1
|
Rong H, Hu Y, Wei W. Curcumol ameliorates diabetic retinopathy via modulating fat mass and obesity-associated protein-demethylated MAF transcription factor G antisense RNA 1. World J Diabetes 2025; 16:97201. [DOI: 10.4239/wjd.v16.i4.97201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/28/2024] [Accepted: 01/06/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major microvascular complication of diabetes mellitus, leading to significant visual impairment and blindness among adults. Current treatment options are limited, making it essential to explore novel therapeutic strategies. Curcumol, a sesquiterpenoid derived from traditional Chinese medicine, has shown anti-inflammatory and anti-cancer properties, but its potential role in DR remains unclear.
AIM To investigate the therapeutic effects of curcumol on the progression of DR and to elucidate the underlying molecular mechanisms, particularly its impact on the fat mass and obesity-associated (FTO) protein and the long non-coding RNA (lncRNA) MAF transcription factor G antisense RNA 1 (MAFG-AS1).
METHODS A streptozotocin-induced mouse model of DR was established, followed by treatment with curcumol. Retinal damage and inflammation were evaluated through histological analysis and molecular assays. Human retinal vascular endothelial cells were exposed to high glucose conditions to simulate diabetic environments in vitro. Cell proliferation, migration, and inflammation markers were assessed in curcumol-treated cells. LncRNA microarray analysis identified key molecules regulated by curcumol, and further experiments were conducted to confirm the involvement of FTO and MAFG-AS1 in the progression of DR.
RESULTS Curcumol treatment significantly reduced blood glucose levels and alleviated retinal damage in streptozotocin-induced DR mouse models. In high-glucose-treated human retinal vascular endothelial cells, curcumol inhibited cell proliferation, migration, and inflammatory responses. LncRNA microarray analysis identified MAFG-AS1 as the most upregulated lncRNA following curcumol treatment. Mechanistically, FTO demethylated MAFG-AS1, stabilizing its expression. Rescue experiments demonstrated that the protective effects of curcumol against DR were mediated through the FTO/MAFG-AS1 signaling pathway.
CONCLUSION Curcumol ameliorates the progression of DR by modulating the FTO/MAFG-AS1 axis, providing a novel therapeutic pathway for the treatment of DR. These findings suggest that curcumol-based therapies could offer a promising alternative for managing this debilitating complication of diabetes.
Collapse
Affiliation(s)
- Han Rong
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
- Department of Ophthalmology, Huai’an Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Huai’an 223002, Jiangsu Province, China
| | - Yu Hu
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an 223002, Jiangsu Province, China
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Wei Wei
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
2
|
Bashir T, Husaini AM. Role of non-coding RNAs in quality improvement of horticultural crops: computational tools, databases, and algorithms for identification and analysis. Funct Integr Genomics 2025; 25:80. [PMID: 40183947 DOI: 10.1007/s10142-025-01592-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Horticultural crops, including fruits, vegetables, flowers, and herbs, are essential for food security and economic sustainability. Advances in biotechnology, including genetic modification and omics approaches, have significantly improved these crops'traits. While initial transgenic efforts focused on protein-coding genes, recent research highlights the crucial roles of non-coding RNAs (ncRNAs) in plant growth, development, and gene regulation. ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), influence key biological processes through transcriptional and post-transcriptional regulation. This review explores the classification, functions, and regulatory mechanisms of ncRNAs, emphasizing their potential in enhancing horticultural crop quality. This growing understanding offers promising avenues for enhancing crop performance and developing new horticultural varieties with improved traits. Additionally, we elucidate the role of ncRNA databases and predictive bioinformatics tools into modern horticultural crop improvement strategies.
Collapse
Affiliation(s)
- Tanzeel Bashir
- Genome Engineering and Societal Biotechnology Lab, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Jammu and Kashmir, India
| | - Amjad M Husaini
- Genome Engineering and Societal Biotechnology Lab, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Jammu and Kashmir, India.
| |
Collapse
|
3
|
Shafiei FS, Abroun S, Vahdat S, Rafiee M. Omics approaches: Role in acute myeloid leukemia biomarker discovery and therapy. Cancer Genet 2025; 292-293:14-26. [PMID: 39798496 DOI: 10.1016/j.cancergen.2024.12.006] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults and has the highest fatality rate. Patients aged 65 and above exhibit the poorest prognosis, with a mere 30 % survival rate within one year. One important issue in optimizing outcomes for AML patients is their limited ability to predict responses to specific therapies, response duration, and likelihood of relapse. Despite rigorous therapeutic interventions, a significant proportion of patients experience relapse. Consequently, there is a pressing need to introduce new targets for therapy. Sequencing and biotechnology have come a long way in the last ten years. This has made it easier for many omics technologies, like genomics, transcriptomics, proteomics, and metabolomics, to study molecular mechanisms of AML. An integrative approach is necessary to understand a complex biological process fully and offers an important opportunity to understand the information underlying diseases. In this review, we studied papers published between 2010 and 2024 employing omics approaches encompassing diagnosis, prognosis, and risk stratification of AML. Finally, we discuss prospects and challenges in applying -omics technologies to the discovery of novel biomarkers and therapy targets. Our review may be helpful for omics researchers who want to study AML from different molecular aspects.
Collapse
Affiliation(s)
- Fatemeh Sadat Shafiei
- MSC student of Hematology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeid Abroun
- PhD in clinical Hematology, Professor of Hematology, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadaf Vahdat
- PhD of Medical Biotechnology, Assistant Professor, Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Rafiee
- PhD of Hematology, Assistant Professor, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
4
|
Yang J, Zhang D, Jiang W. Long noncoding RNA as an emerging regulator of endoderm differentiation: progress and perspectives. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:11. [PMID: 40133743 PMCID: PMC11937447 DOI: 10.1186/s13619-025-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Accumulated studies have demonstrated that long noncoding RNAs (lncRNAs) play crucial regulatory roles in diverse biological processes, such as embryonic development and cell differentiation. Comprehensive transcriptome analysis identifies extensive lncRNAs, gradually elucidating their functions across various contexts. Recent studies have highlighted the essential role of lncRNAs in definitive endoderm differentiation, underscoring their importance in early development. In this review, we have analyzed the features of overlapping, proximal, and desert lncRNAs, classified by genomic location, in pluripotent stem cells (PSCs) and the differentiation derivatives. Furthermore, we focus on the endoderm lineage and review the latest advancements in lncRNA identification and their distinct regulatory mechanisms. By consolidating current knowledge, we aim to provide a clearer perspective on how lncRNAs contribute to endoderm differentiation in different manners.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
5
|
Cao C, Hu Q, Hu X, Zhu L, Jia H, Shen Y, Chen J, Xu B, Zhang B. The role of long non-coding RNA A2M-AS1 in early diagnosis and prognosis evaluation of acute myocardial infarction. J Cardiothorac Surg 2025; 20:163. [PMID: 40133942 PMCID: PMC11934764 DOI: 10.1186/s13019-025-03381-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
AIM The objective was to assess the clinical efficacy of long non-coding RNA (lncRNA) alpha-2-macroglobulin-antisense 1 (A2M-AS1) in acute myocardial infarction (AMI). METHODS One hundred patients with AMI and eighty patients with chest pain were recruited in the case-control study. A2M-AS1 expression was examined by quantitative real-time polymerase chain reaction (qRT-PCR). Receiver operating characteristic (ROC) analysis was utilized for evaluating the diagnostic value. Pearson's correlation analysis was used to analyze the correlation between A2M-AS1 and conventional AMI biomarkers. AMI-associated risk indicators were identified using logistic regression analysis. RESULTS A significant reduction of serum A2M-AS1 was measured in AMI patients relative to chest pain patients. A2M-AS1 had an area under the curve (AUC) of 0.927 to distinguish AMI patients from those with chest pain. Pearson's correlation analysis showed that A2M-AS1 was adversely correlated with white blood cell (WBC) (r=-0.6682, P < 0.001), low density lipoprotein cholesterol (LDL-C) (r=-0.5795, P < 0.001), creatine kinase MB (CK-MB) (r=-0.6022, P < 0.001) and cTnl (r=-0.5473; P < 0.001), while positively correlated with high density lipoprotein cholesterol (HDL-C) (r = 0.6445, P < 0.001). Relative to non-Major Adverse Cardiovascular Events (non-MACE) group, serum A2M-AS1 was obviously declined in the MACE group of AMI patients with high capacity to distinguish the MACE group from the non-MACE patients (AUC = 0.802). Additionally, A2M-AS1 (P = 0.013; OR = 0.268; 95%CI = 0.095-0.760) was a risk indicator for predicting MACE with AMI patients, as well as age (P = 0.014; OR = 3.478; 95%CI = 1.285-9.414). CONCLUSION A reduction in A2M-AS1 expression was observed in AMI patients, suggesting its potential as an underlying indicator for AMI diagnosis.
Collapse
Affiliation(s)
- Chunming Cao
- Department of Cardiology, The Second Affiliated Hospital of Qiqihar Medical university, Qiqihar, 161006, China
| | - Qiyuan Hu
- Department of Cardiology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xinyue Hu
- Shanghai Baoshan Luodian Hospital, No.121, Luoxi Road, Baoshan District, Shanghai, 201908, China
| | - Lijun Zhu
- Department of Intervention Radiology, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Huili Jia
- Shanghai Baoshan Luodian Hospital, No.121, Luoxi Road, Baoshan District, Shanghai, 201908, China
| | - Yongjian Shen
- Shanghai Wusong Central Hospital, Shanghai, 200940, China
| | - Jun Chen
- Baoshan Brance, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200444, China
| | - Bin Xu
- Shanghai Baoshan Luodian Hospital, No.121, Luoxi Road, Baoshan District, Shanghai, 201908, China.
| | - Boqing Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, No. 290, Heyan Road, Qixia District, Nanjing, 210011, China.
| |
Collapse
|
6
|
Wang C, Huang Y, Li L, Huang X, Huang Y, Fang X, Long Y. Antiviral Therapy-Induced Changes in Long Non-Coding RNA Expression Profiles in Umbilical Cord Blood and Placental Tissues of Hepatitis B Virus-Infected Pregnant Women. Int J Womens Health 2025; 17:835-844. [PMID: 40123756 PMCID: PMC11927581 DOI: 10.2147/ijwh.s511524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025] Open
Abstract
Background Hepatitis B virus (HBV) is a major global health concern, with maternal-fetal transmission being the primary route of transmission, which can lead to chronic HBV infection in newborns. Long non-coding RNAs (lncRNAs) play crucial roles in gene regulation and immune responses, but their involvement in HBV transmission during pregnancy remains unclear. This study aimed to assess the impact of tenofovir disoproxil fumarate (TDF)-based antiviral therapy on lncRNA expression profiles and immune signaling pathways in umbilical cord blood and placental tissues and to identify potential therapeutic targets for preventing intrauterine HBV infection. Materials and Methods Umbilical cord serum and placental tissues were collected from six HBV carriers. Three carriers received TDF-based antiviral therapy, and the remaining carriers who did not receive antiviral therapy served as controls. LncRNA microarray analysis and bioinformatics were used to evaluate the effects of antiviral therapy on lncRNA expression profiles and signaling pathways. Results Antiviral therapy exerted minimal effects on lncRNA expression profiles in umbilical cord blood. In placental tissues, significant alterations in lncRNA expression profiles were observed, including 249 upregulated and 381 downregulated lncRNAs. Antiviral therapy activated innate immune pathways, such as intracellular DNA sensing, chemokine signaling, type I interferon, Jak-Stat, and interferon-γ-mediated adaptive immunity. Through intersection analysis, CPED1 was found differentially expressed in both cord blood and placental tissues. KEGG pathway analysis suggested that low CPED1 expression may inhibit virus transmission via the JAK-STAT pathway. Conclusion This study demonstrated that TDF-based antiviral therapy altered lncRNA expression and activated immune signaling pathways in placental tissues, offering insights into the molecular mechanisms of maternal-fetal HBV transmission.
