, 421000, China
- Hunan Provincial Clinical Medical Technology Demonstration Base for Prevention and Treatment of Thyroid Diseases, Shaoyang, 421000, China
| | - Lingli Hu
- Department of Ultrasound, Central Hospital of Hengyang, No.12, Yancheng Road, Yanfeng District, Hengyang, 421000, Hunan, China
| | - Fang Chen
- Department of Ultrasound, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Manlong Long
- Department of Ultrasound, Central Hospital of Hengyang, No.12, Yancheng Road, Yanfeng District, Hengyang, 421000, Hunan, China.
| | - Huayi Zhang
- Department of Radiation Oncology, The First People's Hospital of Yongkang, No.599, Jinshan West Road, Yongkang City, Jinhua City, 321300, Zhejiang, China.
| |
Collapse
|
109
|
Wang Y, Pei W, Yang Y, Xia C, Zhang Q, Geng Z, Shi X, Wang F. Inhibition of XIST restrains paclitaxel resistance in breast cancer cells by targeting hsa-let-7d-5p/ATG16L1 through regulation of autophagy. Cell Signal 2025; 127:111534. [PMID: 39638138 DOI: 10.1016/j.cellsig.2024.111534] [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: 06/03/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer is a fatal malignant tumor in women worldwide. The development of paclitaxel resistance remains a challenge. Autophagy is considered to have a significant part in the chemotherapeutic stress mechanism. This study aimed to investigate the function of long non-coding RNA (lncRNA) in breast cancer cell chemoresistance and autophagy. The paclitaxel (PTX)-resistant breast cancer cells were established. The function of X-inactive specific transcript (XIST) was demonstrated using in vitro and in vivo experiments. Transmission electron microscope (TEM) was used to observe autophagy vesicles. Protein and mRNA levels were determined using western blotting and quantitative real time polymerase chain reaction (qRT-PCR). We discovered that autophagic activity was correlated with chemoresistance in PTX-resistant breast cancer cells. In vitro and in vivo studies showed that XIST inhibition reduced cell resistance to paclitaxel, caused autophagy to be suppressed by regulating hsa-let-7d-5p and ATG16L1 expression. Mechanically, threonine protein kinase B (PKB; also known as AKT) - mammalian target of rapamycin (mTOR) pathway was activated when knockdown of XIST, while was reversed by inhibition of hsa-let-7d-5p. Our results verified that XIST played a significant role in developing chemoresistance via mediating autophagy in PTX-resistant breast cancer cells. It may be a potential target for breast cancer treatment strategies.
Collapse
Affiliation(s)
- Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, Anhui, China
| | - Wenhao Pei
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Yuping Yang
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Chaoqun Xia
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Qiang Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Xiuru Shi
- Department of Blood Transfusion, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Fengchao Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China.
| |
Collapse
|
110
|
Roy B, Verma AK, Funahashi Y, Dwivedi Y. Deciphering the epigenetic role of long non-coding RNAs in mood disorders: Focus on human brain studies. Clin Transl Med 2025; 15:e70135. [PMID: 40038891 PMCID: PMC11879898 DOI: 10.1002/ctm2.70135] [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: 08/22/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 03/06/2025] Open
Abstract
Epigenetics plays a central role in neuropsychiatric disorders, contributing significantly to their complexity and manifestation. Major depressive disorder (MDD) and bipolar disorder (BD) have profound impact on mood, affect and cognition. Emerging evidence suggests that epigenetic modification of genes plays a pivotal role in the pathogenesis of both MDD and BD. Long non-coding RNAs (lncRNA) constitute a heterogeneous class of transcripts and have emerged as crucial regulators of epigenetic processes, offering promising insights into the pathophysiology of various diseases. Despite their limited coding potential, lncRNAs are known to play a critical role in achieving global transcriptomic regulation in a spatiotemporal fashion, especially in complex tissue like the brain. This review aims to discuss the specific dysregulation of lncRNAs so far observed in the brains of MDD and BD patients and understand their mechanistic contributions to the disease pathogenesis. KEY POINTS: Brain-centric lncRNAs regulate gene networks, and their disruption is linked to MDD. In MDD, altered lncRNAs disrupt gene regulation by changing chromatin looping or modifying chromatin accessibility. These changes lead to neuronal dysfunction, affecting neural circuitry and synaptic plasticity. The result is impaired brain function, contributing to the symptoms of MDD.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral NeurobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anuj K. Verma
- Department of Psychiatry and Behavioral NeurobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yu Funahashi
- Department of Psychiatry and Behavioral NeurobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of MedicineToonEhimeJapan
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral NeurobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
111
|
Wang C, Wu H, Xie Y, Wang J, Huang L, Ni X, Deng S, Zhang Y, Chen X, Zhang H, Yuan S, Tang L. Berberine Inhibits Migration and Apoptosis of Rat Podocytes in Diabetic Nephropathy via the Novel lncRNA LOC102549726 Related Pathway. Phytother Res 2025; 39:1615-1631. [PMID: 39916312 DOI: 10.1002/ptr.8452] [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: 04/19/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 03/11/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) stands as one of the most severe complications of diabetes. Podocytes injury, particularly its attachment to the lateral glomerular basement membrane, serves as a crucial indicator of DN. Growing evidence suggests that berberine (BBR) can mitigate the onset and progression of DN. However, the molecular mechanisms through which BBR exerts its beneficial effects in the treatment of DN remain incompletely elucidated. PURPOSE To explore the underlying mechanisms by which BBR exerts its therapeutic effects in DN. METHODS High-throughput lncRNA sequencing on the renal cortex of both the DN model group and the normal SD group was performed to dig for differentially expressed lncRNAs. The expression of LOC102549726 was evaluated using qPCR. The biological functions of LOC102549726 were analyzed in podocyets and DN rats. The bioinformatics techniques, qPCR and WB were used to explore the potential molecular mechanisms. RESULTS We found that lncRNA LOC102549726 was highly expressed in renal cortex of DN rats and podocytes subjected to high glucose conditions. Silencing LOC102549726 inhibited migration and apoptosis of podocytes. Mechanistically, LOC102549726 was identified as a facilitator of the expression of EGF and forkhead box O1 (FOXO1). BBR, a known therapeutic agent for DN, exhibited the ability to diminish the level of LOC102549726, EGF and FOXO1 in both DN rats and podocytes. CONCLUSION Our findings suggested that BBR suppresses migration and apoptosis of podocytes in DN through targeting the LOC102549726/EGF/FOXO1 axis. This sheds light on a potential therapeutic avenue for mitigating the impact of DN on podocyte function.
Collapse
Affiliation(s)
- Chen Wang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Hao Wu
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Yongsheng Xie
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Jiajia Wang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Lingzhi Huang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Shujun Deng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Yang Zhang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Xinyi Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Huihui Zhang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Siming Yuan
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| |
Collapse
|
112
|
Ding D, Zhao G. KLF9 aggravates the cardiomyocyte hypertrophy in hypertrophic obstructive cardiomyopathy through the lncRNA UCA1/p27 axis. Int J Exp Pathol 2025; 106:e12526. [PMID: 39909852 PMCID: PMC11798666 DOI: 10.1111/iep.12526] [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: 11/12/2024] [Accepted: 01/05/2025] [Indexed: 02/07/2025] Open
Abstract
Cardiac hypertrophy refers to an abnormal increase in the thickness of the heart muscle. Our study explores the role of Krüppel-like factor 9 (KLF9) in hypertrophic obstructive cardiomyopathy (HOCM)-induced cardiomyocyte hypertrophy, providing new targets for the treatment of HOCM. Cardiomyocytes were treated with isoproterenol (ISO). The levels of natriuretic peptide B (BNP)/natriuretic peptide A (ANP)/KLF9/long non-coding RNA urothelial carcinoma-associated 1 (lncRNA UCA1)/p27 were measured. Cell surface area and protein/DNA ratio were tested. The binding between KLF9 and the lncRNA UCA1 promoter and between zeste homologue 2 (EZH2) and lncRNA UCA1 was verified. The enrichment of histone H3 lysine 27 tri-methylation (H3K27me3) and EZH2 on the p27 promoter was analysed. ISO treatment increased KLF9 and lncRNA UCA1 expression and decreased p27 expression in cardiomyocytes. KLF9 knockdown inhibited ISO-induced cardiomyocyte hypertrophy, reduced ANP and BNP expression, and alleviated cardiomyocyte damage. KLF9 activated lncRNA UCA1 expression. LncRNA UCA1 recruited EZH2 to the p27 promoter region, increasing the enrichment of H3K27me3, thereby epigenetically suppressing p27 expression. LncRNA UCA1 overexpression or p27 downregulation reduced the protective effect of KLF9 downregulation on cardiomyocyte hypertrophy. In conclusion, KLF9 activates lncRNA UCA1 expression, and lncRNA UCA1 epigenetically suppresses p27 expression, thereby exacerbating cardiomyocyte hypertrophy in HOCM.
Collapse
Affiliation(s)
- Dayou Ding
- School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Guangrong Zhao
- School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| |
Collapse
|
113
|
Shao ZC, Sun WK, Deng QQ, Cheng L, Huang X, Hu LK, Li HN. Identification of Key lncRNAs in Gout Under Copper Death and Iron Death Mechanisms: A Study Based on ceRNA Network Analysis and Random Forest Algorithm. Mol Biotechnol 2025; 67:996-1013. [PMID: 38472694 DOI: 10.1007/s12033-024-01099-5] [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: 01/17/2024] [Indexed: 03/14/2024]
Abstract
This study focused on identifying potential key lncRNAs associated with gout under the mechanisms of copper death and iron death through ceRNA network analysis and Random Forest (RF) algorithm, which aimed to provide new insights into the molecular mechanisms of gout, and potential molecular targets for future therapeutic strategies of gout. Initially, we conducted an in-depth bioinformatics analysis of gout microarray chips to screen the key cuproptosis-related genes (CRGs) and key ferroptosis-related genes (FRGs). Using these data, we constructed a key ceRNA network for gout. Finally, key lncRNAs associated with gout were identified through the RF algorithm combined with ROC curves, and validated using the Comparative Toxicogenomics Database (CTD). We successfully identified NLRP3, LIPT1, and DBT as key CRGs associated with gout, and G6PD, PRKAA1, LIG3, PHF21A, KLF2, PGRMC1, JUN, PANX2, and AR as key FRGs associated with gout. The key ceRNA network identified four downregulated key lncRNAs (SEPSECS-AS1, LINC01054, REV3L-IT1, and ZNF883) along with three downregulated mRNAs (DBT, AR, and PRKAA1) based on the ceRNA theory. According to CTD validation inference scores and biological functions of target mRNAs, we identified a potential gout-associated lncRNA ZNF883/hsa-miR-539-5p/PRKAA1 regulatory axis. This study identified the key lncRNA ZNF883 in the context of copper death and iron death mechanisms related to gout for the first time through the application of ceRNA network analysis and the RF algorithm, thereby filling a research gap in this field and providing new insights into the molecular mechanisms of gout. We further found that lncRNA ZNF883 might function in gout patients by regulating PRKAA1, the mechanism of which was potentially related to uric acid reabsorption in the proximal renal tubules and inflammation regulation. The proposed lncRNA ZNF883/hsa-miR-539-5p/PRKAA1 regulatory axis might represent a potential RNA regulatory pathway for controlling the progression of gout disease. This discovery offered new molecular targets for the treatment of gout, and had significant implications for future therapeutic strategies in managing the gout.
