1
|
Wu H, Xie Y, Li A, Liu X, Guo L, Wu F, Yang Z, Zhang Z, Zhang X. LncRNA RNF144A-AS1 gene polymorphisms and their influence on lung cancer patients in the Chinese Han population. Noncoding RNA Res 2025; 10:252-260. [PMID: 39611047 PMCID: PMC11602538 DOI: 10.1016/j.ncrna.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Lung cancer is primarily classified as NSCLC, which is distinguished by a wide range of genetic variations. This study focused on RNF144A-AS1, a relatively unexplored lncRNA, to explore the impact of its genetic polymorphisms on the susceptibility to NSCLC. We detected RNF144A-AS1 expression and its correlation with prognosis and clinical pathological features using bioinformatics analysis. The association between RNF144A-AS1 polymorphism and NSCLC susceptibility was evaluated using case-control methods. This investigation featured a cohort of 700 NSCLC individuals and 700 healthy controls. The genotype of genetic variation was detected by PCR-RFLP and iMLDR, followed by subsequent calculation of OR and 95 % CI. Our data show that RNF144A-AS1 exhibits high expression levels in LUAD tissues and its expression is closely linked to LUAD progression and prognosis. Carrier of RNF144A-AS1 rs3806609 TT genotype increased NSCLC susceptibility compared to carrier of rs3806609 CC genotype (OR = 2.21, 95%CI = 1.57-3.13). Our study identifies RNF144A-AS1 genetic variants as potential susceptibility markers in NSCLC. RNF144A-AS1 promotes cell proliferation and migration in LUAD through the IFN-γ/JAK2/STAT1 signalling pathway. Collectively, these findings pave the way for developing targeted therapies and diagnostic tools based on RNF144A-AS1 and its variants.
Collapse
Affiliation(s)
- Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, 063210, China
| | - Yuning Xie
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Xiyao Liu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Liwen Guo
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Fengjun Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Zhenbang Yang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Zhi Zhang
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, 063000, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, 063210, China
| |
Collapse
|
2
|
Wang B, Liu W, Song B, Li Y, Wang Y, Tan B. Targeting LINC00665/miR-199b-5p/SERPINE1 axis to inhibit trastuzumab resistance and tumorigenesis of gastric cancer via PI3K/AKt pathway. Noncoding RNA Res 2025; 10:153-162. [PMID: 39399377 PMCID: PMC11467570 DOI: 10.1016/j.ncrna.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 10/15/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) serve as critical mediators of tumor progression and drug resistance in cancer. Herein, we identified a lncRNA, LINC00665, associated with trastuzumab resistance and development in gastric cancer (GC). LINC00665 was highly expressed in GC tissues and high expression of LINC00665 was correlated with poor prognosis. LINC00665 knockdown was verified to suppress migration, invasion, and resistance to trastuzumab in GC. Furthermore, we found that LINC00665 participates in the infiltration of naive B cells, mast cells, and T follicular helper (Tfh) cells. Mechanistically, LINC00665 was confirmed to regulate tumorigenesis and trastuzumab resistance by activating PI3K/AKt pathway. LINC00665 sponged miR-199b-5p to interact with SERPINE1 expression, resulting in the increase of phosphorylation of AKt, thus participating in the PI3K/AKt pathway. To summarize, LINC00665 facilitated the tumorigenesis and trastuzumab resistance of GC by sponging miR-199b-5p and promoting SERPINE1 expression, which further activated PI3K/AKt signaling; this finding reveals a new mechanism by which LINC00665 modulates tumor development and drug resistance in GC.
Collapse
Affiliation(s)
- Bingyu Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Wenbo Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Buyun Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Yingying Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| |
Collapse
|
3
|
Sethi SC, Singh R, Sahay O, Barik GK, Kalita B. Unveiling the hidden gem: A review of long non-coding RNA NBAT-1 as an emerging tumor suppressor and prognostic biomarker in cancer. Cell Signal 2025; 126:111525. [PMID: 39592019 DOI: 10.1016/j.cellsig.2024.111525] [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: 08/20/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Previously considered junk or non-functional, long non-coding RNAs (lncRNAs) have emerged over the past few decades as pivotal components in both physiological and pathological processes, including cancer. Neuroblastoma-associated transcript-1 (NBAT-1) was initially discovered a decade ago as a risk-associated tumor suppressor lncRNA in neuroblastoma (NB). Subsequent studies have consistently demonstrated that NBAT-1 serves as a dedicated tumor suppressor in many cancers. NBAT-1 is significantly downregulated in cancer, which is closely linked to higher histological grades, increased metastasis, and poor survival in cancer patients suggesting NBAT-1's potential as a prognostic biomarker. In this review, we delve into the current body of literature, elucidating the tumor-suppressive roles of NBAT-1 and the underlying regulatory mechanisms in the context of human malignancies. Additionally, we shed light on the mechanisms contributing to the diminished expression of NBAT-1 and its potential as both a prognostic biomarker and a promising therapeutic target in cancer.