Collapse
Affiliation(s)
- Cuimin Wang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yuting Huang
- Department of Obstetrics & Gynecology, Youjiang Medical College for Nationalities, Baise City, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Lanfeng Li
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xizhen Huang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yin Huang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xiang Fang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yu Long
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Medical Simulator Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
7
|
Dai C, Li Q, Wang L, Zhang J, Yang S, Zhang X. Long Noncoding LINC00115 Facilitates Cell Growth and Inhibits Apoptosis by Regulating the miR-4701-5p/P4HB Axis in Bladder Cancer. TOHOKU J EXP MED 2025; 265:69-81. [PMID: 39111879 DOI: 10.1620/tjem.2024.j075] [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: 03/18/2025]
Abstract
Bladder cancer (BCa) is a prevalent urogenital malignancy, imposing a significant burden on health-care systems worldwide. Long noncoding RNAs (lncRNAs) are important regulators of carcinogenesis and affect BCa progression. In this study, the influence of lncRNA LINC00115 on malignant behavior of BCa cells were explored. Bioinformatics method was used for prediction of gene expression and downstream molecules of LIN00115. LINC00115 expression level in BCa cells was measured using RT-qPCR. After LINC00115 depletion, the proportion of viable, proliferative, and apoptotic BCa cells were calculated by methyl thiazolyl tetrazolium (MTT) assays, colony formation assays, and TUNEL staining, respectively. FISH was performed to verify the cellular distribution of LINC00115. The interaction between LINC00115 and miR-4701-5p and the binding between miR-4701-5p and P4HB were confirmed using RNA pulldown, RNA immunoprecipitation (RIP), and luciferase reporter assays. Experimental results showed that LINC00115 was highly expressed in BCa cells. The silencing of LINC00115 restrained BCa cell proliferation and stimulated apoptosis. LINC00115 could directly bind to miR-4701-5p and thus initiate P4HB upregulation in BCa cells. P4HB 3'untranslated region could be targeted by miR-4701-5p. Additionally, Amplification of P4HB expression offset the effects of LINC00115 knockdown on BCa cell proliferative and apoptotic behaviors. In conclusion, LINC00115 facilitates BCa cell growth and inhibits apoptosis via interaction with miR-4701-5p and upregulation of P4HB.
Collapse
Affiliation(s)
- Changyuan Dai
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Qingwen Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Lili Wang
- Department of Emergency medicine, The First Affiliated Hospital of Bengbu Medical College
| | - Jiajun Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Xiaole Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| |
Collapse
|
8
|
Roy D, Bhattacharya B, Chakravarti R, Singh P, Arya M, Kundu A, Patil A, Siva B, Mehta S, Kazi TA, Ghosh D. LncRNAs in oncogenic microenvironment: from threat to therapy. Front Cell Dev Biol 2025; 12:1423279. [PMID: 40176927 PMCID: PMC11962222 DOI: 10.3389/fcell.2024.1423279] [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: 04/25/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
LncRNAs are RNA molecules of more than 200 nucleotides in length and participate in cellular metabolism and cellular responses through their diverse interactomedespite having no protein-coding capabilities. Such significant interactions also implicate the presence of lncRNAs in complex pathobiological pathways of various diseases, affecting cellular survival by modulating autophagy, inflammation and apoptosis. Proliferating cells harbour a complex microenvironment that mainly stimulate growth-specific activities such as DNA replication, repair, and protein synthesis. They also recognise damages at the macromolecular level, preventing them from reaching the next-generation. LncRNAs have shown significant association with the events occurring towards proliferation, regulating key events in dividing cells, and dysregulation of lncRNA transcriptome affects normal cellular life-cycle, promoting the development of cancer. Furthermore, lncRNAs also demonstrated an association with cancer growth and progression by regulating key pathways governing cell growth, epithelial-mesenchymal transition and metastasis. This makes lncRNAs an attractive target for the treatment of cancer and can also be used as a marker for the diagnosis and prognosis of diseases due to their differential expression in diseased samples. This review delves into the correlation of the lncRNA transcriptome with the fundamental cellular signalling and how this crosstalk shapes the complexity of the oncogenic microhabitat.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata, Kolkata, India
| |
Collapse
|
9
|
Wu D, Liu C, Ding L. Follicular metabolic dysfunction, oocyte aneuploidy and ovarian aging: a review. J Ovarian Res 2025; 18:53. [PMID: 40075456 PMCID: PMC11900476 DOI: 10.1186/s13048-025-01633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
With the development of modern society and prolonged education, more women choose to delay their childbearing age, which greatly increases the number of women aged older than 35 years with childbearing needs. However, with increasing age, the quantity and quality of oocytes continue to fall, especially with increasing aneuploidy, which leads to a low in vitro fertilization (IVF) success rate, high abortion rate and high teratogenesis rate in assisted reproduction in women with advanced maternal age. In addition to genetics and epigenetics, follicular metabolism homeostasis is closely related to ovarian aging and oocyte aneuploidy. Glucose, lipid, and amino acid metabolism not only provide energy for follicle genesis but also regulate oocyte development and maturation. This review focuses on the relationships among follicular metabolism, oocyte aneuploidy, and ovarian aging and discusses potential therapeutic metabolites for ovarian aging.
Collapse
Affiliation(s)
- Die Wu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China
| | - Chuanming Liu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China.
- State Key Laboratory of Analytic Chemistry for Life Science, Nanjing University, Nanjing, 210093, China.
- Clinical Center for Stem Cell Research, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
10
|
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
|
11
|
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] [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
|
12
|
Yan J, Li Z, Shu Y, Chen H, Wang T, Li X, Zhang Y, Li L, Zhang Y. The Unveiled Novel regulator of Adeno-associated virus production in HEK293 cells. Gene 2025; 938:149122. [PMID: 39581356 DOI: 10.1016/j.gene.2024.149122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
The field of gene therapy using Adeno-associated viral (AAV) vector delivery is rapidly advancing in the biotherapeutics industry. Despite its successes, AAV manufacturing remains a challenge due to limited production yields. The triple plasmid transfection of HEK293 cells represents the most extensively utilized system for AAV production. The regulatory factors and mechanisms underlying viral production in HEK293 cells are largely unknown. In this study, we isolated high-titer AAV production clones from a parental HEK293 population using a single limiting dilution step, and subsequently elucidating their underlying molecular mechanisms through whole transcriptome analysis. LncRNA TCONS_00160397 was upregulated in clones and shown to promoted HEK293 cells proliferation and improved the titer of AAV production. Mechanistically, results from proteomics and metabolomics indicated that TCONS_00160397 regulated the ABC transporters pathway. These findings furnish a rich repository of knowledge and actionable targets for the rational optimization of HEK293-based producer lines, thereby paving the way for tangible improvements in AAV vector output and expediting the broad implementation of gene therapies.
Collapse
Affiliation(s)
- Junyu Yan
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Ziqian Li
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yue Shu
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Hui Chen
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Tianxingzi Wang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Xin Li
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yuhang Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - LiLi Li
- Beijing Institute of Biological Products Company Limited, Beijing, China.
| | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China; China National Biotec Group Company Limited, Beijing, China.
| |
Collapse
|
13
|
Saeed BI, Kumar A, Oghenemaro EF, Almutairi LA, M RM, Kumawat R, Uthirapathy S, Hulail HM, Sharma S, Ravi Kumar M. Interactions between lncRNAs and cyclins/CDKs complexes; key players in determining cancer cell response to CDKs inhibitors. Exp Cell Res 2025; 445:114406. [PMID: 39761840 DOI: 10.1016/j.yexcr.2025.114406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025]
Abstract
Transcription takes place over a significant portion of the human genome. However, only a small portion of the transcriptome, roughly 1.2 %, consists of RNAs translated into proteins; the majority of transcripts, on the other hand, comprise a variety of RNA families with varying sizes and functions. A substantial portion of this diverse RNA universe consists of sequences longer than 200 bases, called the long non-coding RNA (lncRNA). The control of gene transcription, changes to DNA topology, nucleosome organization and structure, paraspeckle creation, and assistance for developing cellular organelles are only a few of the numerous tasks performed by lncRNA. The main focus of this study is on the function of lncRNA in controlling the levels and actions of cyclin-dependent kinase inhibitors (CDKIs). The enzymes required for the mitotic cycle's regulated progression are called cyclin-dependent kinases (CDKs). They have many degrees of regulation over their activities and interact with CDKIs as their crucial mechanisms. Interestingly, culminating evidence has clarified that lncRNAs are associated with several illnesses and use CDKI regulation to control cellular function. Nonetheless, despite the abundance of solid evidence in the literature, it still seems unlikely that lncRNA will have much of an impact on controlling cell proliferation or modulating CDKIs.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia; Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, 641021, India.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, PMB 1, Abraka, Delta State, Nigeria.
| | - Layla A Almutairi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia.
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Subasini Uthirapathy
- Faculty of Pharmacy, Pharmacology Department, Tishk International University, Erbil, Kurdistan Region of Iraq, Iraq.
| | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq.
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India.
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India.
| |
Collapse
|
14
|
Chen X, Ding W, Liu Y, Liu H, Zhang C, Huang L. Innovative approaches in atherosclerosis treatment: Harnessing traditional Chinese medicine to target long non-coding RNAs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156488. [PMID: 39938175 DOI: 10.1016/j.phymed.2025.156488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Atherosclerosis (AS) is a major contributor to cardiovascular diseases, characterized by high morbidity and mortality rates. Long non-coding RNAs (LncRNAs), as members of non-protein coding RNAs, play a crucial role in various biological processes that maintain homeostasis and influence disease progression. Research indicates that lncRNAs are involved in the pathogenesis of AS. PURPOSE In this study, we aim to explore the role of lncRNAs in the pathogenesis of AS and the latest progress in the prevention and treatment of AS by targeted regulation of lncRNAs by traditional Chinese medicine (TCM), in order to provide more new beneficial targets for the treatment of AS and expand the application of TCM in the treatment of cardiovascular diseases. METHOD The literature was retrieved, analyzed, and collected using PubMed, Web of Science, Sci-Hub, CNKI, Elsevier, ScienceDirect, SpringerLink, and Google Scholar. Search terms include "atherosclerosis", "traditional Chinese medicine", "natural products", "active ingredient", "lncRNAs", "herbal medicine", "cardiovascular diseases", "pharmacology", "toxicology", "clinical trials", etc., and several combinations of these keywords. RESULTS This study examines the primary mechanisms through which lncRNAs induce AS, such as dysfunction in endothelial cells, abnormal proliferation of vascular smooth muscle cells, cholesterol buildup in macrophages, formation of foam cells, inflammatory responses, and imbalances in lipid metabolism. Additionally, it summarizes 16 herbal monomers and 6 Chinese herbal compounds, along with an analysis of the toxicological aspects of TCM. CONCLUSION The study explores the existing approaches for modulating lncRNAs and emphasizes the significance and potential of herbal monomers, extracts, and formulations in this context.