Collapse
Affiliation(s)
- Zi-Chen Shao
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Wei-Kang Sun
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Qin-Qin Deng
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Ling Cheng
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Xin Huang
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Lie-Kui Hu
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Hua-Nan Li
- Affiliated Hospital of Jiangxi University of Chinese Medicine, No.445, Bayi Avenue, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
114
|
Zhang Y, Song JY, Sun ZG. Exploring the impact of environmental factors on male reproductive health through epigenetics. Reprod Toxicol 2025; 132:108832. [PMID: 39778664 DOI: 10.1016/j.reprotox.2025.108832] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Male infertility has become an increasingly severe global health issue, with its incidence significantly rising over the past few decades. This paper delves into the crucial role of epigenetics in male reproductive health, focusing particularly on the effects of DNA methylation, histone modifications, chromatin remodeling and non-coding RNAs regulation on spermatogenesis. Exposure to various environmental factors can cause sperm DNA damage, leading to epigenetic abnormalities. Among these factors, we have discussed heavy metals (including Zinc, Cadmium, Arsenic, Copper), phthalates, electromagnetic radiation, and temperature in detail. Notably, aberrations in DNA methylation are closely associated with various symptoms of male infertility, and histone modifications and chromatin remodeling are essential for sperm maturation and function. By synthesizing existing literature and experimental data, this narrative review investigates how environmental factors influence male reproductive health through epigenetic mechanisms, thus providing new theoretical foundations and practical guidelines for the early diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Yi Zhang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Jing-Yan Song
- Reproductive and Genetic Center, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Zhen-Gao Sun
- Reproductive and Genetic Center, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
115
|
Zhang M, Chen Q, Cheng K, Zhang W, Chen Y. Long non-coding RNA XR008038 promotes the myocardial ischemia/reperfusion injury development through increasing the expressions of galectin-3. Int J Cardiol 2025; 422:132955. [PMID: 39755333 DOI: 10.1016/j.ijcard.2024.132955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Myocardial ischemia/reperfusion (I/R) injury is a common pathophysiological change after myocardial reperfusion therapy. Recent research confirmed that long non-coding RNA (IncRNAs) played an important role in many cardiovascular diseases. This study was carried out to explore the role of lncRNA XR008038 in the I/R progression. METHODS GSE103731 database was downloaded from NCBI Gene Expression Omnibus to analyze the differently expressed lncRNAs. Cell viability was determined by CCK-8 assay. Cell apoptosis was detected by flow cytometry and TUNEL staining. Northern blot and qRT-PCR was carried out to detect the XR008038 levels. The mitochondrial membrane potential was assessed by JC-1 staining. Western blot was conducted to measure the expression of apoptosis related proteins. RNA pull down and RIP assay was carried out to explore the relationship between XR008038 and galectin-3. RESULTS The results showed that XR008038 was up-regulated in the H/R treated H9c2 cells and the myocardial tissues of the I/R rats. XR008038 silencing promoted the cell growth and mitochondrial membrane potential, inhibited the cell apoptosis of the H/R treated H9c2 cells. Additionally, the MDA content was decreased and SOD activity was enhanced in the H/R treated H9c2 cells and the myocardial tissues of the I/R rats after XR008038 knockdown. XR008038 interacted with galectin-3 and further regulated the mRNA stability of galectin-3. Galectin-3 overexpression neutralized the role of si-XR008038 in the H/R treated H9c2 cells. CONCLUSION In conclusion, XR008038 promoted the oxidative damage in I/R progression through regulating the galectin-3 levels.
Collapse
Affiliation(s)
- Meiqi Zhang
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, No.453 Tiyuchang Road, Hangzhou, Zhejiang 310013, China
| | - Qilan Chen
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, No.453 Tiyuchang Road, Hangzhou, Zhejiang 310013, China
| | - Kang Cheng
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, No.453 Tiyuchang Road, Hangzhou, Zhejiang 310013, China
| | - Wei Zhang
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, No.453 Tiyuchang Road, Hangzhou, Zhejiang 310013, China
| | - Yun Chen
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, No.453 Tiyuchang Road, Hangzhou, Zhejiang 310013, China.
| |
Collapse
|
116
|
Zhu J, Jian Z, Liu F, Le L. The emerging landscape of small nucleolar RNA host gene 10 in cancer mechanistic insights and clinical relevance. Cell Signal 2025; 127:111590. [PMID: 39798772 DOI: 10.1016/j.cellsig.2025.111590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/14/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Small nucleolar RNA host gene 10 (SNHG10) is a newly recognized long non-coding RNA (lncRNA) with significant implications in cancer biology. Abnormal expression of SNHG10 has been observed in various solid tumors and hematological malignancies. Research conducted in vivo and in vitro has revealed that SNHG10 plays a pivotal role in numerous biological processes, including cell proliferation, apoptosis, invasion and migration, drug resistance, energy metabolism, immune evasion, as well as tumor growth and metastasis. SNHG10 regulates tumor development through several mechanisms, such as competing with microRNA (miRNA) for binding sites, modulating various signaling pathways, influencing transcriptional activity, and affecting epigenetic regulation. The diverse biological functions and intricate mechanisms of SNHG10 highlight its considerable clinical relevance, positioning it as a potential pan-cancer biomarker and therapeutic target. This review aims to summarize the role of SNHG10 in tumorigenesis and cancer progression, clarify the molecular mechanisms at play, and explore its clinical significance in cancer diagnosis and prognosis prediction, along with its therapeutic potential.
Collapse
Affiliation(s)
- Jingyu Zhu
- Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, China
| | - Zihao Jian
- Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, China
| | - Fangteng Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China.
| | - Lulu Le
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, Jiangxi, China.
| |
Collapse
|
117
|
Hsu CY, Jasim SA, Bansal P, Kaur H, Ahmad I, Saud A, Deorari M, Al-Mashhadani ZI, Kumar A, Zwamel AH. Delving Into lncRNA-Mediated Regulation of Autophagy-Associated Signaling Pathways in the Context of Breast Cancer. Cell Biol Int 2025; 49:221-234. [PMID: 39873206 DOI: 10.1002/cbin.12277] [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: 05/11/2024] [Revised: 12/15/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
Breast cancer is a multifaceted and prevalent malignancy, impacting a considerable proportion of women globally. Numerous signaling pathways intricately regulate cellular functions such as growth, proliferation, and survival. Among the various regulators, lncRNAs have emerged as significant players despite their inability to encode proteins. An expanding body of literature underscores the pivotal roles lncRNAs play in cancer biology, particularly in the context of breast cancer. Autophagy, the cellular process dedicated to the degradation and recycling of cellular components, is now recognized as a crucial factor in cancer initiation and progression. The interplay between lncRNAs, various signaling pathways, and autophagy in the pathophysiology of breast cancer remains an active area of investigation. Researchers have identified specific lncRNAs that are dysregulated in breast cancer patients, influencing the modulation of key signaling pathways. Using experimental methodologies and bioinformatics approaches, multiple lncRNAs have been elucidated, providing deeper insights into their contributions to breast cancer pathogenesis and metastatic processes. In summary, the pathophysiological landscape of breast cancer is characterized by the complex interactions involving lncRNA-mediated autophagy. This understanding paves the way for identifying novel therapeutic targets, prognostic markers, and diagnostic markers, ultimately contributing to improved treatment outcomes in breast cancer management.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, Arizona, USA
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University College, Anbar, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Abdulnaser Saud
- Department of Pharmacy, Al-Hadi University College, Baghdad, Iraq
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, Russia
- Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Ahmed Hussein Zwamel
- Department of Medical Laboratory Technology, College of Medical Technology, The Islamic University, Najaf, Iraq
| |
Collapse
|
118
|
Gao H, Fan L, Gai X, Fu R, Li G, Jing K, Xu J, Sun S. LINC01004/hsa-mir-125b-2-3p axis restrains ferroptosis in hepatocellular carcinoma by targeting HSPA4 via ceRNA mechanism. Technol Health Care 2025; 33:959-973. [PMID: 40105158 DOI: 10.1177/09287329241291430] [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: 03/20/2025]
Abstract
BackgroundHepatocellular carcinoma (HCC) is a primary cancer, accounting for 90% of primary liver cancer, mainly occurring in patients with cirrhosis and chronic liver disease.ObjectiveTo investigate the latent mechanisms of hepatocellular carcinoma (HCC) and find therapeutic targets.MethodsDifferentially expressed and overall survival related genes of HCC, and cell death related genes were intersected to obtain latent target genes. These genes were analyzed using ROC curve for diagnosing HCC. RT-qPCR and Western blot were performed to detect the expression level of genes. Wound healing tests were performed with or without si-HSPA4. Potential ceRNA axis was forecasted using TargetScan and miRanda and the dual luciferase reporter gene assay was used to verify the results. Finally, the images of H&E dye liquor-stained HCC tissue section, the CT images for patients in different tumor stage.ResultsLINC01004/hsa-miR-125b-2-3p/HSPA4 axis was forecasted and then was verified using dual-luciferase reporter assay. HSPA4 knockdown caused significant reduction of cell proliferation and ferroptosis. Si-HSPA4 related ferroptosis was generated through impairing iron transport via targeting restrain GPX4. For human subjects, the RT-qPCR analysis revealed the that the larger the tumor diameter, the higher the LINC01004, HSPA4, and GPX4 expression, and the lower the hsa-miR-125b-2-3p expression.ConclusionLINC01004/hsa-miR-125b-2-3p/HSPA4 regulatory axis involved in the ferroptosis of the progression of HCC via GPX4 dependent method, providing new therapeutic targets for HCC patients.
Collapse
Affiliation(s)
- Han Gao
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, China
| | - Li Fan
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xue Gai
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Rong Fu
- Departmentof Oncology, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Guohua Li
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Kexin Jing
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Jingwei Xu
- General Surgery, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Shengjian Sun
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
119
|
Wang S, Yu ZG, Han GS, Sun XG. CFPLncLoc: A multi-label lncRNA subcellular localization prediction based on Chaos game representation and centralized feature pyramid. Int J Biol Macromol 2025; 297:139519. [PMID: 39761904 DOI: 10.1016/j.ijbiomac.2025.139519] [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: 11/13/2024] [Revised: 01/01/2025] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
There is increasing evidence that the subcellular localization of long noncoding RNAs (lncRNAs) can provide valuable insights into their biological functions. In terms of transcriptomes, lncRNAs were usually found in multiple subcellular localizations. Although several computational methods have been developed to predict the subcellular localization of lncRNAs, few of them were designed for lncRNAs that have multiple subcellular localizations. In this study, we propose a novel deep learning model, called CFPLncLoc, which uses chaos game representation (CGR) images of lncRNA sequences to predict multi-label lncRNA subcellular localization. CFPLncLoc utilizes image update strategy (IUS) to enhance the relative feature representation of the CGR images. To extract higher-level features from CGR images, CFPLncLoc introduces the multi-scale feature fusion (MFF) model, centralized feature pyramid (CFP), from the field of computer vision (CV). Ablation studies confirmed the contribution of the IUS and CFP in improving the prediction performance. Statistical test results verify that CFPLncLoc outperforms existing state-of-the-art predictors under the evaluation metric MaAUC on the hold-out/independent test set. The source code can be obtained from https://github.com/ShengWang-XTU/CFPLncLoc.