Collapse
Affiliation(s)
- Xiaofang Chen
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Wenyan Ding
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yifan Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Hao Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
15
|
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
|
16
|
Yan S, Fu P, Zhu Y, Li H, Shan R, Gong B. Whole transcriptome and proteome analyses identify ncRNAs and mRNAs to predict competing endogenous RNA networks in hepatitis B virus-induced hepatocellular carcinoma. Microb Pathog 2025; 199:107248. [PMID: 39710348 DOI: 10.1016/j.micpath.2024.107248] [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: 09/16/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
The presence of the Hepatitis B virus (HBV) is considered as a valuable risk factor of hepatocellular carcinoma (HCC). To more deeply comprehend the molecular mechanism and transcriptome of HBV-induced HCC, we utilized tandem mass tagging (TMT)-based quantitative proteomics analysis and whole-transcriptome sequencing to analyze three sets of matched HepG2 hepatoma cells and HBV-positive HepAD38 cells. The differentially expressed (DE) proteins (1596), mRNAs (5263), miRNAs (581), lncRNAs (2672) and circRNAs (222) were subjected to differential expression and enrichment analyses in order to thoroughly assess the gene-regulatory circuits of HBV-induced HCC. Subsequently, the amounts of 321 DEproteins-DEmRNAs with common alterations were confirmed. According to functional pathway analysis, the DEproteins-DEmRNAs were primarily linked to signaling pathways, amino acid metabolism, and cellular function. Furthermore, the viability and significance of the ceRNA regulatory networks, LOC105377730/miR-4726-5p/FHL2 and hsa_circ_0001098/miR-2110/IGF2BP1, were randomly chosen and confirmed. Our work provides a valuable asset in terms of understanding regulatory activities at the RNA level, and might reveal fresh information about the fundamental mechanism and potential therapeutic targets of HBV-induced HCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/virology
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/virology
- Liver Neoplasms/genetics
- Hepatitis B virus/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Gene Regulatory Networks
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Gene Expression Profiling
- Transcriptome/genetics
- Proteomics
- Hep G2 Cells
- Proteome
- RNA, Circular/genetics
- RNA, Untranslated/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Hepatitis B/complications
- Hepatitis B/virology
- Gene Expression Regulation, Neoplastic
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Shaoying Yan
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Diagnosis of Infectious Diseases, Nanchang, Jiangxi, China
| | - Peng Fu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yali Zhu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huiming Li
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Renfeng Shan
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
17
|
Liu F, Yang H, Liu X, Ning Y, Wu Y, Yan X, Zheng H, Liu C. LncRNA CCAT1 knockdown suppresses tongue squamous cell carcinoma progression by inhibiting the ubiquitination of PHLPP2. Mol Cell Biochem 2025; 480:1063-1075. [PMID: 38763996 DOI: 10.1007/s11010-024-05004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 05/21/2024]
Abstract
Tongue squamous cell carcinoma (TSCC) is prevailing malignancy in the oral and maxillofacial region, characterized by its high frequency. LncRNA CCAT1 can promote tumorigenesis and progression in many cancers. Here, we investigated the regulatory mechanism by which CCAT1 influences growth and metastasis of TSCC. Levels of CCAT1, WTAP, TRIM46, PHLPP2, AKT, p-AKT, and Ki67 in TSCC tissues and cells were assessed utilizing qRT-PCR, Western blot and IHC. Cell proliferation, migration, and invasion were evaluated utilizing CCK8, colony formation, wound healing and transwell assays. Subcellular localization of CCAT1 was detected utilizing FISH assay. m6A level of CCAT1 was assessed using MeRIP. RNA immunoprecipitation (RIP), Co-immunoprecipitation (Co-IP) and RNA pull down elucidated binding relationship between molecules. Nude mouse tumorigenesis experiments were used to verify the TSCC regulatory function of CCAT1 in vivo. Metastatic pulmonary nodules were observed utilizing hematoxylin and eosin (HE) staining. CCAT1 silencing repressed TSCC cell proliferation, migration and invasion. Expression of CCAT1 was enhanced through N6-methyladenosine (m6A) modification of its RNA, facilitated by WTAP. Moreover, IGF2BP1 up-regulated CCAT1 expression by stabilizing its RNA transcript. CCAT1 bond to PHLPP2, inducing its ubiquitination and activating AKT signaling. CCAT1 mediated the ubiquitination and degradation of PHLPP2 by TRIM46, thereby promoting TSCC growth and metastasis. CCAT1/TRIM46/PHLPP2 axis regulated proliferation and invasion of TSCC cells, implying that CCAT1 would be a novel therapeutic target for TSCC patients.
Collapse
MESH Headings
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Humans
- Tongue Neoplasms/pathology
- Tongue Neoplasms/genetics
- Tongue Neoplasms/metabolism
- Ubiquitination
- Mice
- Phosphoprotein Phosphatases/metabolism
- Phosphoprotein Phosphatases/genetics
- Animals
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Cell Proliferation
- Mice, Nude
- Cell Line, Tumor
- Female
- Cell Movement
- Male
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Disease Progression
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
Collapse
Affiliation(s)
- Feng Liu
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China.
- Department of Stomatology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China.
| | - Hanlin Yang
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Xiongwei Liu
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Yangbo Ning
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Yiwei Wu
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Xinglan Yan
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Huixi Zheng
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| | - Chang Liu
- Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan Province, China
| |
Collapse
|
18
|
Valverde A, George A, Nares S, Naqvi AR. Emerging therapeutic strategies targeting bone signaling pathways in periodontitis. J Periodontal Res 2025; 60:101-120. [PMID: 39044454 PMCID: PMC11873684 DOI: 10.1111/jre.13326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/22/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024]
Abstract
Periodontitis is a multifactorial immune-mediated disease exacerbated by dysregulated alveolar bone homeostasis. Timely intervention is crucial for disease management to prevent tooth loss. To successfully manage periodontitis, it is imperative to understand the cellular and molecular mechanisms involved in its pathogenesis to develop novel treatment modalities. Non-surgical periodontal therapy (NSPT) such as subgingival instrumentation/debridement has been the underlying treatment strategy over the past decades. However, new NSPT approaches that target key signaling pathways regulating alveolar bone homeostasis have shown positive clinical outcomes. This narrative review aims to discuss endogenous bone homeostasis mechanisms impaired in periodontitis and highlight the clinical outcomes of preventive periodontal therapy to avoid invasive periodontal therapies. Although the anti-resorptive therapeutic adjuncts have demonstrated beneficial outcomes, adverse events have been reported. Diverse immunomodulatory therapies targeting the osteoblast/osteoclast (OB/OC) axis have shown promising outcomes in vivo. Future controlled randomized clinical trials (RCT) would help clinicians and patients in the selection of novel preventing therapies targeting key molecules to effectively treat or prevent periodontitis.
Collapse
Affiliation(s)
- Araceli Valverde
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Anne George
- Department of Oral BiologyCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Salvador Nares
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Afsar R. Naqvi
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
- Department of Microbiology and ImmunologyUniversity of Illinois ChicagoChicagoIllinoisUSA
| |
Collapse
|
19
|
Heidari R, Assadollahi V, Marashi SN, Elahian F, Mirzaei SA. Identification of Novel lncRNAs Related to Colorectal Cancer Through Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5538575. [PMID: 39949372 PMCID: PMC11824705 DOI: 10.1155/bmri/5538575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/15/2024] [Indexed: 02/16/2025]
Abstract
Long noncoding RNA (lncRNA) plays a critical role in cancer cell proliferation, invasion, metastasis, and chemoresistance. The current study introduces novel lncRNAs in colorectal cancer (CRC) through bioinformatics analysis. GSE134834 CRC-related microarray of Gene Expression Omnibus (GEO) was analyzed to identify differentially expressed genes (DEGs) in CRC samples against normal samples. Analysis revealed 6763 DEGs (p < 0.05 and |log fold change (FC)| ≥ 0.5) that include differentially expressed mRNA (DEmRNA) and differentially expressed long noncoding RNA (DElncRNA). Novel lncRNAs were identified, and to better understand the biological function of the identified lncRNAs, gene modules were constructed using weighted gene coexpression network analysis (WGCNA), and finally, two modules for lncRNAs were obtained. The coexpression modules with these lncRNAs were subjected to enrichment analysis in FunRich software to predict their functions through their coexpressed genes. Gene ontology results of modules related to novel lncRNA revealed they significantly enriched the cellular pathways regulation in cancer. The protein-protein interaction (PPI) network of novel lncRNAs-related modules was constructed using Search Tool for the Retrieval of Interacting Genes (STRING) and visualized using the Cytoscape software. Hub genes were screened from the PPI network by the CytoHubba plug-in of Cytoscape. The hub genes were MRTO4, CDK1, CDC20, RPF2, NOP58, NIFK, GTPBP4, BUB1, BUB1B, and BOP1 for the lightpink4 module and BYSL, RPS23 (ribosomal protein S23), RSL1D1 (ribosomal L1 domain containing 1), NAT10, NOP14, GNL2, MRPS12, NOL6 (nucleolar protein 6), IMP4, and RRP12 (ribosomal RNA processing 12 homolog) for the pink module. The expression levels of the top DEmRNA and module hub genes in CRC were validated using the Gene Expression Profiling Interactive Analysis (GEPIA) database. Generally, our findings offer crucial insight into the hub genes and novel lncRNAs in the development of CRC by bioinformatics analysis, information that may prove useful in the identification of new biomarkers and treatment targets in CRC; however, more experimental investigation is required to validate the findings of the present study.
Collapse
Affiliation(s)
- Razieh Heidari
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahideh Assadollahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyedeh Negar Marashi
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Advanced Technologies Core, Baylor College of Medicine, Houston, Texas, USA
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
20
|
Shirani N, Abdi N, Chehelgerdi M, Yaghoobi H, Chehelgerdi M. Investigating the role of exosomal long non-coding RNAs in drug resistance within female reproductive system cancers. Front Cell Dev Biol 2025; 13:1485422. [PMID: 39925739 PMCID: PMC11802832 DOI: 10.3389/fcell.2025.1485422] [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: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Exosomes, as key mediators of intercellular communication, have been increasingly recognized for their role in the oncogenic processes, particularly in facilitating drug resistance. This article delves into the emerging evidence linking exosomal lncRNAs to the modulation of drug resistance mechanisms in cancers such as ovarian, cervical, and endometrial cancer. It synthesizes current research findings on how these lncRNAs influence cancer cell survival, tumor microenvironment, and chemotherapy efficacy. Additionally, the review highlights potential therapeutic strategies targeting exosomal lncRNAs, proposing a new frontier in overcoming drug resistance. By mapping the interface of exosomal lncRNAs and drug resistance, this article aims to provide a comprehensive understanding that could pave the way for innovative treatments and improved patient outcomes in female reproductive system cancers.
Collapse
Affiliation(s)
- Nooshafarin Shirani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Neda Abdi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
21
|
Ambalavanan N, Cotten CM, Erickson SW, Mathur R, Torgerson D, Ballard PL. Genomic Differences between Spontaneous versus Indicated Extreme Preterm Birth. Am J Perinatol 2025; 42:238-249. [PMID: 38889886 PMCID: PMC11693484 DOI: 10.1055/a-2347-3751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
OBJECTIVE Extremely preterm infants are at high risk of neonatal mortality and morbidity. Extreme preterm birth (PTB) may result from spontaneous preterm labor or preterm premature rupture of membranes or may be indicated due to preeclampsia, eclampsia, hypertension, or other causes. Our objective was to identify single nucleotide polymorphisms (SNPs) and biological pathways associated with spontaneous versus indicated extreme PTB using the neonatal genome. STUDY DESIGN We evaluated 523 spontaneous births and 134 indicated births weighing 401 to 1,000 g at birth from the Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network's Genomics dataset by genome-wide association study (GWAS) and pathway analysis. The TOLSURF cohort was used to replicate the results. RESULTS In the NRN GWAS, no statistically significant results were found, although the Manhattan plot showed one almost significant peak (rs60854043 on chromosome 14 at p = 1.03E-07) along with many other modest peaks at p = 1-9E-06, for a total of 15 suggestive associations at this locus. In the NRN pathway analysis, multiple pathways were identified, with the most significant being "GO_mf:go_low_density_lipoprotein_particle_receptor_activity" at p = 1.14E-06. However, these results could not be replicated in the TOLSURF cohort. CONCLUSION Genomic differences are seen between infants born by spontaneous versus indicated extreme PTB. Due to the limited sample size, there is a need for larger studies. KEY POINTS · Genomic differences are seen between infants born by spontaneous versus indicated very PTB.. · Future studies with large sample sizes evaluating extreme PTB are necessary.. · Spontaneous PTB is more common than indicated extreme PTB..