Collapse
Affiliation(s)
- Sheng Wang
- National Center for Applied Mathematics in Hunan, Xiangtan University, Hunan 411105, China; Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Hunan 411105, China
| | - Zu-Guo Yu
- National Center for Applied Mathematics in Hunan, Xiangtan University, Hunan 411105, China; Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Hunan 411105, China.
| | - Guo-Sheng Han
- National Center for Applied Mathematics in Hunan, Xiangtan University, Hunan 411105, China; Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Hunan 411105, China.
| | - Xin-Gen Sun
- National Center for Applied Mathematics in Hunan, Xiangtan University, Hunan 411105, China; Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Hunan 411105, China
| |
Collapse
|
120
|
Yan C, Li Y, Li X, Li Y, Zhang Y, Sun J, Ding Q, Zhao X, Li B. LncRNA MALAT1/Calpain-1 Axis in ATO Induced hERG Channel Deficiency. Drug Des Devel Ther 2025; 19:1475-1487. [PMID: 40041754 PMCID: PMC11878130 DOI: 10.2147/dddt.s502776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/21/2025] [Indexed: 05/12/2025] Open
Abstract
Background KCNH2 encodes the hERG potassium channel, which is associated with drug-induced long QT syndrome. Arsenic trioxide (ATO) is an effective therapeutic agent for acute promyelocytic leukemia; however, its long-term use can lead to cardiotoxicity, particularly in cases of acquired long QT syndrome (acLQTS), which may result in torsade de pointes (TdP). Therefore, it is essential to comprehend the mechanisms behind acLQTS and to develop effective preventive and therapeutic strategies. Aim This study sought to investigate the role and molecular mechanism of MALAT1 in ATO-induced acLQTS. Furthermore, it sought to identify pharmacological agents that could mitigate the cardiotoxic effects of ATO and establish viable intervention targets for the prevention and management of acLQTS. Methods First, we employed gene chip arrays to identify target long noncoding RNAs (lncRNAs). Subsequently, we performed quantitative qRT-PCR and RNA-binding protein immunoprecipitation (RIP) to assess lncRNA levels. Next, we utilized Western blotting for protein expression analysis, and finally, we conducted whole-cell patch-clamp recordings to evaluate hERG currents. Results Our results revealed a significant upregulation of lncRNA MALAT1 expression in HEK293-hERG cells treated with ATO. Mechanistically, MALAT1 interacts with calpain-1, inhibiting its ubiquitin-mediated degradation and enhancing the cleavage activity of calpain-1 on the hERG channel. FEX and TAN were found to mitigate the effects of ATO on the MALAT1/calpain-1 pathway, ultimately restoring hERG protein levels. Conclusion This study demonstrated that ATO-induced enhancement of calpain-1 and reduction of hERG may be linked to the aberrant overexpression of lncRNA MALAT1. Tanshinone IIA and fexofenadine restored the hERG protein levels potentially by decreasing MALAT1 expression and counteracting ATO's effects on the MALAT1/calpain-1 pathway. Collectively, our research uncovers a previously unreported regulatory mechanism underlying ATO-induced acLQTS. Moreover, it identifies potential molecular targets and intervention strategies for acLQTS therapy.
Collapse
Affiliation(s)
- Caichuan Yan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Yuexin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Xiaoxu Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Yang Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Yuhao Zhang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Jinyang Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Qirui Ding
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Xin Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| | - Baoxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
121
|
Shi L, Li B, Tan J, Zhu L, Zhang S, Zhang Y, Xiang M, Li J, Chen Y, Han X, Xie J, Tang Y, Rosie Xing H, Li J, Wang J. Exosomal lncRNA Mir100hg from lung cancer stem cells activates H3K14 lactylation to enhance metastatic activity in non-stem lung cancer cells. J Nanobiotechnology 2025; 23:156. [PMID: 40022086 PMCID: PMC11869636 DOI: 10.1186/s12951-025-03198-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025] Open
Abstract
The mean survival of metastatic lung adenocarcinoma is less than 1 year, highlighting the urgent need to understand the mechanisms underlying its high mortality rate. The role of Extracellular vesicles (EVs) in facilitating the interactions between cancer cells and the metastatic microenvironment has garnered increasing attention. Previous studies on the role of EVs in metastasis have been primarily focused on cancer cell-derived EVs in modulating the functions of stromal cells. However, whether cancer stem cells (CSCs) can alter the metastatic properties of non-CSC cells, and whether EV crosstalk can mediate such interaction, have not been demonstrated prior to this report. In the present study, we integrated multi-omics sequencing and public database analysis with experimental validation to demonstrate, for the first time, the exosomal Mir100hg, derived from CSCs, could enhance the metastatic potential of non-CSCs both in vitro and in vivo. Mechanistically, HNRNPF and HNRNPA2B1 directly binds to Mir100hg, facilitating its trafficking via exosomes to non-CSCs. In non-CSCs, Mir100hg upregulates ALDOA expression, subsequently leading to elevated lactate production. Consequently, the increased lactate levels enhance H3K14 lactylation by 2.5-fold and promote the transcription of 169 metastasis-related genes. This cascade of events ultimately results in enhanced ALDOA-driven glycolysis and histone lactylation-mediated metastatic potential of non-CSC lung cancer cells. We have delineated a complex regulatory network utilized by CSCs to transfer their high metastatic activity to non-CSCs through exosomal Mir100hg, providing new mechanistic insights into the communication between these two heterogeneous tumor cell populations. These mechanistic insights provide novel therapeutic targets for metastatic lung cancer, including HNRNPF/HNRNPA2B1-mediated Mir100hg trafficking and the histone lactylation pathway, advancing our understanding of CSC-mediated metastasis while suggesting promising strategies for clinical intervention.
Collapse
Affiliation(s)
- Lei Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350000, Fujian Province, China
| | - Bowen Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jiyu Tan
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Ling Zhu
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Women and Children'S Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Sicheng Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhan Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Meng Xiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Li
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xue Han
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yao Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - H Rosie Xing
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| | - Jingyu Li
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Women and Children'S Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Jianyu Wang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
122
|
Moras B, Sissi C. Unravelling the Regulatory Roles of lncRNAs in Melanoma: From Mechanistic Insights to Target Selection. Int J Mol Sci 2025; 26:2126. [PMID: 40076754 PMCID: PMC11900516 DOI: 10.3390/ijms26052126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Melanoma is the deadliest form of skin cancer, and its treatment poses significant challenges due to its aggressive nature and resistance to conventional therapies. Long non-coding RNAs (lncRNAs) represent a new frontier in the search for suitable targets to control melanoma progression and invasiveness. Indeed, lncRNAs exploit a wide range of regulatory functions along chromatin remodeling, gene transcription, post-transcription, transduction, and post-transduction to ultimately tune multiple cellular processes. The understanding of this intricate and flexible regulatory network orchestrated by lncRNAs in pathological conditions can strategically support the rational identification of promising targets, ultimately speeding up the setup of new therapeutics to integrate the currently available approaches. Here, the most recent findings on lncRNAs involved in melanoma will be analyzed. In particular, the functional links between their mechanisms of action and some frequently underestimated features, like their different subcellular localizations, will be highlighted.
Collapse
Affiliation(s)
| | - Claudia Sissi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| |
Collapse
|
123
|
Hu W, Du X, Wang X, Zhang K, Li J, Gao Y, An T, Zhang H, Zhang Y, Ren Z, Xu Y, Liu S. Explore autophagy-related lncRNA-miRNA-mRNA ceRNA networks for diagnosis of early-onset schizophrenia through transcriptome analysis. Front Psychiatry 2025; 16:1567148. [PMID: 40078531 PMCID: PMC11897231 DOI: 10.3389/fpsyt.2025.1567148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Background The severe functional impairment and poor prognosis of early-onset schizophrenia (EOS) create a great need to identify effective biomarkers for early diagnosis in young psychiatric patients. Current research indicates a potential link between loss of autophagy function and emotional and behavioral abnormalities in individuals with psychiatric disorders. Materials and Methods This study aimed to explore diagnostic autophagy-related endogenous competitive RNA (ceRNA) networks for EOS patients. The messenger RNAs (mRNAs) and long non-coding RNAs (lncRNAs) expression profiles were obtained from peripheral blood mononuclear cells of 18 EOS patients and 12 healthy controls (HC). A co-expression analysis was performed between 365 core lncRNAs and 55 differentially expressed autophagy-related genes (ARGs) to identify differentially expressed autophagy-related lncRNAs. Subsequently, five diagnostic autophagy-related lncRNAs were identified as candidate genes to construct a ceRNA regulatory network using least absolute shrinkage and selection operator (LASSO) Cox regression, and receiver operating characteristic (ROC) curve analysis was performed to evaluate their predictive accuracy. Then, putative interactions among lncRNA-microRNAs (miRNAs)-mRNA were determined based on the lncRNASNP2 and TarBase databases. Results Three lncRNAs, twenty miRNAs, and ten mRNAs were selected to construct an autophagy-associated ceRNA network associated with EOS occurrence. Through protein-protein interaction network analysis, five hub mRNAs were identified, which exhibited good predictive ability in distinguishing EOS patients from healthy individuals. ROC curve analysis demonstrated that integrating three diagnostic lncRNAs (RP1-135L22.1, RP5-884C9.2, RP11-390F4.3) along with five hub mRNAs (EIF4G1, AKT1, BAX, WIPI2, MAPT) appeared to yield better diagnostic accuracy compared to using either lncRNAs or mRNAs alone. Furthermore, all three diagnostic lncRNAs and five hub mRNAs were positively correlated with at least two types of immune infiltration. Conclusion Through transcriptome analysis, we searched for diagnostic autophagy-related ceRNA networks, which provided valuable candidates for the early diagnosis of EOS.
Collapse
Affiliation(s)
- Wei Hu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Basic Medical College, Shanxi Medical University, Taiyuan, China
| | - Xinzhe Du
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinxia Wang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Kexin Zhang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Junxia Li
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yao Gao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ting An
- Shanxi Province Mental Health Center, Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Hong Zhang
- Shanxi Province Mental Health Center, Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Yu Zhang
- Basic Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhiyong Ren
- Shanxi Province Mental Health Center, Taiyuan Psychiatric Hospital, Taiyuan, China
| | - Yong Xu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Sha Liu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
124
|
He X, Li H. Role of LncRNA in Pathogenesis, Diagnosis and Treatment of Chronic Kidney Disease. Cell Biochem Biophys 2025:10.1007/s12013-025-01698-2. [PMID: 40000585 DOI: 10.1007/s12013-025-01698-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Chronic kidney disease (CKD) is a clinical syndrome of metabolic disorder caused by progressive kidney impairment for more than 3 months. CKD has become a global public health problem due to its high morbidity and mortality, which is difficult to be cured for most patients. The pathogenesis of CKD is still unclear, which is closely related to glomerulosclerosis, kidney tubular injury and kidney fibrosis. LncRNA is a non-coding RNA with a length of more than 200 nucleotides. It not only participates in intracellular transcriptional regulation, post-transcriptional regulation and epigenetic activities, but also forms a regulatory network together with miRNA and mRNA, to further conduct the reticular regulation in cells. Recently, it has been found that lncRNA participates in pathophysiological mechanism of CKD by regulating glomerulosclerosis, kidney tubular injury and kidney fibrosis. This has also become a new direction of lncRNA in early diagnosis and targeted therapy of CKD.