Collapse
Affiliation(s)
| | | | | | - Ravi Mathur
- Genomics Research Center, RTI International, Research Triangle Park, NC
| | - Dara Torgerson
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA
| | - Philip L. Ballard
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | | | | |
Collapse
|
22
|
Sun L, Ye X, Wang L, Yu J, Wu Y, Hua Y, Dai L. Dysregulated Long Non-coding RNAs in Myasthenia Gravis- A Mini-Review. Curr Mol Med 2025; 25:2-12. [PMID: 38192147 DOI: 10.2174/0115665240281531231228051037] [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: 09/27/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
Myasthenia gravis (MG) is an acquired autoimmune disease that is mediated by humoral immunity, supplemented by cellular immunity, along with participation of the complement system. The pathogenesis of MG is complex; although autoimmune dysfunction is clearly implicated, the specific mechanism remains unclear. Long non-coding RNAs (lncRNAs) are a class of non-coding RNA molecules with lengths greater than 200 nucleotides, with increasing evidence of their rich biological functions and high-level structure conservation. LncRNAs can directly interact with proteins and microRNAs to regulate the expression of target genes at the transcription and post-transcription levels. In recent years, emerging studies have suggested that lncRNAs play roles in the differentiation of immune cells, secretion of immune factors, and complement production in the human body. This suggests the involvement of lncRNAs in the occurrence and progression of MG through various mechanisms. In addition, the differentially expressed lncRNAs in peripheral biofluid may be used as a biomarker to diagnose MG and evaluate its prognosis. Moreover, with the development of lncRNA expression regulation technology, it is possible to regulate the differentiation of immune cells and influence the immune response by regulating the expression of lncRNAs, which will provide a potential therapeutic option for MG. Here, we review the research progress on the role of lncRNAs in different pathophysiological events contributing to MG, focusing on specific lncRNAs that may largely contribute to the pathophysiology of MG, which could be used as potential diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Liying Sun
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| | - Xuhui Ye
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| | - Linlin Wang
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| | - Junping Yu
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| | - Yan Wu
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| | - Yun Hua
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Lihua Dai
- Intensive Care Unit, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
23
|
Ammar S, Abdelbaki T, Elsabaa B, El Assi H, Kassem H. Changes in Circulating Levels of Long Non-Coding RNA p5549 and p19461 Following Metabolic Bariatric Surgery (MBS): A Prospective Study. Obes Surg 2025; 35:131-140. [PMID: 39652216 PMCID: PMC11717812 DOI: 10.1007/s11695-024-07596-5] [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: 06/05/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Obesity is attributed to a combination of factors such as lifestyle, environmental influences, and genetic background. Nowadays, the issue of obesity has grown to an epidemic scale. Environmental changes, having contributed to the sharp rise in obesity prevalence, are not the only contributing etiologic factors. Inherent biological variables interact with environmental factors resulting in obesity. Epigenetic mechanisms may explain part of obesity heritability. One of the recently discovered epigenetic mechanisms for controlling gene expression is long non-coding RNAs (lncRNAs). Circulating lncRNA p5549 and p19461 levels were reported to be significantly lower in individuals with obesity. This study aimed to evaluate whether weight loss following metabolic/bariatric surgery (MBS) can be related to altered expression levels of those lncRNAs, which have been reported to be reduced in individuals with obesity. METHODS Comparison of circulating levels of lncRNA p5549 and p19461 before and 12 weeks after MBS in thirty-four patients was conducted to evaluate whether MBS can revert the altered levels of these lncRNAs. None of the participating patients were lost to follow-up, and all underwent re-evaluation of post-surgical expression levels. RESULTS lncRNA p5549 expression levels in serum were found to increase significantly in the postoperative samples compared to preoperative samples (fold increase: 4.63 ± 7.68, p = 0.014). CONCLUSION Epigenetic changes in patients with obesity, specifically lncRNA-p5549 expression levels, are reversed after MBS. The postoperative increase in the expression levels of lncRNA- p19461 was not statistically significant.
Collapse
|
24
|
Pinskaya M, Jarroux J, Cipolla R, Morillon A. Nascent and Mature RNA Profiling by Subcellular Fractionation in Human Cells. Methods Mol Biol 2025; 2863:283-296. [PMID: 39535716 DOI: 10.1007/978-1-0716-4176-7_17] [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: 11/16/2024]
Abstract
Transcription and RNA decay determine steady-state RNA levels in cells available for translation and RNA-mediated regulatory functions. Both processes can be assessed by various techniques, for majority, based on RNA labelling or chromatin immunoprecipitation, but require a high level of expertise. Here, we describe a cost-effective, fast, and simple protocol that enables the profiling of nascent and mature RNA in the cytoplasm, nucleoplasm, and chromatin through subcellular fractionation. The workflow can include α-amanitin inhibition of RNA Polymerase II to assess nascent RNAs as a proxy of transcriptional activity, or it can be used without this treatment to investigate distribution of partially processed or mature transcripts across distinct subcellular compartments. It is applicable for studying any of RNA biotypes, including small and long noncoding RNAs, mRNAs, and their splice variants, on both transcript-specific and transcriptome-wide scales. Nascent or mature RNAs isolated from each fraction can be further analyzed by any technique of choice (northern blot, reverse transcription, RNA sequencing).
Collapse
Affiliation(s)
- Marina Pinskaya
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne Université, PSL University, Institut Curie, Centre de Recherche, Paris, France.
| | - Julien Jarroux
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Rocco Cipolla
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne Université, PSL University, Institut Curie, Centre de Recherche, Paris, France
| | - Antonin Morillon
- ncRNA, Epigenetic and Genome Fluidity, CNRS UMR3244, Sorbonne Université, PSL University, Institut Curie, Centre de Recherche, Paris, France.
| |
Collapse
|
25
|
Poloni JF, Oliveira FHS, Feltes BC. Localization is the key to action: regulatory peculiarities of lncRNAs. Front Genet 2024; 15:1478352. [PMID: 39737005 PMCID: PMC11683014 DOI: 10.3389/fgene.2024.1478352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
To understand the transcriptomic profile of an individual cell in a multicellular organism, we must comprehend its surrounding environment and the cellular space where distinct molecular stimuli responses are located. Contradicting the initial perception that RNAs were nonfunctional and that only a few could act in chromatin remodeling, over the last few decades, research has revealed that they are multifaceted, versatile regulators of most cellular processes. Among the various RNAs, long non-coding RNAs (LncRNAs) regulate multiple biological processes and can even impact cell fate. In this sense, the subcellular localization of lncRNAs is the primary determinant of their functions. It affects their behavior by limiting their potential molecular partner and which process it can affect. The fine-tuned activity of lncRNAs is also tissue-specific and modulated by their cis and trans regulation. Hence, the spatial context of lncRNAs is crucial for understanding the regulatory networks by which they influence and are influenced. Therefore, predicting a lncRNA's correct location is not just a technical challenge but a critical step in understanding the biological meaning of its activity. Hence, examining these peculiarities is crucial to researching and discussing lncRNAs. In this review, we debate the spatial regulation of lncRNAs and their tissue-specific roles and regulatory mechanisms. We also briefly highlight how bioinformatic tools can aid research in the area.
Collapse
Affiliation(s)
| | | | - Bruno César Feltes
- Department of Biophysics, Laboratory of DNA Repair and Aging, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
26
|
Lin K, Yi Z, Lv S, Zhang B, Guo Z, Li Y. Uncovering the key lncRNAs in regulating cadmium accumulation and translocation in sweet sorghum. PLANTA 2024; 261:12. [PMID: 39661199 DOI: 10.1007/s00425-024-04589-7] [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: 09/07/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
MAIN CONCLUSION 1988 lncRNAs were identified in sweet sorghum roots under cadmium treatment; lncRNA 15962 and lncRNA 11558 were validated to be the key lncRNAs involved in regulating cadmium accumulation and translocation. Cadmium (Cd) has become one of the most harmful and widespread pollutants with industry development. Sweet sorghum is an ideal plant for phytoremediation of Cd-contaminated soil. However, little is known about the regulatory role of long non-coding RNAs (lncRNAs) associated with Cd stress response in sweet sorghum. Here, lncRNA-seq was carried out in the roots of two contrasting sweet sorghum genotypes (high-Cd accumulation genotype 'H18', and low-Cd accumulation genotype 'L69'). A total of 1988 lncRNAs were characterized, including 52 and 69 differentially expressed lncRNAs in 'H18' and 'L69' in response to Cd stress, respectively. Furthermore, the trans- or cis-target genes of lncRNAs were investigated. Then, 65 lncRNAs were characterized as the probable target of 117 miRNAs and 1888 genes were identified as putative cis-target genes of Cd-responsive lncRNAs. The dual-luciferase reporter assay indicated lncRNA 15962 may serve as the endogenous target mimics of sbi-miR5565e, which targeted two genes (Sobic.005G212900 and Sobic.009G144700) involved in cell wall metabolism. Four cis-target genes including SbYS1 which encoding a Cd chelate transporter, were up-regulated by overexpression of their corresponding lncRNAs in sweet sorghum protoplasts, suggesting the positive regulatory role of lncRNAs to these cis-target genes. Moreover, the expression of SbYS1 decreased when lncRNA 11558 was inhibited by exogenous miRNA application in 'H18' seedlings, further demonstrating the positive regulatory role of lncRNA 11558 to SbYS1. Altogether, our findings shed light on the regulatory role of lncRNAs associated with Cd accumulation and translocation in sweet sorghum.
Collapse
Affiliation(s)
- Kangqi Lin
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China
- China National Botanical Garden, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ze Yi
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sulian Lv
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China
- China National Botanical Garden, Beijing, China
| | - Bo Zhang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China
- China National Botanical Garden, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zijin Guo
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China
- China National Botanical Garden, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yinxin Li
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, China.
- China National Botanical Garden, Beijing, China.
| |
Collapse
|
27
|
Hussain S, Gupta G, Shahwan M, Bansal P, Kaur H, Deorari M, Pant K, Ali H, Singh SK, Rama Raju Allam VS, Paudel KR, Dua K, Kumarasamy V, Subramaniyan V. Non-coding RNA: A key regulator in the Glutathione-GPX4 pathway of ferroptosis. Noncoding RNA Res 2024; 9:1222-1234. [PMID: 39036600 PMCID: PMC11259992 DOI: 10.1016/j.ncrna.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/26/2024] [Accepted: 05/19/2024] [Indexed: 07/23/2024] Open
Abstract
Ferroptosis, a form of regulated cell death, has emerged as a crucial process in diverse pathophysiological states, encompassing cancer, neurodegenerative ailments, and ischemia-reperfusion injury. The glutathione (GSH)-dependent lipid peroxidation pathway, chiefly governed by glutathione peroxidase 4 (GPX4), assumes an essential part in driving ferroptosis. GPX4, as the principal orchestrator of ferroptosis, has garnered significant attention across cancer, cardiovascular, and neuroscience domains over the past decade. Noteworthy investigations have elucidated the indispensable functions of ferroptosis in numerous diseases, including tumorigenesis, wherein robust ferroptosis within cells can impede tumor advancement. Recent research has underscored the complex regulatory role of non-coding RNAs (ncRNAs) in regulating the GSH-GPX4 network, thus influencing cellular susceptibility to ferroptosis. This exhaustive review endeavors to probe into the multifaceted processes by which ncRNAs control the GSH-GPX4 network in ferroptosis. Specifically, we delve into the functions of miRNAs, lncRNAs, and circRNAs in regulating GPX4 expression and impacting cellular susceptibility to ferroptosis. Moreover, we discuss the clinical implications of dysregulated interactions between ncRNAs and GPX4 in several conditions, underscoring their capacity as viable targets for therapeutic intervention. Additionally, the review explores emerging strategies aimed at targeting ncRNAs to modulate the GSH-GPX4 pathway and manipulate ferroptosis for therapeutic advantage. A comprehensive understanding of these intricate regulatory networks furnishes insights into innovative therapeutic avenues for diseases associated with perturbed ferroptosis, thereby laying the groundwork for therapeutic interventions targeting ncRNAs in ferroptosis-related pathological conditions.