Collapse
Affiliation(s)
- Xin He
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Han Li
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
125
|
Wang Y, Yuan J, Guo K, Zhang Z, Zhu J, Arya S, Huang G, Li S, Chen Q, Liu X, Jia J. Identification and functional characterization of T-cell exhaustion-associated lncRNA AL031775.1 in osteosarcoma: a novel therapeutic target. Front Immunol 2025; 16:1517971. [PMID: 40066455 PMCID: PMC11891247 DOI: 10.3389/fimmu.2025.1517971] [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: 10/27/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Background Osteosarcoma, an aggressive bone malignancy predominantly affecting children and adolescents, presents significant therapeutic challenges with a 5-year survival rate below 30% in metastatic cases. T-cell exhaustion, characterized by the overexpression of immune checkpoint molecules, contributes to osteosarcoma progression and immune evasion. Although targeting these inhibitory pathways has shown potential in restoring T-cell activity, the molecular regulators of T-cell depletion in osteosarcoma are poorly understood. Methods This study employed comprehensive bioinformatics analyses on osteosarcoma samples from the TARGET database, combined with normal tissue data from the GTEx database, to identify T-cell exhaustion-associated genes and their co-expressed long non-coding RNAs (lncRNAs). Gene ontology and KEGG pathway analyses were used to elucidate immune-related pathway enrichments. A six-lncRNA prognostic model was established using LASSO regression and validated in separate cohorts. Functional assays evaluated the impact of the lncRNA AL031775.1 on osteosarcoma cell behavior and T-cell function. Results Twenty-four key T-cell exhaustion-related genes were identified and significantly enriched in immune-related pathways, indicating their importance in the osteosarcoma immune microenvironment. The constructed six-lncRNA model stratified patients by survival prognosis, showing robust predictive performance across cohorts. Among the six identified lncRNAs, AL031775.1 is notably downregulated in osteosarcoma patients and significantly promotes osteosarcoma cell proliferation, migration, and invasion while contributing to T-cell exhaustion. In T cells, downregulation of AL031775.1 impairs antitumor immunity, upregulates immune checkpoint molecules LAG3, PD1, and CTLA4, and diminishes T-cell cytotoxic activity against tumor cells. Conclusion This study identifies a novel six-lncRNA prognostic model and highlights the therapeutic potential of AL031775.1 in managing osteosarcoma by enhancing T-cell immunity and counteracting tumor progression. Targeting AL031775.1 represents a promising approach to improve immunotherapy efficacy in osteosarcoma. These findings provide critical insights into the molecular regulation of T-cell exhaustion and suggest a new avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Yameng Wang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jinghong Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Keying Guo
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Zhuoer Zhang
- Department of Bone and Soft Tissue Tumors, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Junchao Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Shahrzad Arya
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Guowen Huang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Shengqin Li
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Qi Chen
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xijuan Liu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jingyu Jia
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Institute of Orthopaedics of Jiangxi Province, Nanchang, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| |
Collapse
|
126
|
Shu A, Tian X, Yue J, Jiang Y, Liu Y. Unveiling the role of lncRNA ERDR1 in immune cell regulation. Heliyon 2025; 11:e42085. [PMID: 39991241 PMCID: PMC11847233 DOI: 10.1016/j.heliyon.2025.e42085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of RNA molecules that exceed 200 nucleotides in length and lack the capacity to encode proteins. In recent years, there has been a surge of interest in lncRNA research, leading to the discovery of their diverse structures and functions. This review focused on elucidating the regulatory roles of lncRNA erythroid differentiation regulatory 1 (Erdr1) within immune cells and its involvement in related disorders. By synthesizing findings from recent studies sourced from PubMed, this paper examined the biological functions and underlying mechanisms by which lncRNA Erdr1 influences immune cells and contributes to various diseases. Emerging research highlights that lncRNA Erdr1 exerts significant effects on the functionality of immune cells, particularly T lymphocytes (T cells), natural killer (NK) cells, and macrophages. Furthermore, Erdr1 has been implicated in the mitigation of several diseases, including acne, wound healing, osteoarthritis, melanoma, gastric cancer, obesity, and autism. Given its complex biological functions and mechanisms, Erdr1 presents itself as a promising biomarker and a potential therapeutic target for a range of immune cell-related disorders.
Collapse
Affiliation(s)
- Aihua Shu
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Xu Tian
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Jie Yue
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Yuxia Jiang
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Yifei Liu
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| |
Collapse
|
127
|
Li Y, Xu Y, Yu X, Su S, Wu B, Su Y, Guo L. Long non-coding RNA NEAT1 promotes colorectal cancer progression via interacting with SIRT1. Sci Rep 2025; 15:5673. [PMID: 39955367 PMCID: PMC11830096 DOI: 10.1038/s41598-025-90416-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Nuclear-enriched abundant transcript 1 (NEAT1), a long noncoding RNA, is found to be significantly dysregulated in different types of cancer, including colorectal cancer (CRC). Nevertheless, there is still much to learn about the precise functions and processes of NEAT1 in the progression of CRC. Using The Cancer Genome Atlas (TCGA) database and 50 CRC specimens from the First Affiliated Hospital of Dali University, we assessed the expression of NEAT1 to determine its clinical impact. Through gene set enrichment analysis (GSEA), Cancer Single-cell State Atlas (CancerSEA), and immune infiltration studies, we elucidated key functions of NEAT1. We utilized Cell Counting Kit-8 (CCK8), wound healing, and Transwell assays to investigate the role of NEAT1 in the progression of CRC. Through the use of GSEA and immunohistochemistry, additional investigations were conducted to unveil the downstream targets of NEAT1 and gain insights into their regulatory dynamics. Our in vitro studies confirmed the regulatory role of NEAT1 in CRC. Findings indicate that increased NEAT1 expression correlates with adverse outcomes in colorectal tissues. In the CRC model, reduced levels of NEAT1 lead to reduced cell proliferation, invasion, and migration. Additionally, NEAT1 influenced immune cell infiltration in CRC and functioned as an oncogene by upregulating Sirtuin 1 (SIRT1) expression. This study demonstrates that NEAT1 promotes CRC progression and metastasis through a SIRT1-mediated mechanism, suggesting its potential as a prognostic biomarker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Yuwei Li
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Yunchun Xu
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Xinya Yu
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Shuangyan Su
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Bihua Wu
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Yunpeng Su
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China
| | - Le Guo
- Department of Medical Microbiology and Immunology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, P.R. China.
| |
Collapse
|
128
|
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
|
129
|
Long L, Zhang C, He Z, Liu O, Yang H, Fan Z. LncRNA NR_045147 modulates osteogenic differentiation and migration in PDLSCs via ITGB3BP degradation and mitochondrial dysfunction. Stem Cells Transl Med 2025; 14:szae088. [PMID: 39674578 PMCID: PMC11878762 DOI: 10.1093/stcltm/szae088] [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: 06/27/2024] [Accepted: 10/16/2024] [Indexed: 12/16/2024] Open
Abstract
Periodontitis is an inflammation of the alveolar bone and soft tissue surrounding the teeth. Although mesenchymal stem cells (MSCs) have been implicated in periodontal regeneration, the mechanisms by which they promote osteogenesis remain unclear. We examined whether epigenetic modifications mediated by the long-noncoding RNA (lncRNA) NR_045147, which plays a crucial role in cancer, influence the osteogenic differentiation of periodontal ligament stem cells (PDLSCs). Alkaline phosphatase staining, alizarin red staining, and western blotting were used to detect the effects of NR_045147 on PDLSC osteogenic differentiation. Scratch migration and transwell chemotaxis assays were used to evaluate the effects of NR_045147 on PDLSC migration. Mitochondrial function was evaluated via Seahorse XF analysis to measure changes in cellular respiration upon manipulation of NR_045147 expression. Ubiquitination assays were performed to examine the protein stability and degradation pathways affected by the NR_045147-MDM2 interaction. An in vivo nude rat calvarial defect model was established and gene-edited PDLSCs were re-implanted to examine the osteogenic effects of NR_045147. NR_045147 significantly reduced PDLSC osteogenic differentiation and migration ability both in vitro and in vivo. Under inflammatory conditions, the loss of NR_045147 rescued osteogenesis. NR_045147 significantly blocked the expression of integrin beta3-binding protein (ITGB3BP). Mechanistically, NR_045147 promoted the ITGB3BP-MDM2 interaction, thus increasing ITGB3BP ubiquitination and degradation. NR_045147 regulated PDLSC mitochondrial respiration and ITGB3BP upregulation efficiently promoted their osteogenic differentiation and migration ability. Concluding, NR_045147 downregulation enhances PDLSC osteogenic differentiation and migration, connects changes in cellular metabolism to functional outcomes via mitochondrial respiration, and promotes ITGB3BP degradation by mediating its interaction with MDM2.
Collapse
Affiliation(s)
- Lujue Long
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, People’s Republic of China
| | - Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, People’s Republic of China
| | - Zhengquan He
- Department of Orthodontics, Changsha Stomatology Hospital, Changsha, Hunan, People’s Republic of China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-Maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, People’s Republic of China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, People’s Republic of China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, People’s Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People’s Republic of China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
130
|
Long L, Zhang C, He Z, Liu O, Yang H, Fan Z. LncRNA NR_045147 modulates osteogenic differentiation and migration in PDLSCs via ITGB3BP degradation and mitochondrial dysfunction. Stem Cells Transl Med 2025; 14. [DOI: 2.doi: 10.1093/stcltm/szae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025] Open
Abstract
Abstract
Periodontitis is an inflammation of the alveolar bone and soft tissue surrounding the teeth. Although mesenchymal stem cells (MSCs) have been implicated in periodontal regeneration, the mechanisms by which they promote osteogenesis remain unclear. We examined whether epigenetic modifications mediated by the long-noncoding RNA (lncRNA) NR_045147, which plays a crucial role in cancer, influence the osteogenic differentiation of periodontal ligament stem cells (PDLSCs). Alkaline phosphatase staining, alizarin red staining, and western blotting were used to detect the effects of NR_045147 on PDLSC osteogenic differentiation. Scratch migration and transwell chemotaxis assays were used to evaluate the effects of NR_045147 on PDLSC migration. Mitochondrial function was evaluated via Seahorse XF analysis to measure changes in cellular respiration upon manipulation of NR_045147 expression. Ubiquitination assays were performed to examine the protein stability and degradation pathways affected by the NR_045147–MDM2 interaction. An in vivo nude rat calvarial defect model was established and gene-edited PDLSCs were re-implanted to examine the osteogenic effects of NR_045147. NR_045147 significantly reduced PDLSC osteogenic differentiation and migration ability both in vitro and in vivo. Under inflammatory conditions, the loss of NR_045147 rescued osteogenesis. NR_045147 significantly blocked the expression of integrin beta3-binding protein (ITGB3BP). Mechanistically, NR_045147 promoted the ITGB3BP-MDM2 interaction, thus increasing ITGB3BP ubiquitination and degradation. NR_045147 regulated PDLSC mitochondrial respiration and ITGB3BP upregulation efficiently promoted their osteogenic differentiation and migration ability. Concluding, NR_045147 downregulation enhances PDLSC osteogenic differentiation and migration, connects changes in cellular metabolism to functional outcomes via mitochondrial respiration, and promotes ITGB3BP degradation by mediating its interaction with MDM2.
Collapse
Affiliation(s)
- Lujue Long
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing ,
| | - Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing ,
| | - Zhengquan He
- Department of Orthodontics, Changsha Stomatology Hospital , Changsha, Hunan ,
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-Maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University , Changsha, Hunan ,
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing ,
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing ,
- Beijing Laboratory of Oral Health, Capital Medical University , Beijing ,
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences , Beijing ,
| |
Collapse
|
131
|
Chen Y, Shi H, Dong Y, Cui W. LncRNA MSTO2P affects the proliferation, invasion and migration of non-small cell lung cancer by regulating the Wnt/β-catenin pathway. Discov Oncol 2025; 16:150. [PMID: 39928213 PMCID: PMC11811353 DOI: 10.1007/s12672-025-01920-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
This study aimed to investigate the regulatory effects of the long non-coding RNA (lncRNA) MISATO family member 2 (MSTO2P) on non-small cell lung cancer (NSCLC) cell viability, invasion, and migration, as well as the underlying mechanism. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze the expression levels of MSTO2P. The effects of MSTO2P on cell viability, invasion, and migration were assessed using cell counting kit-8 (CCK-8), Transwell invasion, and wound healing assays in A549 and H1229 cells. A human phospho-kinase array kit was employed to identify potential phosphorylated kinases or signaling nodes affected by MSTO2P. The interaction between MSTO2P and β-catenin was evaluated using RNA pull-down and RNA immunoprecipitation (RIP) assays. A xenograft tumor mouse model was established to evaluate tumor growth. The results demonstrated that MSTO2P expression was elevated in NSCLC tissues and cells compared to normal counterparts. Silencing MSTO2P inhibited the viability, invasion, and migration of A549 and H1229 cells. MSTO2P interacted with β-catenin, thereby activating the Wnt/β-catenin pathway. Overexpression of MSTO2P or β-catenin promoted the viability, invasion, and migration of A549 and H1229 cells, effects that were reversed by treatment with XAV-939. In vivo studies showed that silencing MSTO2P suppressed tumor growth. In conclusion, MSTO2P promoted NSCLC cell viability, invasion, and migration by regulating the Wnt/β-catenin pathway, suggesting that MSTO2P may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yunfeng Chen
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Hai Shi
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Yuan Dong
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China.
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| | - Wenjie Cui
- Department of Respiratory and Critial Care Medicine, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou City, 221116, Jiangsu Province, China.
- Department of Respiratory and Critial Care Medicine, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| |
Collapse
|
132
|
Wu Y, Wang D, Zhao J, Guo J, Gao Z, Xu Q, Hu X. NRAV promoted the malignant progression of gastric cancer. Gene 2025; 937:149134. [PMID: 39615808 DOI: 10.1016/j.gene.2024.149134] [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: 06/04/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
Gastric cancer (GC) has been ranked as the third incidence tumors globally. Long non-coding RNA (lncRNA) NRAV has been reported as a tumor-enhancer in the development of human cancers, whereas the function of NRAV in GC remains to be elucidated. The aim of this research was to explore the underlying function of NRAV in GC. Through comprehensive bioinformatics analysis, a significantly elevation of NRAV was found in both human GC tissues and cell lines, which indicated the poor prognosis of GC patients. Then, we conducted a series of functional experiments to illustrate the role of NRAV in GC. The results showed that the down-regulation of NRAV exhibited a significant inhibitory effect on GC cell proliferation and migration, while NRAV overexpression promoted GC cell proliferation and migration. Through xenograft mouse tumor model, the suppression of NRAV led to a reduction in the growth of tumor mice, whereas overexpression of NRAV notably enhanced tumor growth. Finally, EFHC1 was revealed as the potential target gene of NRAV. Overall, our findings indicated the promising application of NRAV as a therapeutic target and prognostic biomarker for GC.
Collapse
Affiliation(s)
- Yuchen Wu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Bengbu Medical University, Bengbu 233030, China
| | | | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Zhenyu Gao
- Wenzhou Medical University, Wenzhou 325035, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiaoge Hu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
133
|
Luo Y, Ye Z, Li Y, Wong CW, Xu S, Deng Y, Su Z, Li X, Huang Y, Han B. Analysis of long noncoding gene expression and its interactions with protein-coding genes in vascular endothelial cells in keloids. Eur J Med Res 2025; 30:86. [PMID: 39920823 PMCID: PMC11806810 DOI: 10.1186/s40001-025-02271-6] [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: 10/09/2023] [Accepted: 01/01/2025] [Indexed: 02/09/2025] Open
Abstract
OBJECTIVES The purpose of this study was to determine the relationship between protein-coding RNA (messenger RNA, mRNA) and long noncoding RNA (lncRNA) expressed in vascular endothelial cells (VECs) in keloids by reanalyzing Gene Expression Omnibus (GEO) microarray chip data. MATERIALS AND METHODS The GSE121618 database and clinical information of these samples were downloaded and reanalyzed by the R language package. Expression differences in mRNA and lncRNA between keloids and normal skin were calculated. GO/KEGG enrichment analysis was conducted to determine the function of these genes, and an interaction network of lncRNAs-mRNAs was constructed. Magnetic Sorting of VECs and qRT-PCR were used to verify these bioinformatic results. RESULTS The expression of three hundred and five mRNAs in the keloid group was significantly different from that in the normal group, and 98 lncRNAs were different, 21 of which were upregulated and 118 of which were downregulated. The hub relationship between the upregulated lncRNA‒mRNA interaction was lncRNA LINC01546-RASAL3/COL13A1, while the downregulated hub was lncRNA LOC101929787-PRKAA2/KRT71/SSTR1. qPCR verification result showed no obvious statistical differences. CONCLUSIONS Through the in-depth mining of keloid microarray data using bioinformatic methods, we speculated that VECs can affect the development and progression of keloids by epigenomic regulation via lncRNA‒mRNA interactions.
Collapse
Affiliation(s)
- Yunxiang Luo
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zi Ye
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yi Li
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Chau Wei Wong
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
- LIHE Hospital, 1428 GuangZhou Avenue, Guangzhou, 510060, China.
| | - Shuqia Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yu Deng
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhicheng Su
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Xueqing Li
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yingxiong Huang
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Bing Han
- Scar and Wound Treatment Department, Plastic Surgery Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, 100144, China.
| |
Collapse
|
134
|
Gu X, Hu X, Zhang S, Zhang X, Wang Y, Li L. The diagnostic and prognostic significance of HOXC13-AS and its molecular regulatory mechanism in human cancer. Front Mol Biosci 2025; 12:1540048. [PMID: 39981436 PMCID: PMC11839424 DOI: 10.3389/fmolb.2025.1540048] [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: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
HOXC13 antisense RNA (HOXC13-AS, also known as HOXC-AS5) is a long non-coding RNA that is expressed abnormally in various types of tumors and is closely related to clinical staging, clinical pathological features, and patient survival. HOXC13-AS is involved in the occurrence and development of tumors, affecting cell proliferation, migration, invasion, epithelial-mesenchymal transition, and tumor growth. This review summarizes the clinical significance of HOXC13-AS as a biomarker for human tumor diagnosis and prognosis and outlines the function and molecular regulation mechanism of HOXC13-AS in various types of cancer, including nasopharyngeal carcinoma, breast cancer, oral squamous cell carcinoma, glioma, and cervical cancer. Overall, this review emphasizes the potential of HOXC13-AS as a human tumor predictive biomarker and therapeutic target, paving the way for its clinical application.
Collapse
Affiliation(s)
- Xiaosi Gu
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, China
| | - Xin Hu
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, China
| | - Sijia Zhang
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoyu Zhang
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, China
| | - Yong Wang
- Shandong Provincial Engineering Research Center for Bacterial Oncolysis and Cell Treatment, Jinan, Shandong, China
| | - Lianlian Li
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, China
- Laboratory of Metabolism and Gastrointestinal Tumor, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
135
|
Lu Q, Ling H, Lao Y, Liu J, Su W, Huang Z. Immune-mediated mechanisms in acute osteofascial compartment syndrome: insights from multi-omics analysis. Eur J Med Res 2025; 30:79. [PMID: 39910406 PMCID: PMC11796005 DOI: 10.1186/s40001-025-02285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Acute Osteofascial Compartment Syndrome (AOCS) stands as a critical surgical emergency, often secondary to various diseases. Its clinical manifestation arises from increased pressure within the fascial compartment, resulting in diminished tissue perfusion and consequential ischemic damage. Presently, clinical diagnostics lack effective biological markers, and patients face a grim prognosis, experiencing muscle contractures, necrosis, amputations, renal failure, and even mortality. The primary treatment, fasciotomy, poses infection risks and potential nerve damage. Hence, there is an urgent need for research elucidating AOCS's pathogenic mechanism and exploring novel treatments. METHODS To address this, we established a rat model of AOCS, extracting toe flexor muscles from both experimental and control groups. Employing second-generation high-throughput sequencing, we obtained comprehensive mRNA, lncRNA, circRNA, and miRNA data. Comparative analysis of expression differences between AOCS and control groups, followed by in-depth examination, allowed us to unravel the intricacies of AOCS occurrence from a multi-omics perspective. RESULTS Our research findings indicate that AOCS is an immune-mediated inflammatory disease, primarily involving immune cells, especially neutrophils. In addition, genes associated with ferroptosis, a form of regulated cell death, are found to be upregulated in the rat model, with non-coding RNAs playing a role in regulatory interactions. CONCLUSIONS These results suggest that neutrophils may undergo ferroptosis, thereby enhancing inflammation and immune responses in the fascial compartment, which promotes disease progression. Furthermore, these findings reveal the interactions between immune molecules and pathways in AOCS, which are significant for a deeper understanding of the pathogenesis of the disease and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Qinzhen Lu
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China
| | - He Ling
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China
| | - Yonghui Lao
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China
| | - Junjie Liu
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China
| | - Wei Su
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China
| | - Zhao Huang
- Department of Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 Shuang Yong Road, Nanning, 530022, Guangxi, China.