Collapse
Affiliation(s)
- Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, 346, United Arab Emirates
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, 346, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
- Graphic Era Hill University, Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, NSW, 2007, Australia
| | - Kamal Dua
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, P.O. Box: 123 Broadway, Ultimo, NSW, 2007, Australia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
28
|
Liu Y, Yang H, Lv G, Duan J, Zhao W, Shi Y, Lei Y. Integration analysis of cis- and trans-regulatory long non-coding RNAs associated with immune-related pathways in non-small cell lung cancer. Biochem Biophys Rep 2024; 40:101832. [PMID: 39539669 PMCID: PMC11558640 DOI: 10.1016/j.bbrep.2024.101832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are importantly involved in the initiation and progression of non-small cell lung cancer (NSCLC). However, the classification and mechanisms of lncRNAs remain largely elusive. Aim Hence, we addressed this through bioinformatics analysis. Methods and results We utilized microarray technology to analyze lncRNAs and mRNAs in twenty paired NSCLC tumor tissues and adjacent normal tissues. Gene set enrichment analysis, Kyoto Encyclopedia of Genes and Genomes, and Gene Ontology were conducted to discern the biological functions of identified differentially expressed transcripts. Additionally, networks of lncRNA-mRNA co-expression, including cis-regulation, lncRNA-transcription factor (TF)-mRNA, trans-regulation, and lncRNA-miRNA-mRNA interactions were explored. Furthermore, the study examined differentially expressed transcripts and their prognostic values in a large RNA-seq dataset of 1016 NSCLC tumors and normal tissues extracted from the Cancer Genome Atlas (TCGA). The analysis revealed 391 lncRNAs and 344 mRNAs with differential expression in NSCLC tumor tissues compared to adjacent normal tissues. Subsequently, 43,557 co-expressed lncRNA-mRNA pairs were identified, including 27 lncRNA-mRNA pairs in cis, 9 lncRNA-TF-mRNA networks, 34 lncRNA-mRNA pairs in trans, and 8701 lncRNA-miRNA-mRNA competing endogenous RNA (ceRNA) networks. Notably, these lncRNAs were found to be involved in immune-related pathways. Six significant transcripts, including NTF4, PTPRD-AS, ITGA11, HID1-AS1, RASGRF2-AS1, and TBX2-AS1, were identified within the ceRNA network and trans-regulation. Conclusion This study brings important insights into the regulatory roles of lncRNAs in NSCLC, providing a fresh perspective on lncRNA research in tumor biology.
Collapse
Affiliation(s)
| | | | - Guoli Lv
- Department of Geriatric Thoracic Surgery, the First Hospital of Kunming Medical University, Kunming, China
| | - Jin Duan
- Department of Geriatric Thoracic Surgery, the First Hospital of Kunming Medical University, Kunming, China
| | - Wei Zhao
- Department of Geriatric Thoracic Surgery, the First Hospital of Kunming Medical University, Kunming, China
| | - Yunfei Shi
- Department of Geriatric Thoracic Surgery, the First Hospital of Kunming Medical University, Kunming, China
| | - Youming Lei
- Department of Geriatric Thoracic Surgery, the First Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
29
|
Fosseprez O, Cuvier O. Uncovering the functions and mechanisms of regulatory elements-associated non-coding RNAs. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195059. [PMID: 39226990 DOI: 10.1016/j.bbagrm.2024.195059] [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: 06/26/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
Over the past decade, regulatory non-coding RNAs (ncRNAs) produced by RNA Pol II have been revealed as meaningful players in various essential cellular functions. In particular, thousands of ncRNAs are produced at transcriptional regulatory elements such as enhancers and promoters, where they may exert multiple functions to regulate proper development, cellular programming, transcription or genomic stability. Here, we review the mechanisms involving these regulatory element-associated ncRNAs, and particularly enhancer RNAs (eRNAs) and PROMoter uPstream Transcripts (PROMPTs). We contextualize the mechanisms described to the processing and degradation of these short lived RNAs. We summarize recent findings explaining how ncRNAs operate locally at promoters and enhancers, or further away, either shortly after their production by RNA Pol II, or through post-transcriptional stabilization. Such discoveries lead to a converging model accounting for how ncRNAs influence cellular fate, by acting on transcription and chromatin structure, which may further involve factors participating to 3D nuclear organization.
Collapse
Affiliation(s)
- Olivier Fosseprez
- Chromatin Dynamics and Cell Proliferation team; Center of Integrative Biology (CBI), Molecular Cellular and Developmental Biology Unit (MCD/UMR5077) Center of Integrative Biology (CBI-CNRS), Université de Toulouse (UPS), F-31000, France.
| | - Olivier Cuvier
- Chromatin Dynamics and Cell Proliferation team; Center of Integrative Biology (CBI), Molecular Cellular and Developmental Biology Unit (MCD/UMR5077) Center of Integrative Biology (CBI-CNRS), Université de Toulouse (UPS), F-31000, France.
| |
Collapse
|
30
|
Zhou J, Zhang L, Wu H, Gao SL, Chen XP, Zhang LF, Zhao CP, Wei BB, Bai Y. Ferroptosis-related lncRNA AL136084.3 is associated with NUPR1 in bladder cancer. Discov Oncol 2024; 15:730. [PMID: 39613992 DOI: 10.1007/s12672-024-01564-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND LncRNAs are critical regulators of bladder cancer (BLCA), and ferroptosis is a newly discovered cell death responsible for mediating apoptosis and tumorigenesis. The present study aims to establish a prognostic signature of differentially expressed ferroptosis-related lncRNAs (DEFRlncRNAs) and explore the DEFRlncRNA associated with NUPR1 in BLCA. METHODS DEFRlncRNAs in BLCA patients were screened using univariate and multivariate Cox and LASSO regression analyses. In vitro experiments were performed to detect the regulatory effects of DEFRlncRNAs on BLCA cells. A prognostic signature of DEFRlncRNAs in BLCA was created and validated. Moreover, we used RNA-binding protein immunoprecipitation (RIP) to evaluate the correlated DEFRlncRNA with NUPR1. RESULTS A prognostic signature involving 18 DEFRlncRNAs in BLCA was created. Overexpression of AL355353.2 or knockdown of AL136084.3 promoted apoptosis in 5637 and T24 cells in vitro. Results from Starbase database estimated that AL136084.3 was positively associated with NUPR1 (R = 0.229, p < 0.001). RIP analysis revealed the reciprocal binding of NUPR1 and AL136084.3 in BLCA. CONCLUSION The identified FRlncRNA pair signature has a good prognostic and clinical predictive value. The ferroptosis-related lncRNA AL136084.3 is correlated with NUPR1 in BLCA.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Hefeng Road 1000, Wuxi, 214000, China
| | - Li Zhang
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Hao Wu
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
- Department of Urology, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Sheng-Lin Gao
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Xiao-Ping Chen
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Hefeng Road 1000, Wuxi, 214000, China
| | - Li-Feng Zhang
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Cui-Ping Zhao
- Department of Geriatrics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, China.
| | - Bing-Bing Wei
- Department of Urology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, China.
| | - Yu Bai
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China.
- Department of Urology, Gonghe County Traditional Chinese Medicine Hospital, 277 South Street of Qinghai Lake, Gonghe County, Hainan Prefecture, 813099, Qinghai, China.
| |
Collapse
|
31
|
de Oliveira JCC, Barbosa EDS, Silva NB, Silva TDC, Matos AGDM, Pinho JD. Non-coding rnas in Turner syndrome: a systematic review. REVISTA PAULISTA DE PEDIATRIA : ORGAO OFICIAL DA SOCIEDADE DE PEDIATRIA DE SAO PAULO 2024; 43:e2024029. [PMID: 39630788 PMCID: PMC11606598 DOI: 10.1590/1984-0462/2025/43/2024029] [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: 02/22/2024] [Accepted: 08/24/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE The aim of this study was to summarize the main findings of non-coding RNA (ncRNAs) in Turner syndrome (TS), correlating these biomolecules with the clinical manifestations in affected patients. DATA SOURCE Searches were conducted in the databases of the United States National Library of Medicine (PubMed), Scientific Electronic Library Online (SciELO), and ScienceDirect, covering original English articles published from 2014 to 2023. Descriptors used included "lncRNAs and Turner Syndrome," "miRNAs and Turner Syndrome," and "circRNAs and Turner Syndrome." The studies that were included addressed the role of ncRNAs in the clinical characteristics of patients with TS. Exclusion criteria comprised texts in abstracts, reports, reviews, and monographs. DATA SYNTHESIS We identified 147 studies, of which seven were included. In the analysis of microRNAs, miR-486-5p and miR-320a stood out, being associated with ovarian development; miR-126-3p and miR-126-5p were related to greater aortic stiffness. Regarding long non-coding RNAs, the downregulation of XIST indicated dysfunctions in X chromosome inactivation. Concerning circular RNAs, circPPP2R3B, circCSF2RA, and circPCTN were related to immunological functions, while circ_0090421, circ_0090392, and circ_0089945 were linked to cardiac development. CONCLUSIONS The data from these studies demonstrate that these biomolecules play crucial roles in processes related to specific characteristics observed in TS patients. Besides being suggested as potential biomarkers, they may be useful in clinical practice.
Collapse
|
32
|
Nadhan R, Isidoro C, Song YS, Dhanasekaran DN. LncRNAs and the cancer epigenome: Mechanisms and therapeutic potential. Cancer Lett 2024; 605:217297. [PMID: 39424260 DOI: 10.1016/j.canlet.2024.217297] [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: 08/23/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators of epigenome, modulating gene expression through DNA methylation, histone modification, and/or chromosome remodeling. Dysregulated lncRNAs act as oncogenes or tumor suppressors, driving tumor progression by shaping the cancer epigenome. By interacting with the writers, readers, and erasers of the epigenetic script, lncRNAs induce epigenetic modifications that bring about changes in cancer cell proliferation, apoptosis, epithelial-mesenchymal transition, migration, invasion, metastasis, cancer stemness and chemoresistance. This review analyzes and discusses the multifaceted role of lncRNAs in cancer pathobiology, from cancer genesis and progression through metastasis and therapy resistance. It also explores the therapeutic potential of targeting lncRNAs through innovative diagnostic, prognostic, and therapeutic strategies. Understanding the dynamic interplay between lncRNAs and epigenome is crucial for developing personalized therapeutic strategies, offering new avenues for precision cancer medicine.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 151-921, South Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
33
|
Chen J, Gao Y, Zhong J, Wu X, Leng Z, Liu M, Wang Y, Wang Y, Yang X, Huang N, Xiao F, Zhang M, Liu X, Zhang N. Lnc-H19-derived protein shapes the immunosuppressive microenvironment of glioblastoma. Cell Rep Med 2024; 5:101806. [PMID: 39481387 PMCID: PMC11604490 DOI: 10.1016/j.xcrm.2024.101806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/02/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
The immunosuppressive tumor microenvironment (TME) is a prominent feature of glioblastoma (GBM), the most lethal primary brain cancer resistant to current immunotherapies. The mechanisms underlying GBM-TME remain to be explored. We report that long non-coding RNA (LncRNA) H19 encodes an immune-related protein called H19-IRP. Functionally separated from H19 RNA, H19-IRP promotes GBM immunosuppression by binding to the CCL2 and Galectin-9 promoters and activating their transcription, thereby recruiting myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), leading to T cell exhaustion and an immunosuppressive GBM-TME. H19-IRP, overexpressed in clinical GBM samples, acts as a tumor-associated antigen (TAA) presented by major histocompatibility complex class I (MHC-I). A circular RNA vaccine targeting H19-IRP (circH19-vac) triggers a potent cytotoxic T cell response against GBM and inhibits GBM growth. Our results highlight the unrevealed function of H19-IRP in creating immunosuppressive GBM-TME by recruiting MDSCs and TAMs, supporting the idea of targeting H19-IRP with cancer vaccine for GBM treatment.