| |
Collapse
|
136
|
Zhao SS, Tao DL, Chen JM, Zhang MY, Yang X, Song JK, Liu Q, Zhao GH. Neospora caninum infection specifically suppresses the expression of a host lncRNA XR_001919077.1 to facilitate parasite propagation by modulating host cell mitochondrial function and autophagy. Microbiol Spectr 2025; 13:e0158024. [PMID: 39714178 PMCID: PMC11792476 DOI: 10.1128/spectrum.01580-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Neospora caninum is one of the most common pathogens causing reproductive failure in ruminants (e.g., cattle and goats) worldwide. However, due to a poor understanding of the pathogenic mechanisms of N. caninum infection, no effective drugs and vaccines are currently available. Long non-coding RNAs (lncRNAs) have been reported to be important regulators involved in a great number of physiological and pathological processes. Our previous study found that N. caninum infection induced significantly aberrant expression of lncRNA profiles in caprine endometrial epithelial cells (EECs). In the present study, we found that N. caninum infection specifically suppressed the expression of a novel lncRNA, XR_001919077.1, and knockdown of XR_001919077.1 with small interfering RNA significantly promoted the propagation of N. caninum in caprine EECs. Rapid amplification of cDNA ends analysis generated six splice variants of XR_001919077.1, with lengths ranging from 592 to 694 nt. Transfection of the full length of each variant markedly inhibited the propagation of N. caninum in caprine EECs. Further study suggested that XR_001919077.1 acted as a sponge of Chi-miR-93-5p to promote the expression of sirt1, and the XR_001919077.1/Chi-miR-93-5p/sirt1 axis significantly delayed the in vitro growth of N. caninum in caprine EECs by regulating host cell mitochondrial function and autophagy. Our findings provide a novel insight to understand the interactions between N. caninum and host cells.IMPORTANCEThe uterus is an indispensable reproductive organ for embryo implantation and fetal growth. The endometrium is more vulnerable to infection by pathogenic microorganisms resulting in an increased risk of miscarriage. Neospora caninum is one of the most common pathogens causing miscarriage in ruminants and is able to naturally inhabit the uterus, with N. caninum tissue cysts found in the endometrium. Recent advances in N. caninum research have revealed aberrant expression of long non-coding RNA (lncRNA) profiles in infected caprine endometrial epithelial cells. In the present study, N. caninum, but not Toxoplasma gondii, which has similar morphological and biological features to N. caninum, specifically suppresses the expression of a host lncRNA, XR_ 001919077.1, to impair host's defense through the competitive endogenous RNA mechanism to modulate the host cell mitochondrial function and autophagy to facilitate parasite propagation. The findings suggest a novel immune evasion strategy of N. caninum to facilitate intracellular propagation and provide an alternative path to develop control strategies against neosporosis.
Collapse
Affiliation(s)
- Shan-Shan Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - De-Liang Tao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jin-Ming Chen
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ming-Yi Zhang
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xin Yang
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jun-Ke Song
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qun Liu
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Guang-Hui Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
137
|
Saadh MJ, Menon SV, Verma R, Siva Prasad GV, Allela OQB, Mahdi MS, Ahmad N, Husseen B. LncRNA CRNDE and HOTAIR: Molecules behind the scenes in the progression of gastrointestinal cancers through regulating microRNAs. Pathol Res Pract 2025; 266:155778. [PMID: 39721094 DOI: 10.1016/j.prp.2024.155778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Gastrointestinal (GI) cancers, such as gastric cancer, hepatocellular carcinoma, colorectal cancer, and esophageal cancer, pose a significant medical and economic burden globally, accounting for the majority of new cancer cases and deaths each year. A lack of knowledge about the molecular mechanisms of GI cancers is reflected in the low efficacy of treatment for individuals with late stage and recurring illness. Understanding the molecular pathways that promote the growth of GI cancers may open doors for their therapy. Numerous long non-coding RNAs (lncRNAs) that are produced differently in normal and malignant tissues have been discovered by genome-wide techniques. The role of lncRNAs in the diagnosis, proliferation, metastasis, and drug resistance of different GI cancers has been investigated in recent research. LncRNAs may affect transcription, epigenetic modifications, protein/RNA stability, translation, and post-translational modifications via their interactions with DNA, RNAs, and proteins. Also, by functioning as competing endogenous RNAs (ceRNAs), they control the synthesis of certain microRNAs (miRNAs), which in turn modify the downstream target molecules of these miRNAs. Based on recent studies, lncRNAs in particular, CRNDE and HOTAIR, sponge different miRNAs and their downstream genes, which in turn regulate GI cancers development, including cell proliferation, invasion, migration, and chemoresistance. In this comprehensive review, we present an overview of the biological roles of CRNDE and HOTAIR and their associated mechanisms, miRNAs/mRNA pathways, in various GI cancers, encompassing colorectal cancer, hepatocellular carcinoma, esophageal cancer, and gastric cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Sahibzada Ajit Singh Nagar, Jhanjeri, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | | | | | - Nabeel Ahmad
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India; Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand- 831001, India.
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
138
|
Tang L, Wang Y, Chen Y, Xu B, Miao L, Zhong L. LncRNA MIR17HG drives cisplatin resistance partially via miR-138-5p/AKAP9 axis in cholangiocarcinoma. Scand J Gastroenterol 2025; 60:184-196. [PMID: 39773276 DOI: 10.1080/00365521.2025.2450024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
OBJECTIVES This study aims to discover the role of lncRNA MIR17HG, referred to as MIR17HG, in cisplatin resistance for cholangiocarcinoma (CCA). METHODS QRT-PCR was conducted to measure the expression of MIR17HG in cisplatin-resistant/sensitive CCA cells and clinical CCA specimens. Log-rank test was used to analyze the survival curve. Cck8-assay and flow cytometry were employed to detect the sensitivity of CCA cells to cisplatin and the apoptosis rate following different treatments, respectively. The next-generation sequencing was carried out to get gene transcripts after silencing MIR17HG in HCCC-9810 cells. The LncBase database was used to predict the target miRNA of MIR17HG, and MS2 RIP assay and dual luciferase assay were conducted to confirm their binding. MiRwalk database and the RNA sequencing data were utilized to screen the key genes regulated by MIR17HG/miR-138-5p axis and a dual luciferase assay was performed to confirm the binding site of miR-138-5p with AKAP9. Immunoblotting was further employed to give assistant evidence. Rescue experiments were performed to observe the function of miR-138-5p and AKAP9 in MIR17HG-induced cisplatin resistance. RESULTS MIR17HG overexpression predicts cisplatin resistance and poor prognosis in CCA. MIR17HG could bind with miR-138-5p to release AKAP9, thereby inhibiting cisplatin-induced apoptosis and promoting cisplatin resistance in CCA. MIR17HG silencing in CCA cells leads to expression alteration of genes, which are enriched in platinum resistance-related pathways. CONCLUSIONS LncRNA MIR17HG regulates platinum resistance-associated genes and promotes cisplatin resistance partially via the miR-138-5p/AKAP9 axis by inhibiting cisplatin-induced apoptosis in CCA.
Collapse
Affiliation(s)
- Lingyu Tang
- Department of Gastroenterology and Endoscopy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuting Wang
- Department of Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongzhen Chen
- Department of general practice, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Boming Xu
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Lin Miao
- Department of Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Zhong
- Department of Gastroenterology and Endoscopy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
139
|
Chen X, Zhang Y, Hu N, Pan Q, Wang K, Yin Y. Regulatory mechanism of LncRNA GAS5 in cognitive dysfunction induced by sevoflurane anesthesia in neonatal rats. Brain Dev 2025; 47:104295. [PMID: 39550980 DOI: 10.1016/j.braindev.2024.10.003] [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: 05/09/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND AND OBJECTIVES Sevoflurane (Sev) exposure may provoke deleterious effects on cognitive function. This study explores the mechanism of long non-coding RNA growth arrest specific transcript 5 (LncRNA GAS5) in Sev-induced cognitive dysfunction in neonatal rats. METHODS Cognitive dysfunction was induced by Sev anesthesia in 7-day-old Sprague-Dawley rats, followed by open field test, novel object recognition, radial arm maze, and Morris water maze to evaluate cognitive function of rats. The subcellular localization of LncRNA GAS5 was detected by nucleocytoplasmic isolation assay, and the binding of miR-137 to LncRNA GAS5 and NKCC1 was detected by RNA pull down and dual-luciferase reporter assay, respectively. Adenovirus-packaged sh-LncRNA GAS5 was injected into the hippocampus of Sev rats. qRT-PCR and Western blot were performed to detect the expressions of LncRNA GAS5, miR-137 and NKCC1 in the hippocampus of rats. RESULTS Sev anesthesia led to cognitive dysfunction in neonatal rats. LncRNA GAS5 was highly expressed in Sev rats, and inhibition of LncRNA GAS5 alleviated Sev-induced cognitive dysfunction in rats. LncRNA GAS5 targeted miR-137, and miR-137 inhibited NKCC1 expression. Knockdown of miR-137 or overexpression of NKCC1 reversed the effect of LncRNA GAS5 inhibition on cognitive dysfunction in sev rats. CONCLUSION LncRNA GAS5 promotes Sev-induced cognitive dysfunction in neonatal rats via the miR-137/NKCC1 axis.
Collapse
Affiliation(s)
- Xi Chen
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yu Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Nan Hu
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Qian Pan
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Kaiyuan Wang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
140
|
Xiao H, Fei M, Xu Q, Gao Y, Feng R, Liang C, Wang B, Li H. MATN1-AS1 Promotes Tumour Metastasis and Sunitinib Resistance via E2F2 in Clear Cell Renal Cell Carcinoma. J Cell Mol Med 2025; 29:e70428. [PMID: 39999286 PMCID: PMC11855375 DOI: 10.1111/jcmm.70428] [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: 08/22/2024] [Revised: 12/14/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
It has become increasingly recognised that MATN1-AS1 is involved in multiple tumour development. The role of MATN1-AS1 in clear cell renal cell carcinoma (ccRCC), however, is still largely unrecognised. This study investigated the molecular functions of MATN1-AS1 in promoting ccRCC metastasis and sunitinib resistance. MATN1-AS1 was found to be mainly located in the cytoplasm and was upregulated in ccRCC, and a positive association was seen between greater levels of MATN1-AS1 expression and worse clinical outcomes. Downregulating MATN1-AS1 significantly hindered cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT). MATN1-AS1 promoted tumour growth and metastasis in vivo. Mechanismly, MATN1-AS1 targeted microRNA miR-214-5p, thereby upregulating E2F2 and promoting E2F2-mediated EMT. We discovered that MATN1-AS1 also promoted sunitinib resistance via E2F2 in vitro. Collectively, our research uncovered the protumor characteristics of MATN1-AS1 and suggested it as a therapeutic target for reverse sunitinib resistance in ccRCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Epithelial-Mesenchymal Transition/genetics
- Epithelial-Mesenchymal Transition/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Line, Tumor
- Animals
- Cell Movement/genetics
- Cell Movement/drug effects
- Cell Proliferation/genetics
- Cell Proliferation/drug effects
- E2F2 Transcription Factor/genetics
- E2F2 Transcription Factor/metabolism
- Mice
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Neoplasm Metastasis
- Male
- Female
- MicroRNAs/genetics
- Mice, Nude
Collapse
Affiliation(s)
- Haibing Xiao
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Mintian Fei
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Qili Xu
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Yu Gao
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Rui Feng
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
- Department of UrologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chaozhao Liang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Baojun Wang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Haolin Li
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
- Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
141
|
Hassan FU, Safdar M, Younus M, Arain MA. Regulation of energy metabolism by non-coding RNAs in livestock species: a review. J Comp Physiol B 2025; 195:1-12. [PMID: 39638953 DOI: 10.1007/s00360-024-01596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
The optimisation of livestock production relies on efficient energy metabolism. This review focused on elaborate regulatory processes governed by non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). It explores the complex energy metabolism processes in livestock, elucidating the functions of ncRNAs in the expression of genes and pathways. miRNAs have been identified as significant regulators of glycolysis and glucose metabolism, whereas lncRNAs are known to affect adipogenesis and mitochondrial activity. Moreover, circRNAs have a substantial influence on the regulation of energy. In addition, this is not only enriching non-coding RNA-mediated energy control but also sheds light on possible applications. It is derived from its ability to condense complex molecular systems, thereby offering crucial insights to researchers. Through a comprehensive analysis of the intricate relationship between ncRNAs and energy metabolism, the information of this review provides a valuable framework for the implementation of focused interventions that hold the potential to significantly enhance the efficiency of livestock production.
Collapse
Affiliation(s)
- Faiz-Ul Hassan
- Department of Breeding and Genetics, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan.
| | - Muhammad Safdar
- Department of Breeding and Genetics, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan
| | - Muhammad Younus
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan
| | - Muhammad Asif Arain
- Faculty of Veterinary and Animal Sciences, Water and Marine Sciences, Lasbela University of Agriculture, Uthal, 90150, Balochistan, Pakistan
| |
Collapse
|
142
|
Wang Y, Zhou Y, Li K. The role of lncRNA in the differentiation of adipose-derived stem cells: from functions to mechanism. J Mol Med (Berl) 2025; 103:125-135. [PMID: 39708157 DOI: 10.1007/s00109-024-02507-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Adipose-derived stem cells (ADSCs) have become one of the best seed cells widely studied and concerned in tissue engineering because of their rich sources and excellent multi-directional differentiation ability, which are expected to play a practical application role in tissue defect, osteoporosis, plastic surgery, and other fields. However, the differentiation direction of ADSCs is regulated by complex factors. Long non-coding RNAs (lncRNAs) are RNA molecules longer than 500 nucleotides that do not encode proteins and can act as signaling RNAs in response to intracellular and extracellular stimuli. Recently, accumulating evidence has revealed that lncRNAs could regulate the cell cycle and differentiation direction of ADSCs through various mechanisms, including histone modification, binding to RNA-binding proteins, and regulating the expression of miRNAs. Therefore, enriching and elucidating its mechanism of action as well as targeting lncRNAs to regulate ADSCs differentiation have potential prospects in tissue regeneration applications such as bone, blood vessels, and adipose. In this review, we summarize the role and mechanism of lncRNAs and its complexes in the multi-directional differentiation of ADSCs and discuss some potential approaches that can exert therapeutic effects on tissue defects by modulating the expression level of lncRNAs in ADSCs. Our work might provide some new research directions for the clinical applications of tissue engineering.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China
| | - Yuxi Zhou
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China
| | - Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Central South University, Changsha, 410000, China.
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
143
|
Li Z, Wang D, Zhu X. Unveiling the functions of five recently characterized lncRNAs in cancer progression. Clin Transl Oncol 2025; 27:458-465. [PMID: 39066874 DOI: 10.1007/s12094-024-03619-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: 04/29/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Numerous studies over the past few decades have shown that RNAs are multifaceted, multifunctional regulators of most cellular processes, contrary to the initial belief that they only act as mediators for translating DNA into proteins. LncRNAs, which refer to transcripts longer than 200nt and lack the ability to code for proteins, have recently been identified as central regulators of a variety of biochemical and cellular processes, particularly cancer. When they are abnormally expressed, they are closely associated with tumor occurrence, metastasis, and tumor staging. Therefore, through searches on Google Scholar, PubMed, and CNKI, we identified five five recently characterized lncRNAs-Lnc-SLC2A12-10:1, LncRNA BCRT1, lncRNA IGFBP4-1, LncRNA PCNAP1, and LncRNA CDC6-that have been linked to the promotion of cancer cell proliferation, invasion, and metastasis. Consequently, this review encapsulates the existing research and molecular underpinnings of these five newly identified lncRNAs across various types of cancer. It suggests that these novel lncRNAs hold potential as independent biomarkers for clinical diagnosis and prognosis, as well as candidates for therapeutic intervention. In parallel, we discuss the challenges inherent in the research on these five newly discovered lncRNAs and look forward to the avenues for future exploration in this field.
Collapse
Affiliation(s)
- Zhicheng Li
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Dan Wang
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Xiaojun Zhu
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China.
| |
Collapse
|
144
|
Ding Z, Wu B, Yang J, Wang D, Qiao J, Guo F. Glycolysis regulated exosomal LINC01214 inhibited CD8 + T cell function and induced anti-PD1 resistance in melanoma via modulating miR-4492/PPP1R11 axis. Noncoding RNA Res 2025; 10:242-251. [PMID: 39559293 PMCID: PMC11570817 DOI: 10.1016/j.ncrna.2024.10.005] [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: 08/29/2024] [Revised: 10/16/2024] [Accepted: 10/27/2024] [Indexed: 11/20/2024] Open
Abstract
Background Long non-coding RNAs (lncRNAs) can be incorporated into exosomes to mediate the intercellular communication, regulating the occurrence, development, and immunosuppression of cancers. T cell dysfunction has been a hallmark of many cancers, including melanoma, which enables cancer cells escape from host immune surveillance. However, the molecular mechanism of exosome-transmitted lncRNAs in CD8+ T cell dysfunction in melanoma remains largely unclear. Method The expression of circulating LINC01214 (cirLINC01214) was detected by quantitative real-time polymerase chain reaction (RT-qPCR). Exosomes were isolation from the culture medium and plasma of melanoma patients via ultracentrifugation and characterized by transmission electronic microscopy. The regulation of exosomal LINC01214 on CD8+ T cell function was determined by ELISA. The molecular mechanism of exosomal LINC01214 in CD8+ T cells were assessed by the RNA immunoprecipitation and pull-down assay. A mouse model with reconstituted human immune system was used to explore the role of exosomal LINC01214 in the resistance to anti-PD1 therapy. Results LINC01214 was highly expressed in melanoma tissues compared with matched adjacent normal tissues. Increased levels of circulating LINC01214 (cirLINC01214) was observed in melanoma patient plasma and correlated with poor PD-1 immunotherapy response. The cirLINC01214 was predominantly released by melanoma cells in an exosome manner. Melanoma cell-derived exosomal LINC01214 inhibits the production of IFN-γ, TNF-α, Granzyme-B and Perforin by CD8+ T cells. Further mechanism study found that cirLINC01214 delivered by exosomes suppressed CD8+ T cell function by up-regulating the expression of Protein Phosphatase 1 Regulatory Inhibitor Subunit 11 (PPP1R11) through sponging miR-4492. CirLINC01214 conferred resistance to PD-1 immunotherapy in melanoma xenograft mouse model. Melanoma patients with poor prognosis after PD-1 treatment carried high levels of exosomal LINC01214. Additionally, the secretion of exosomal cirLINC01214 was enhanced by the Warburg effect, which was consistent with the reprogrammed glucose metabolism of melanoma. Conclusions Our results demonstrated that exosomal LINC01214 released by melanoma cells promoted immunotherapy resistance by inducing CD8+ T cell dysfunction via the miR-4492/PPP1R11 regulatory loop. Targeting cirLINC01214 might be a potential therapeutic strategy to enhance the outcome of immunotherapy in melanoma.
Collapse
Affiliation(s)
- Zhi Ding
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Baojin Wu
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Junyi Yang
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Daohe Wang
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Qiao
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fanli Guo
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
145
|
Fan Y, Tian D, Lv Z, Peng S, Zhu S. LncRNA-THBS4 affects granulosa cell proliferation and apoptosis in diminished ovarian reserve by regulating PI3K/AKT/mTOR signaling pathway. J Reprod Immunol 2025; 167:104419. [PMID: 39732055 DOI: 10.1016/j.jri.2024.104419] [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: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUNDS Recent studies have found Several lncRNAs were proved differential expression in diminished ovarian reserve (DOR) patients, however, the mechanism of DOR caused by lncRNAs is still largely unclear. METHODS High throughput sequencing was performed in ovarian GCs extracted from women with normal ovarian function and women with DOR. Bioinformation analysis was used to analyze the sequencing data and identify the differential expression of lncRNAs. Quantitative RT-PCR (qRT-PCR) was used to verify the sequencing results. Situ fluorescence hybridization (FISH) followed by confocal microscopy and qRT-PCR were used to explore the location and expression of LncRNA-THBS4 in GCs. The significantly enriched signaling pathways of LncRNA-THBS4 were identified by KEGG. The study used RNA interference technology to decipher LncRNA-THBS4 function by silencing LncRNA-THBS4 in GCs. Western blot and qRT-PCR were used to explore the mRNA and protein expressions of key factors of PI3Ks pathway. The pro-apoptotic protein and anti-apoptotic protein were detected by western blot. The proliferation and apoptosis of GCs were detected by MTT assay and Flow cytometry. RESULTS 197 lncRNAs with significant differences in expression levels were detected between control and DOR group by high throughput sequencing. The study found the expression of LncRNA-THBS4 in GCs was positively correlated with Anti-Mullerian hormone (AMH) (p = 0.0020, r = 0.4742)、antral follicle count (AFC) (p = 0.0007, r = 0.5130)、good embryo rate (p = 0.0006, r = 0.5210), negatively correlated with basal FSH level (p = 0.0007, r = -0.5152). LncRNA-THBS4 was mainly localized in the cytoplasm of GCs. LncRNA-THBS4 silencing could inhibit the PI3Ks pathway; decrease the levels of anti-apoptotic protein, inhibit the proliferation of GCs; increase the levels of apoptosis protein, enhance the apoptosis of GCs. CONCLUSIONS The expression level of lncRNA-THBS4 is correlated with ovarian function indicators and pregnancy outcomes in women. LncRNA-THBS4 may participate in the pathogenesis of DOR by affecting the proliferation and apoptosis of GCs via regulating PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yiyue Fan
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Beiiing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China.
| | - Dongmei Tian
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Zili Lv
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shiyang Peng
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shaomi Zhu
- Chengdu Fifth People's Hospital, (School of Medical and Life Sciences/Affiliated Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine), Chengdu, China.
| |
Collapse
|
146
|
Han Y, Pu Q, Fan T, Wei T, Xu Y, Zhao L, Liu S. Long non-coding RNAs as promising targets for controlling disease vector mosquitoes. INSECT SCIENCE 2025; 32:24-41. [PMID: 38783627 DOI: 10.1111/1744-7917.13383] [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: 01/09/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Hematophagous female mosquitoes are important vectors of numerous devastating human diseases, posing a major public health threat. Effective prevention and control of mosquito-borne diseases rely considerably on progress in understanding the molecular mechanisms of various life activities, and accordingly, the molecules that regulate the various life activities of mosquitoes are potential targets for implementing future vector control strategies. Many long non-coding RNAs (lncRNAs) have been identified in mosquitoes and significant progress has been made in determining their functions. Here, we present a comprehensive overview of the research advances on mosquito lncRNAs, including their molecular identification, function, and interaction with other non-coding RNAs, as well as their synergistic regulatory roles in mosquito life activities. We also highlight the potential roles of competitive endogenous RNAs in mosquito growth and development, as well as in insecticide resistance and virus-host interactions. Insights into the biological functions and mechanisms of lncRNAs in mosquito life activities, viral replication, pathogenesis, and transmission will contribute to the development of novel drugs and safe vaccines.