Collapse
MESH Headings
- Glioblastoma/immunology
- Glioblastoma/pathology
- Glioblastoma/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/immunology
- Tumor Microenvironment/immunology
- Humans
- Animals
- Galectins/metabolism
- Galectins/genetics
- Galectins/immunology
- Cell Line, Tumor
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- Brain Neoplasms/immunology
- Brain Neoplasms/pathology
- Brain Neoplasms/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL2/immunology
- Chemokine CCL2/genetics
- Mice
- Gene Expression Regulation, Neoplastic
- Macrophages/immunology
- Macrophages/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Mice, Inbred C57BL
- Cancer Vaccines/immunology
- Promoter Regions, Genetic/genetics
Collapse
Affiliation(s)
- Junju Chen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Yixin Gao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Jian Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Xujia Wu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Zhaojie Leng
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Ming Liu
- Guangzhou Geneseed Biotech. Co., Ltd, Guangzhou, Guangdong Province, China
| | - Yesheng Wang
- Guangzhou Geneseed Biotech. Co., Ltd, Guangzhou, Guangdong Province, China
| | - Yuan Wang
- Guangzhou Geneseed Biotech. Co., Ltd, Guangzhou, Guangdong Province, China
| | - Xuesong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Nunu Huang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China
| | - Feizhe Xiao
- Department of Scientific Research Section, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Maolei Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China.
| | - Xuesong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China.
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
34
|
Zhang Y, Shen Z, Han X, Wu Y, Huang T. Long non-coding RNA AC105118.1 affects glycolysis to facilitate oxaliplatin resistance in colorectal cancer cells by modulating the miR-378a-3p/KIF26B axis. Int J Biochem Cell Biol 2024; 177:106692. [PMID: 39536859 DOI: 10.1016/j.biocel.2024.106692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Oxaliplatin is a first-line chemotherapy drug for colorectal cancer (CRC), but many patients eventually lose treatment efficacy due to acquired resistance. AC105118.1 is a long non-coding RNA with unknown biological function. This research attempts to probe into the molecular regulatory mechanism of AC105118.1 in CRC oxaliplatin resistance. METHODS The expression level of AC105118.1 in CRC tissues and cells was measured based on The Cancer Genome Atlas (TCGA) data and quantitative reverse transcription polymerase chain reaction (qRT-PCR). We utilized dual-luciferase assay and RNA immunoprecipitation to analyze the interaction between AC105118.1, miR-378a-3p, and their downstream target KIF26B. CCK-8, colony formation assay, and flow cytometry were employed to assess the half inhibitory concentration (IC50), cell proliferation, and apoptosis rate of HCT116/L-OHP cells treated with oxaliplatin. The glycolysis evaluation was completed by measuring the extracellular acidification rate (ECAR), glucose consumption, lactate production, and glycolysis-related proteins (HK2, GLUT1, and LDHA). TUNEL staining was used to detect the level of apoptosis. RESULTS AC105118.1 was specifically upregulated in CRC tissues and cells. AC105118.1 indirectly facilitated the expression of miRNA target gene KIF26B by sequestering miR-378a-3p. In HCT116/L-OHP cells, transfection with si-AC105118.1 resulted in a decrease in glycolysis level, a lower maximum IC50 required for oxaliplatin-treated cells, inhibited cell proliferation, and an increase in apoptosis rate. All of these effects were alleviated when simultaneously transfecting miR-378a-3p inhibitor or oe-KIF26B. Knockdown of AC105118.1 significantly inhibited oxaliplatin resistance to CRC in mice. CONCLUSION AC105118.1 facilitates glycolysis and increases CRC cells' resistance to oxaliplatin by targeting the miR-378a-3p/KIF26B axis. The present work shed new insights into the function and mechanism of AC105118.1 in molecular function and suggested that the AC105118.1/miR-378a-3p/KIF26B axis is a promising target for intervening CRC oxaliplatin resistance.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Surgery, AnYang Tumor Hospital, Anyang 455000, China
| | - Zhiling Shen
- Department of Surgery, AnYang Tumor Hospital, Anyang 455000, China
| | - Xiaodong Han
- Department of Surgery, AnYang Tumor Hospital, Anyang 455000, China
| | - Yachao Wu
- Department of Surgery, AnYang Tumor Hospital, Anyang 455000, China
| | - Tianchen Huang
- Department of Surgery, AnYang Tumor Hospital, Anyang 455000, China.
| |
Collapse
|
35
|
Chang J, Zhang L, Li Z, Qian C, Du J. Exosomal non-coding RNAs (ncRNAs) as potential biomarkers in tumor early diagnosis. Biochim Biophys Acta Rev Cancer 2024; 1879:189188. [PMID: 39313040 DOI: 10.1016/j.bbcan.2024.189188] [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: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Exosomes, extracellular vesicles carrying a cargo rich in various non-coding RNAs (ncRNAs), have emerged as crucial mediators of intercellular communication. Their stability, abundance, and specificity make exosomal ncRNAs promising candidates for biomarker discovery. The discovery of exosomal ncRNAs has unveiled a novel avenue for the exploration of biomarkers in tumor early diagnosis. This review consolidates current knowledge on the role of exosomal ncRNAs as potential biomarkers in the early detection of various tumors. We provide an overview of recent studies demonstrating the diagnostic potential of exosomal ncRNAs across multiple cancer types, highlighting their sensitivity, specificity, and feasibility for early detection. This review underscores the potential of exosomal ncRNAs as non-invasive biomarkers for early tumor diagnosis, paving the way for improved clinical outcomes through timely intervention and personalized management strategies.
Collapse
Affiliation(s)
- Jingyue Chang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Lingquan Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Zeting Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China
| | - Chungen Qian
- Department of Reagent Research and Development, Shenzhen YHLO Biotech Co., Ltd., Shenzhen 518172, Guangdong, China.
| | - Juan Du
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong, China.
| |
Collapse
|
36
|
Yang Y, Cheng H. Emerging Roles of ncRNAs in Type 2 Diabetes Mellitus: From Mechanisms to Drug Discovery. Biomolecules 2024; 14:1364. [PMID: 39595541 PMCID: PMC11592034 DOI: 10.3390/biom14111364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM), a high-incidence chronic metabolic disorder, has emerged as a global health issue, where most patients need lifelong medication. Gaining insights into molecular mechanisms involved in T2DM development is expected to provide novel strategies for clinical prevention and treatment. Growing evidence validates that non-coding RNAs (ncRNAs) including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) function as crucial regulators in multiple biological processes of T2DM, inspiring various potential targets and drug candidates. In this review, we summarize the current understanding of ncRNA roles in T2DM and discuss the potential use of ncRNAs as targets and active molecules for drug discovery.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
37
|
Oguntoyinbo IO, Goyal R. The Role of Long Intergenic Noncoding RNA in Fetal Development. Int J Mol Sci 2024; 25:11453. [PMID: 39519006 PMCID: PMC11546696 DOI: 10.3390/ijms252111453] [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: 09/25/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The role of long intergenic noncoding RNAs (lincRNAs) in fetal development has emerged as a significant area of study, challenging the traditional protein-centric view of gene expression. While messenger RNAs (mRNAs) have long been recognized for their role in encoding proteins, recent advances have illuminated the critical functions of lincRNAs in various biological processes. Initially identified through high-throughput sequencing technologies, lincRNAs are transcribed from intergenic regions between protein-coding genes and exhibit unique regulatory functions. Unlike mRNAs, lincRNAs are involved in complex interactions with chromatin and chromatin-modifying complexes, influencing gene expression and chromatin structure. LincRNAs are pivotal in regulating tissue-specific development and embryogenesis. For example, they are crucial for proper cardiac, neural, and reproductive system development, with specific lincRNAs being associated with organogenesis and differentiation processes. Their roles in embryonic development include regulating transcription factors and modulating chromatin states, which are essential for maintaining developmental programs and cellular identity. Studies using RNA sequencing and genetic knockout models have highlighted the importance of lincRNAs in processes such as cell differentiation, tissue patterning, and organ development. Despite their functional significance, the comprehensive annotation and understanding of lincRNAs remain limited. Ongoing research aims to elucidate their mechanisms of action and potential applications in disease diagnostics and therapeutics. This review summarizes current knowledge on the functional roles of lincRNAs in fetal development, emphasizing their contributions to tissue-specific gene regulation and developmental processes.
Collapse
Affiliation(s)
- Ifetoluwani Oluwadunsin Oguntoyinbo
- School of Animal and Comparative Biomedical Sciences, College of Agriculture, Life & Environmental Sciences, University of Arizona, Tucson, AZ 85721, USA;
| | - Ravi Goyal
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
38
|
Wang C, Liang X, Jia Z, Huang Y, Chen H, Wei H, Huang Y, Huang X, Fang X. Changes in the expression profile of serum lncRNAs in pregnant women with high hepatitis B viral load during antiviral and non-antiviral treatment. BMC Pregnancy Childbirth 2024; 24:696. [PMID: 39449132 PMCID: PMC11515369 DOI: 10.1186/s12884-024-06907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE This research analyzes the potential of long non-coding RNAs (lncRNAs) as markers in determining the necessity of antiviral treatment in pregnant women by examining alterations in the expression profile of serum lncRNAs in pregnant women with elevated hepatitis B viral load (HBVL) under antiviral and non-antiviral treatment regimens between the second trimester and delivery. METHODS Serum was obtained from 6 s-trimester pregnant women with high HBVL and no intrauterine infection. Then, 3 of these women were randomly selected for antiviral treatment, with the remaining 3 women undergoing non-antiviral treatment as control. Serum samples were again collected from these 6 women before delivery. The expression profile of lncRNAs was analyzed with microarray technology, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. The axes of hub lncRNA-miRNA-mRNA were identified based on the competing endogenous RNA (ceRNA) network. RESULTS The expression profile of serum lncRNAs in pregnant women with high HBVL changed significantly from the second trimester of pregnancy until delivery under antiviral or non-antiviral treatment. The Venn diagram was utilized to screen out the jointly up-regulated and down-regulated lncRNAs in the serum of pregnant women under antiviral and non-antiviral treatment before delivery. Additionally, the KEGG pathway enrichment analysis results showed that lncRNAs might mediate the Hippo pathway in HBV infection. Based on the ceRNA network, 3 hub lncRNAs (CATG00000076041.1, LINC01310, and G014655) were found to potentially regulate the key gene TP73 in the Hippo pathway. CONCLUSION In this study, we retrieved co-differentially expressed lncRNAs in pregnant women with high HBVL under antiviral or non-antiviral treatment, which may be used as markers for evaluating whether pregnant women with high HBVL may be free of antiviral treatment. This study may provide a basis for preventing potential adverse effects of antiviral treatment on maternal and fetal health.
Collapse
Affiliation(s)
- Cuimin Wang
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China.
- Department of Obstetrics and Gynecology, People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, No. 6, Taoyuan Road, Qingxiu District, Nanning, China.
| | - Xuxia Liang
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China.
- Department of Obstetrics and Gynecology, People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, No. 6, Taoyuan Road, Qingxiu District, Nanning, China.
| | - Zaiming Jia
- Department of Obstetrics and Gynecology, Youjiang Medical College for Nationalities, Baise City, China
| | - Yuting Huang
- Department of Obstetrics and Gynecology, Youjiang Medical College for Nationalities, Baise City, China
| | - Hui Chen
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Haitang Wei
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yin Huang
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Xizhen Huang
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Xiang Fang
- Department of Obstetrics and Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| |
Collapse
|
39
|
Sun Z, Tang J, You T, Zhang B, Liu Y, Liu J. lncRNA OIP5-AS1 promotes mitophagy to alleviate osteoarthritis by upregulating PPAR-γ to activate the AMPK/Akt/mTOR pathway. Mod Rheumatol 2024; 34:1265-1276. [PMID: 38441253 DOI: 10.1093/mr/roae015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/21/2024] [Indexed: 10/17/2024]
Abstract
OBJECTIVES Osteoarthritis (OA) is the most common chronic joint degenerative disease. Herein, we investigated long non-coding RNA Opa-interacting protein 5-antisense transcript 1's (OIP5-AS1) in regulating mitophagy during OA. METHODS RNA immunoprecipitation and RNA pull-down verified the relationship between molecules. Cell counting kit-8 detected cell viability. Enzyme-linked immunosorbent assay evaluated inflammatory cytokines secretion. Flow cytometry measured the contents of reactive oxygen species (ROS) and calcium. Immunofluorescence staining analysed TOMM20 and LC3B levels. JC-1 staining was adopted to measure mitochondrial membrane potential. The changes of mitophagy were analysed by transmission electron microscopy. RESULTS Lipopolysaccharide (LPS) treatment contributed to the decrease of chondrocyte viability, and calcium level and inhibited mitochondrial membrane potential, while elevating the secretion of inflammatory factors, ROS, and TOMM20 expression. OIP5-AS1 overexpression inhibited LPS-induced chondrocyte injury and activated mitophagy. OIP5-AS1 upregulated the peroxisome proliferator-activated receptor-γ (PPAR-γ) mRNA level to regulate adenosine monophosphate-activated protein kinase (AMPK)/v-akt murine thymoma viral oncogene homolog (Akt)/mammalian target of rapamycin (mTOR) signalling by interacting with FUS. PPAR-γ overexpression alleviated LPS-induced chondrocyte injury by activating AMPK/Akt/mTOR signalling. PPAR-γ knockdown reversed the promotion of OIP5-AS1 upregulation on mitophagy. CONCLUSIONS OIP5-AS1 promotes PPAR-γ expression to activate the AMPK/Akt/mTOR signalling, thereby enhancing mitophagy and alleviating OA progression.