Collapse
Affiliation(s)
- Yujiao Han
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Qian Pu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Ting Fan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Tianqi Wei
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Yankun Xu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Lu Zhao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| |
Collapse
|
147
|
Zhou W, Tan C, Xiong D, Chen C, Zhao Y, Xie Y, Sun B, Wang Z, Xia P, Ye Z. LncRNA-MEG3 Mediated Diabetic Cerebral Ischemia-Reperfusion Injury-Induced Apoptosis via Modulating Interaction Between Annexin A2 and Akt in Mitochondria. CNS Neurosci Ther 2025; 31:e70242. [PMID: 39912333 PMCID: PMC11799844 DOI: 10.1111/cns.70242] [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: 07/09/2024] [Revised: 10/14/2024] [Accepted: 12/14/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND In clinical domains, encompassing neurosurgery and macrovascular cardiac procedures, certain interventions result in cerebral ischemia- reperfusion injury (CIRI). Diabetes mellitus (DM) increases the risk of CIRI and worsens the severity of neurological impairment. It was documented that lncRNA-MEG3 contributed to the pathogenesis of CIRI. However, the pivotal significance of lncRNA-MEG3 in diabetic CIRI has never been studied. AIMS This study's aims were two-fold, to (1) figure out the influence of lncRNA-MEG3 on neurological dysfunction subsequent to diabetic cerebral ischemic injury, (2) elucidate its potential role in mitochondria-related apoptosis via modulating the Anxa2 signaling pathway. MATERIALS AND METHODS We mainly collected plasma from clinical patients to measure the expression of lncRNA-MEG3, and explored the molecular mechanism of lncRNA-MEG3 in CIRI combined with DM by immunofluorescence, western blot, co-ip and other molecular biology experiments in rat MACO+DM model and cellular OGD/R+HG model. RESULTS LncRNA-MEG3 expression in DM+AIS cases was remarkably higher than that in cases with AIS and healthy controls. Moreover, lncRNA-MEG3 expression was strongly linked to the National Institutes of Health Stroke Scale (NIHSS) score. Additionally, the findings unveiled that lncRNA-MEG3 depletion alleviated neurological impairments following CIRI in diabetic rats, and cellular death resulted from Oxygen-glucose deprivation (OGD) plus hyperglycemic reperfusion in rat brain microvascular endothelial cells (RBMVECs) that was concomitant with the increased phosphorylation of Annexin A2 (Anxa2) at Tyr23. Meanwhile, over expression of Anxa2, identified as a lncRNA-MEG3-associated mitochondrial protein, remarkably suppressed mitochondria-derived apoptosis. Importantly, lncRNA-MEG3 knockdown enhanced the mitochondrial translocation of Anxa2 via promoting its phosphorylation at Tyr23 in OGD+HG-treated RBMVECs. Furthermore, Anxa2 enhanced Akt phosphorylation at Ser473 and bound to Akt in mitochondria, which was involved in lncRNA-MEG3 depletion-induced neuroprotection. However, lncRNA-MEG3 mobilized to mitochondria in a Plectin-dependent manner and subsequently impeded the interaction between p-Anxa2 and p-Akt. DISCUSSION AND CONCLUSION The outcomes provided clinical evidence that lncRNA-MEG3 appeared as an unfavorable prognostic factor for diabetic CIRI and revealed that lncRNA-MEG3 knockdown could be protective against diabetic CIRI-induced mitochondria-related apoptosis through modulating Anxa2 binding to Akt in mitochondria.
Collapse
Affiliation(s)
- Wanqing Zhou
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
| | - Chongyi Tan
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
| | - Di Xiong
- Department of General PracticeZhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South UniversityZhuzhouChina
| | - Cheng Chen
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
| | - Yanfei Zhao
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
| | - Yongqiu Xie
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina
| | - Bei Sun
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina
| | - Zhihua Wang
- Department of AnesthesiologyHainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital)HaikouChina
| | - Pingping Xia
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina
| | - Zhi Ye
- Department of AnesthesiologyXiangya Hospital of Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina
| |
Collapse
|
148
|
Huang LX, Sun T, Sun J, Wu ZM, Ling C, Zhang BY, Chen C, Wang H. Non-Coding RNA in Schwann Cell and Peripheral Nerve Injury: A Review. Adv Biol (Weinh) 2025; 9:e2400357. [PMID: 39185790 DOI: 10.1002/adbi.202400357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/28/2024] [Indexed: 08/27/2024]
Abstract
Peripheral nerve injury (PNI) can result in severe disabilities, profoundly impacting patients' quality of life and potentially endangering their lives. Therefore, understanding the potential molecular mechanisms that facilitate the regeneration of damaged nerves is crucial. Evidence indicates that Schwann cells (SCs) play a pivotal role in repairing peripheral nerve injuries. Previous studies have shown that RNA, particularly non-coding RNA (ncRNA), plays a crucial role in nerve regeneration, including the proliferation and dedifferentiation of SCs. In this review, the individual roles of ncRNA in SCs and PNI are analyzed. This review not only enhances the understanding of ncRNA's role in nerve injury repair but also provides a significant theoretical foundation and inspiration for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Li-Xin Huang
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Tao Sun
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Zhi-Min Wu
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Bao-Yu Zhang
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, 600 Tianhe Road, Guangzhou, Guangdong, 510630, China
| |
Collapse
|
149
|
Zhu Y, Lv Y, Yao H, Chen Z, Yang W, Tian C, Yang W, Li M, Jia Q, Wang L. Qiu's Cervical Prescription inhibit the invasion and growth of cervical cancer through LncRNA ATB/miR-126 pathway. Pathol Res Pract 2025; 266:155784. [PMID: 39709873 DOI: 10.1016/j.prp.2024.155784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Cervical cancer (CC) is one of the most deadly cancers in women, its current treatments still result in poor outcomes and developing the novel targets and therapeutic strategies are urgently needed. Qiu's Cervical Prescription (QCP) is one of the traditional Chinese medicines used in the treatment of cervical cancer in China. Although its curative effect is remarkable, the internal mechanism of its treatment is still poorly understood. Recent studies have shown that LncRNA ATB might be used as a new proliferation marker for cancer diagnosis and prognosis. This study aimed to investigate the possible mechanism of action of QCP in the treatment of cervical cancer. METHODS The functional assays of migration and invasion in vitro using transwell assays and wound healing assays was performed to confirm the pro-carcinogenic effect of LncRNA ATB, and the changes of migration and invasion of HeLa cells were observed after treatment with QCP containing drug serum. The changes in tumor volume, general condition of transplanted tumor-bearing mice and expression of LncRNA ATB pathway-related proteins were detected by qPCR, Western blotting and HE staining after treatment with the QCP. RESULTS We induced LncRNA ATB knockdown and overexpression in cervical cancer cell lines and detected the biological behavior changes in vitro. Furthermore, we established murine models using stable LncRNA ATB-shRNA HeLa cells or overexpression LncRNA ATB cells or normal Hela cells with QCP to evaluate how suppression of LncRNA ATB affects tumor growth. CONCLUSION We showed that potential mechanism of QCP in the treatment of cervical cancer may be through inhibition of the LncRNA ATB/miR-126/TGFβ1 signaling axis. In conclusion, QCP may be a promising approach for the treatment of CC.
Collapse
Affiliation(s)
- Yingping Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Lv
- Department of Pathology, No. 942 Hospital of the Chinese People's Liberation Army Logistic Support Force, Yinchuan, China
| | - Haili Yao
- Department of Central Sterile Supply, Navy Qingdao Special Service Rehabilitation Center, Qingdao, China
| | - Zhilei Chen
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenjuan Yang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuntao Tian
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Weiyi Yang
- Department of Neurology, Xi'an Daxing Hospital, Xi'an, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| | - Liangping Wang
- Center for Reproductive Medicine, Department of Reproductive Endocrinology,Zhejiang Provincial People's Hospital, Hangzhou, China.
| |
Collapse
|
150
|
Liu J, Han S, Cui Y, Zhao Q, Wang Y, Li T, Li X. LncRNA FAM30A Suppresses Proliferation and Metastasis of Colorectal Carcinoma by Blocking the JAK-STAT Signalling. J Cell Mol Med 2025; 29:e70421. [PMID: 39972942 PMCID: PMC11839745 DOI: 10.1111/jcmm.70421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/15/2025] [Accepted: 01/30/2025] [Indexed: 02/21/2025] Open
Abstract
Colorectal carcinoma (CRC) poses a serious risk to global human health. Long non-coding RNAs (LncRNAs) play an important role in the pathogenesis of CRC. There is a scarcity of data about a newly identified lncRNA, FAM30A. Our major objective is to investigate the role of FAM30A in the process of CRC. Gene expression data and correlated clinical information were retrieved and downloaded from public databases to identify differentially expressed genes linked to CRC. The expression of FAM30A was identified in clinical samples and CRC cell lines using via Quantitative Real-time Polymerase Chain Reaction (qPCR) assay also. The survival significance of FAM30A was determined via R package "survival." Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed to identify FAM30A-related signalling pathway. The levels of proteins expression were determined by western blot assay. The effect of FAM30A on CRC cell biological behaviours was evaluated by cell function experiments. FAM30A was identified down-regulated in CRC based on the data from public database. FAM30A had lower expression in CRC clinical samples and cell lines. Low FAM30A expression was positively related to a poor prognosis in CRC patients. After FAM30A was overexpressed, the proliferation, invasion, and migration abilities of CRC cells were decreased, and the rate of CRC cell apoptosis increased. Furthermore, overexpression of FAM30A could block JAK-STAT signalling. FAM30A suppresses proliferative, invasive, and migratory abilities of CRC through blocking JAK-STAT signalling. Thus, it can be a novel biomarker of CRC prognosis.
Collapse
Affiliation(s)
- Jin Liu
- Department of GastroenterologyHenan Provincial People's HospitalZhengzhouChina
| | - Shuangyin Han
- Department of GastroenterologyHenan Provincial People's HospitalZhengzhouChina
| | - Yuanbo Cui
- Trauma and Metabolic InstituteZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Qiuyan Zhao
- Department of GastroenterologyHenan Provincial People's HospitalZhengzhouChina
| | - Yunfei Wang
- Department of GastroenterologyHenan Provincial People's HospitalZhengzhouChina
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease‐Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal DiseasesTianjin Nankai Hospital, Tianjin Medical UniversityTianjinChina
| | - Xiuling Li
- Department of GastroenterologyHenan Provincial People's HospitalZhengzhouChina
| |
Collapse
|