Collapse
Affiliation(s)
- Zhilu Sun
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| | - Jie Tang
- The First Affiliated Hospital, Department of Pain, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| | - Ting You
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| | - Bihong Zhang
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| | - Yu Liu
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| | - Jing Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang, Hunan Province, P.R. China
| |
Collapse
|
40
|
Shou F, Li G, Morshedi M. Long Non-coding RNA ANRIL and Its Role in the Development of Age-Related Diseases. Mol Neurobiol 2024; 61:7919-7929. [PMID: 38443729 DOI: 10.1007/s12035-024-04074-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/23/2024] [Indexed: 03/07/2024]
Abstract
ANRIL is known as a lncRNA that has many linear and circular isoforms and its polymorphisms are observed to be associated with the pathogenesis of many diseases including age-related diseases. Age-related diseases including atherosclerosis, ischemic heart disease, and Alzheimer's and Parkinson's disease are the most common cause of mortality in both developed and undeveloped countries and that is why a better understanding of their pathogenesis and underlying mechanisms is necessary for controlling their healthcare burden.In this review, we aim to gather the data of researches which have investigated the role of ANRIL in aging and its related diseases. The conclusions of this paper might give a new insight for decreasing the mortality rate of these diseases.
Collapse
Affiliation(s)
- Feiyan Shou
- Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Gang Li
- Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.
| | - Mohammadamin Morshedi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
41
|
Xu B, Ye X, Wen Z, Chen S, Wang J. Epigenetic regulation of megakaryopoiesis and platelet formation. Haematologica 2024; 109:3125-3137. [PMID: 38867584 PMCID: PMC11443398 DOI: 10.3324/haematol.2023.284951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Indexed: 06/14/2024] Open
Abstract
Platelets, produced by megakaryocytes, play unique roles in physiological processes, such as hemostasis, coagulation, and immune regulation, while also contributing to various clinical diseases. During megakaryocyte differentiation, the morphology and function of cells undergo significant changes due to the programmed expression of a series of genes. Epigenetic changes modify gene expression without altering the DNA base sequence, effectively affecting the inner workings of the cell at different stages of growth, proliferation, differentiation, and apoptosis. These modifications also play important roles in megakaryocyte development and platelet biogenesis. However, the specific mechanisms underlying epigenetic processes and the vast epigenetic regulatory network formed by their interactions remain unclear. In this review, we systematically summarize the key roles played by epigenetics in megakaryocyte development and platelet formation, including DNA methylation, histone modification, and non-coding RNA regulation. We expect our review to provide a deeper understanding of the biological processes underlying megakaryocyte development and platelet formation and to inform the development of new clinical interventions aimed at addressing platelet-related diseases and improving patients' prognoses.
Collapse
Affiliation(s)
- Baichuan Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038
| | - Xianpeng Ye
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038
| | - Zhaoyang Wen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038
| | - Shilei Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038.
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038.
| |
Collapse
|
42
|
Song Y, Wen H, Zhai X, Jia L, Li L. Functional Bidirectionality of ERV-Derived Long Non-Coding RNAs in Humans. Int J Mol Sci 2024; 25:10481. [PMID: 39408810 PMCID: PMC11476766 DOI: 10.3390/ijms251910481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Human endogenous retroviruses (HERVs) are widely recognized as the result of exogenous retroviruses infecting the ancestral germline, stabilizing integration and vertical transmission during human genetic evolution. To date, endogenous retroviruses (ERVs) appear to have been selected for human physiological functions with the loss of retrotransposable capabilities. ERV elements were previously regarded as junk DNA for a long time. Since then, the aberrant activation and expression of ERVs have been observed in the development of many kinds of human diseases, and their role has been explored in a variety of human disorders such as cancer. The results show that specific ERV elements play respective crucial roles. Among them, long non-coding RNAs (lncRNAs) transcribed from specific long-terminal repeat regions of ERVs are often key factors. lncRNAs are over 200 nucleotides in size and typically bind to DNA, RNA, and proteins to perform biological functions. Dysregulated lncRNAs have been implicated in a variety of diseases. In particular, studies have shown that the aberrant expression of some ERV-derived lncRNAs has a tumor-suppressive or oncogenic effect, displaying significant functional bidirectionality. Therefore, theses lncRNAs have a promising future as novel biomarkers and therapeutic targets to explore the concise relationship between ERVs and cancers. In this review, we first summarize the role of ERV-derived lncRNAs in physiological regulation, mainly including immunomodulation, the maintenance of pluripotency, and erythropoiesis. In addition, pathological regulation examples of their aberrant activation and expression leading to carcinogenesis are highlighted, and specific mechanisms of occurrence are discussed.
Collapse
Affiliation(s)
- Yanmei Song
- Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Shandong University, Key Laboratory for the Prevention and Control of Emerging Infectious Diseases and Biosafety, Jinan 250012, China; (Y.S.); (H.W.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100850, China;
| | - Hongling Wen
- Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Shandong University, Key Laboratory for the Prevention and Control of Emerging Infectious Diseases and Biosafety, Jinan 250012, China; (Y.S.); (H.W.)
| | - Xiuli Zhai
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100850, China;
- Department of Microbiology, School of Basic Medicine, Anhui Medical University, Hefei 230000, China
| | - Lei Jia
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100850, China;
| | - Lin Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100850, China;
| |
Collapse
|
43
|
Arshi A, Mahmoudi E, Raeisi F, Dehghan Tezerjani M, Bahramian E, Ahmed Y, Peng C. Exploring potential roles of long non-coding RNAs in cancer immunotherapy: a comprehensive review. Front Immunol 2024; 15:1446937. [PMID: 39257589 PMCID: PMC11384988 DOI: 10.3389/fimmu.2024.1446937] [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: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer treatment has long been fraught with challenges, including drug resistance, metastasis, and recurrence, making it one of the most difficult diseases to treat effectively. Traditional therapeutic approaches often fall short due to their inability to target cancer stem cells and the complex genetic and epigenetic landscape of tumors. In recent years, cancer immunotherapy has revolutionized the field, offering new hope and viable alternatives to conventional treatments. A particularly promising area of research focuses on non-coding RNAs (ncRNAs), especially long non-coding RNAs (lncRNAs), and their role in cancer resistance and the modulation of signaling pathways. To address these challenges, we performed a comprehensive review of recent studies on lncRNAs and their impact on cancer immunotherapy. Our review highlights the crucial roles that lncRNAs play in affecting both innate and adaptive immunity, thereby influencing the outcomes of cancer treatments. Key observations from our review indicate that lncRNAs can modify the tumor immune microenvironment, enhance immune cell infiltration, and regulate cytokine production, all of which contribute to tumor growth and resistance to therapies. These insights suggest that lncRNAs could serve as potential targets for precision medicine, opening up new avenues for developing more effective cancer immunotherapies. By compiling recent research on lncRNAs across various cancers, this review aims to shed light on their mechanisms within the tumor immune microenvironment.
Collapse
Affiliation(s)
- Asghar Arshi
- Department of Biology, York University, Toronto, ON, Canada
| | - Esmaeil Mahmoudi
- Young Researchers and Elite Club, Islamic Azad University, Shahrekord, Iran
| | | | - Masoud Dehghan Tezerjani
- Department of bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Bahramian
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Yeasin Ahmed
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
44
|
Liu Y, Gao J, Xu Q, Wang X, Zhong W, Wu F, Lin X, Zhang Q, Ye Q. Long non-coding RNA NEAT1 exacerbates NLRP3-mediated pyroptosis in allergic rhinitis through regulating the PTBP1/FOXP1 cascade. Int Immunopharmacol 2024; 137:112337. [PMID: 38861915 DOI: 10.1016/j.intimp.2024.112337] [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: 11/15/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Allergic Rhinitis (AR) is a prevalent chronic non-infectious inflammation affecting the nasal mucosa. NLRP3-mediated pyroptosis of epithelial cells plays a pivotal role in AR pathogenesis. Herein, we evaluated the impact of the long non-coding RNA nuclear paraspeckle assembly transcript 1 (lncRNA NEAT1) on NLR family pyrin domain containing 3 (NLRP3)-mediated pyroptosis in AR. METHODS Nasal inflammation levels in ovalbumin (OVA)-induced AR mice were assessed using HE staining, and NLRP3 expression was evaluated through immunohistochemistry. ELISA was utilized to detect OVA-specific IgE, IL-6, IL-5, and inflammatory cytokines (IL-1β, IL-18). Human nasal epithelial cells (HNEpCs) stimulated with IL4/IL13 were used to analyze the mRNA and protein levels of associated genes utilizing RT-qPCR and western blot, respectively. Cell viability and pyroptosis were assessed by CCK-8 and flow cytometry. The targeting relationship between NEAT1, PTBP1 and FOXP1 were analyzed by RIP and RNA pull down assays. FISH and IF analysis were performed to assess the co-localization of NEAT1 and PTBP1. RESULTS In both the AR mouse and cellular models, increased levels of NEAT1, PTBP1 and FOXP1 were observed. AR mice exhibited elevated inflammatory infiltration and pyroptosis, evidenced by enhanced expressions of OVA-specific IgE, IL-6, and IL-5, NLRP3, Cleaved-caspase 1, GSDMD-N, IL-1β and IL-18. Functional assays revealed that knockdown of PTBP1 or NEAT1 inhibited pyroptosis while promoting the proliferation of IL4/IL13-treated HNEpCs. Mechanistically, NEAT1 directly interacted with PTBP1, thereby maintaining FOXP1 mRNA stability. Rescue assays demonstrated that FOXP1 upregulation reversed the inhibitory effects of silencing NEAT1 or PTBP1 on IL4/IL13-stimulated pyroptosis activation in HNEpCs. CONCLUSION NEAT1 acts as a RNA scaffold for PTBP1, activating the PTBP1/FOXP1 signaling cascade, subsequently triggering NLRP3-mediated pyroptosis in HNEpCs, and ultimately promoting AR progression. These findings highlight some new insights into the pathogenesis of AR.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Pyroptosis
- Rhinitis, Allergic/immunology
- Rhinitis, Allergic/pathology
- Rhinitis, Allergic/genetics
- Rhinitis, Allergic/metabolism
- Humans
- Mice
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Nasal Mucosa/immunology
- Nasal Mucosa/pathology
- Nasal Mucosa/metabolism
- Mice, Inbred BALB C
- Ovalbumin/immunology
- Heterogeneous-Nuclear Ribonucleoproteins/metabolism
- Heterogeneous-Nuclear Ribonucleoproteins/genetics
- Signal Transduction
- Disease Models, Animal
- Female
- Cytokines/metabolism
Collapse
Affiliation(s)
- Yunliang Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China; Department of Otolaryngology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian Province, PR China; Department of Otolaryngology, Fujian Children's Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Jing Gao
- Health Medicine Department, The 900th Hospital of Chinese PLA Joint Logistics Support Force, Fuzhou 350025, Fujian Province, PR China
| | - Qingqing Xu
- Department of Otolaryngology, Fujian Children's Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Xiaoyan Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China; Department of Otorhinolaryngology-Head & Neck Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - Wenhui Zhong
- Department of Clinical Laboratory, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - Fengfang Wu
- Department of Otorhinolaryngology-Head & Neck Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, Fujian Province, PR China
| | - Xianghang Lin
- Department of Otolaryngology, Fujian Children's Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Qiuyun Zhang
- Department of Otolaryngology, Fujian Children's Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Qing Ye
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China; Department of Otorhinolaryngology-Head & Neck Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China.
| |
Collapse
|
45
|
Sweef O, Mahfouz R, Taşcıoğlu T, Albowaidey A, Abdelmonem M, Asfar M, Zaabout E, Corcino YL, Thomas V, Choi ES, Furuta S. Decoding LncRNA in COPD: Unveiling Prognostic and Diagnostic Power and Their Driving Role in Lung Cancer Progression. Int J Mol Sci 2024; 25:9001. [PMID: 39201688 PMCID: PMC11354875 DOI: 10.3390/ijms25169001] [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: 07/01/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer represent formidable challenges in global health, characterized by intricate pathophysiological mechanisms and multifaceted disease progression. This comprehensive review integrates insights from diverse perspectives to elucidate the intricate roles of long non-coding RNAs (lncRNAs) in the pathogenesis of COPD and lung cancer, focusing on their diagnostic, prognostic, and therapeutic implications. In the context of COPD, dysregulated lncRNAs, such as NEAT1, TUG1, MALAT1, HOTAIR, and GAS5, emerge as pivotal regulators of genes involved in the disease pathogenesis and progression. Their identification, profiling, and correlation with the disease severity present promising avenues for prognostic and diagnostic applications, thereby shaping personalized disease interventions. These lncRNAs are also implicated in lung cancer, underscoring their multifaceted roles and therapeutic potential across both diseases. In the domain of lung cancer, lncRNAs play intricate modulatory roles in disease progression, offering avenues for innovative therapeutic approaches and prognostic indicators. LncRNA-mediated immune responses have been shown to drive lung cancer progression by modulating the tumor microenvironment, influencing immune cell infiltration, and altering cytokine production. Their dysregulation significantly contributes to tumor growth, metastasis, and chemo-resistance, thereby emphasizing their significance as therapeutic targets and prognostic markers. This review summarizes the transformative potential of lncRNA-based diagnostics and therapeutics for COPD and lung cancer, offering valuable insights into future research directions for clinical translation and therapeutic development.
Collapse
Affiliation(s)
- Osama Sweef
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Reda Mahfouz
- Core Laboratory, University Hospital Cleveland Medical Center, Department of Pathology, School of Medicine, Case Western Reserve University, 1100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Clinical Pathology, Faculty of Medicine, Menofia University, Shebin-Elkom 32511, Egypt
| | - Tülin Taşcıoğlu
- Department of Molecular Biology and Genetics, Demiroglu Bilim University, Esentepe Central Campus, Besiktas, 34394 Istanbul, Turkey
| | - Ali Albowaidey
- The Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Mohamed Abdelmonem
- Department of Pathology, Transfusion Medicine Service, Stanford Healthcare, Stanford, CA 94305, USA
| | - Malek Asfar
- Department of Pathology, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Elsayed Zaabout
- Department of Therapeutics & Pharmacology, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
| | - Yalitza Lopez Corcino
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Venetia Thomas
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Eun-Seok Choi
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Saori Furuta
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| |
Collapse
|
46
|
Hernández-Contreras KA, Martínez-Díaz JA, Hernández-Aguilar ME, Herrera-Covarrubias D, Rojas-Durán F, Chi-Castañeda LD, García-Hernández LI, Aranda-Abreu GE. Alterations of mRNAs and Non-coding RNAs Associated with Neuroinflammation in Alzheimer's Disease. Mol Neurobiol 2024; 61:5826-5840. [PMID: 38236345 DOI: 10.1007/s12035-023-03908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Alzheimer's disease is a neurodegenerative pathology whose pathognomonic hallmarks are increased generation of β-amyloid (Aβ) peptide, production of hyperphosphorylated (pTau), and neuroinflammation. The last is an alteration closely related to the progression of AD and although it is present in multiple neurodegenerative diseases, the pathophysiological events that characterize neuroinflammatory processes vary depending on the disease. In this article, we focus on mRNA and non-coding RNA alterations as part of the pathophysiological events characteristic of neuroinflammation in AD and the influence of these alterations on the course of the disease through interaction with multiple RNAs related to the generation of Aβ, pTau, and neuroinflammation itself.
Collapse
Affiliation(s)
- Karla Aketzalli Hernández-Contreras
- Doctorado en Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Jorge Antonio Martínez-Díaz
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - María Elena Hernández-Aguilar
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Deissy Herrera-Covarrubias
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Lizbeth Donají Chi-Castañeda
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Luis Isauro García-Hernández
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México
| | - Gonzalo Emiliano Aranda-Abreu
- Instituto de Investigaciones Cerebrales/Universidad Veracruzana, Av. Luis Castelazo Ayala S/N, Carr. Xalapa-Veracruz, Km 3.5, C.P. 91190, Xalapa, Veracruz, México.
| |
Collapse
|
47
|
Kafida M, Karela M, Giakountis A. RNA-Independent Regulatory Functions of lncRNA in Complex Disease. Cancers (Basel) 2024; 16:2728. [PMID: 39123456 PMCID: PMC11311644 DOI: 10.3390/cancers16152728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
During the metagenomics era, high-throughput sequencing efforts both in mice and humans indicate that non-coding RNAs (ncRNAs) constitute a significant fraction of the transcribed genome. During the past decades, the regulatory role of these non-coding transcripts along with their interactions with other molecules have been extensively characterized. However, the study of long non-coding RNAs (lncRNAs), an ncRNA regulatory class with transcript lengths that exceed 200 nucleotides, revealed that certain non-coding transcripts are transcriptional "by-products", while their loci exert their downstream regulatory functions through RNA-independent mechanisms. Such mechanisms include, but are not limited to, chromatin interactions and complex promoter-enhancer competition schemes that involve the underlying ncRNA locus with or without its nascent transcription, mediating significant or even exclusive roles in the regulation of downstream target genes in mammals. Interestingly, such RNA-independent mechanisms often drive pathological manifestations, including oncogenesis. In this review, we summarize selective examples of lncRNAs that regulate target genes independently of their produced transcripts.
Collapse
Affiliation(s)
| | | | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|
48
|
Chaudhary U, Banerjee S. Decoding the Non-coding: Tools and Databases Unveiling the Hidden World of "Junk" RNAs for Innovative Therapeutic Exploration. ACS Pharmacol Transl Sci 2024; 7:1901-1915. [PMID: 39022352 PMCID: PMC11249652 DOI: 10.1021/acsptsci.3c00388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024]
Abstract
Non-coding RNAs are pivotal regulators of gene and protein expression, exerting crucial influences on diverse biological processes. Their dysregulation is frequently implicated in the onset and progression of diseases, notably cancer. A profound comprehension of the intricate mechanisms governing ncRNAs is imperative for devising innovative therapeutic interventions against these debilitating conditions. Significantly, nearly 80% of our genome comprises ncRNAs, underscoring their centrality in cellular processes. The elucidation of ncRNA functions is pivotal for grasping the complexities of gene regulation and its implications for human health. Modern genome sequencing techniques yield vast datasets, stored in specialized databases. To harness this wealth of information and to understand the crosstalk of non-coding RNAs, knowledge of available databases is required, and many new sophisticated computational tools have emerged. These tools play a pivotal role in the identification, prediction, and annotation of ncRNAs, thereby facilitating their experimental validation. This Review succinctly outlines the current understanding of ncRNAs, emphasizing their involvement in disease development. It also highlights the databases and tools instrumental in classifying, annotating, and evaluating ncRNAs. By extracting meaningful biological insights from seemingly "junk" data, these tools empower scientists to unravel the intricate roles of ncRNAs in shaping human health.
Collapse
Affiliation(s)
- Uma Chaudhary
- Department of Biotechnology,
School of Biosciences and Technology, Vellore
Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Satarupa Banerjee
- Department of Biotechnology,
School of Biosciences and Technology, Vellore
Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| |
Collapse
|
49
|
Zhang L, Yu F, Zhang Y, Li P. Implications of lncRNAs in Helicobacter pylori-associated gastrointestinal cancers: underlying mechanisms and future perspectives. Front Cell Infect Microbiol 2024; 14:1392129. [PMID: 39035354 PMCID: PMC11257847 DOI: 10.3389/fcimb.2024.1392129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a harmful bacterium that is difficult to conveniently diagnose and effectively eradicate. Chronic H. pylori infection increases the risk of gastrointestinal diseases, even cancers. Despite the known findings, more underlying mechanisms are to be deeply explored to facilitate the development of novel prevention and treatment strategies of H. pylori infection. Long noncoding RNAs (lncRNAs) are RNAs with more than 200 nucleotides. They may be implicated in cell proliferation, inflammation and many other signaling pathways of gastrointestinal cancer progression. The dynamic expression of lncRNAs indicates their potential to be diagnostic or prognostic biomarkers. In this paper, we comprehensively summarize the processes of H. pylori infection and the treatment methods, review the known findings of lncRNA classification and functional mechanisms, elucidate the roles of lncRNAs in H. pylori-related gastrointestinal cancer, and discuss the clinical perspectives of lncRNAs.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | | | | | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
50
|
Sivagurunathan N, Rahamathulla MP, Al-Dossary H, Calivarathan L. Emerging Role of Long Noncoding RNAs in Regulating Inflammasome-Mediated Neurodegeneration in Parkinson's Disease. Mol Neurobiol 2024; 61:4619-4632. [PMID: 38105409 DOI: 10.1007/s12035-023-03809-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
Parkinson's disease (PD) is one of the complex neurodegenerative disorders, primarily characterized by motor deficits, including bradykinesia, tremor, rigidity, and postural instability. The underlying pathophysiology involves the progressive loss of dopaminergic neurons within the substantia nigra pars compacta, leading to dopamine depletion in the basal ganglia circuitry. While motor symptoms are hallmark features of PD, emerging research highlights a wide range of non-motor symptoms, including cognitive impairments, mood disturbances, and autonomic dysfunctions. Inflammasome activation is pivotal in inducing neuroinflammation and promoting disease onset, progression, and severity of PD. Several studies have shown that long noncoding RNAs (lncRNAs) modulate inflammasomes in the pathogenesis of neurodegenerative diseases. Dysregulation of lncRNAs is linked to aberrant gene expression and cellular processes in neurodegeneration, causing the activation of inflammasomes that contribute to neuroinflammation and neurodegeneration. Inflammasomes are cytosolic proteins that form complexes upon activation, inducing inflammation and neuronal cell death. This review explores the significance of lncRNAs in regulating inflammasomes in PD, primarily focusing on specific lncRNAs such as nuclear paraspeckle assembly transcript 1 (NEATNEAT1), X-inactive specific transcript (XIST), growth arrest-specific 5 (GAS5), and HOX transcript antisense RNA (HOTAIR), which have been shown to activate or inhibit the NLRP3 inflammasome and induce the release of proinflammatory cytokines. Moreover, some lncRNAs mediate inflammasome activation through miRNA interactions. Understanding the roles of lncRNAs in inflammasome regulation provides new therapeutic targets for controlling neuroinflammation and reducing the progression of neurodegeneration. Identifying lncRNA-mediated regulatory pathways paves the way for novel therapies in the battle against these devastating neurodegenerative disorders.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Neelakudi Campus, Thiruvarur, 610005, India
| | - Mohamudha Parveen Rahamathulla
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Kingdom of Saudi Arabia
| | - Hussein Al-Dossary
- University Hospital, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Kingdom of Saudi Arabia
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Neelakudi Campus, Thiruvarur, 610005, India.
| |
Collapse
|