Collapse
Affiliation(s)
- Subhash Chandra Sethi
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ragini Singh
- Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Osheen Sahay
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ganesh Kumar Barik
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Bhargab Kalita
- Amrita Research Center, Amrita Vishwa Vidyapeetham, Amrita Hospital, Mata Amritanandamayi Marg, Faridabad 121002, India.
| |
Collapse
|
4
|
Shi H, Ding G, Wang Y, Wang J, Wang X, Wang D, Lu P. Genome-wide identification of long non-coding RNA for Botrytis cinerea during infection to tomato (Solanum lycopersicum) leaves. BMC Genomics 2025; 26:7. [PMID: 39762752 PMCID: PMC11702200 DOI: 10.1186/s12864-024-11171-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Long non-coding RNA (lncRNA) plays important roles in animals and plants. In filamentous fungi, however, their biological function in infection stage has been poorly studied. Here, we investigated the landscape and regulation of lncRNA in the filamentous plant pathogenic fungus Botrytis cinerea by strand-specific RNA-seq of multiple infection stages. In total, 1837 lncRNAs have been identified in B. cinerea. A large number of lncRNAs were found to be antisense to mRNAs, forming 743 sense-antisense pairs, of which 55 antisense lncRNAs and their respective sense transcripts were induced in parallel as the infection stage. Although small RNAs were produced from these overlapping loci, antisense lncRNAs appeared not to be involved in gene silencing pathways. In addition, we found the alternative splicing events occurred in lncRNA. These results highlight the developmental stage-specific nature and functional potential of lncRNA expression in the infection stage and provide fundamental resources for studying infection stage-induced lncRNAs.
Collapse
Affiliation(s)
- Haojie Shi
- The Key Lab for Biology of Crop Pathogens and Insect Pests and Their Ecological Regulation of Zhejiang Province, College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, 311300, China
| | - Guijuan Ding
- The Key Lab for Biology of Crop Pathogens and Insect Pests and Their Ecological Regulation of Zhejiang Province, College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, 311300, China
| | - Yun Wang
- The Key Lab for Biology of Crop Pathogens and Insect Pests and Their Ecological Regulation of Zhejiang Province, College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, 311300, China
| | - Jiaqi Wang
- The Key Lab for Biology of Crop Pathogens and Insect Pests and Their Ecological Regulation of Zhejiang Province, College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, 311300, China
| | - Xiaoli Wang
- Jiangsu Provincial Key Construction Laboratory of Probiotics Preparation, College of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Dan Wang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A & F University, Hangzhou, 311300, China.
| | - Ping Lu
- The Key Lab for Biology of Crop Pathogens and Insect Pests and Their Ecological Regulation of Zhejiang Province, College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, 311300, China.
| |
Collapse
|
5
|
Gao Z, Chen S, Ye W. Cuproptosis related lncRNA signature as a prognostic and therapeutic biomarker in osteosarcoma immunity. Sci Rep 2025; 15:221. [PMID: 39747262 PMCID: PMC11696132 DOI: 10.1038/s41598-024-84024-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Osteosarcoma is one of the most common malignant bone tumours in children. In this study, we aimed to construct a cuproptosis-related lncRNAs signature to predict the prognosis and immune landscape of osteosarcoma patients. Databases from TARGET were used to acquire osteosarcoma patient datasets, which included clinical information and RNA sequencing data. Cuproptosis-related lncRNAs was obtained by correlation analysis. Through univariate Cox regression analysis, prognosis-related lncRNAs were obtained. We used nonnegative matrix factorization clustering to identify potential molecular subgroups with different cuproptosis-related lncRNA expression patterns. The least absolute shrinkage and selection operator algorithm and multivariate Cox regression analysis were used to construct the prognostic signature. The ESTIMATE algorithm, Gene Ontology and Kyoto Encyclopaedia of Genes and Genomes were applied to explore the underlying mechanisms in the immune landscape of osteosarcoma. We used gene set enrichment analysis to compare the different enrichments in the high-risk group and the low-risk group. Furthermore, we predicted the response to targeted drugs in patients with different risk groups. Using multivariable analysis, we developed a risk scoring model based on 7 long noncoding RNAs and calculated two molecular subgroups from osteosarcoma patients from the database. There is a better immune microenvironment in the low-risk group compared to the high-risk group. At the same time, the gene functional enrichment analysis based on the differently expressed genes obtained by grouping showed they were mainly related to immunity, indicating that cuproptosis-related lncRNAs may affect the prognosis of osteosarcoma by regulating immunity. Moreover, these patients in high-risk group were more susceptible to targeted drugs than the low-risk group. We identified a cuproptosis-related lncRNA prognostic signature for osteosarcoma and showed a close connection in terms of immunity. Moreover, we provided some potential targeted drugs for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Ziwei Gao
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Siqi Chen
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wei Ye
- Department of Gastroenterology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Han X, Shi F, Guo S, Li Y, Wang H, Song C, Wu S. LncRNA LINC00466 Promotes the Progression of Breast Cancer via miR-4731-5p/EPHA2 Pathway. Curr Pharm Biotechnol 2025; 26:120-131. [PMID: 38726776 DOI: 10.2174/0113892010290582240419051056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Breast Cancer (BC) is a female malignancy with a high mortality rate. Novel diagnostic and prognostic biomarkers are valuable for reducing BC mortality. Our study is designed to undrape the precise role of the LINC00466/miR-4731-5p/EPHA2 axis in BC. METHODS The Cancer Genome Atlas (TCGA) sequencing dataset was utilized to compare the levels of LINC00466. The levels of LINC00466, miR-4731-5p, and EPHA2 were tested by qRTPCR. Cell proliferation and cycle were detected by CCK-8 assay and flow cytometer. In vivo role of LINC00466 was tested by Xenograft nude models. Binding sites were predicted by TargetScan and Starbase. The binding relationship was employed by Dual-luciferase reporter gene assay and RNA pull-down assay. RESULTS LINC00466 was increased in human breast cancer tissues. LINC00466 was negatively associated with miR-4731-5p and positively correlated with EPHA2 in human breast cancer tissues. Down-regulation of LINC00466 suppressed the proliferation and arrested the cell cycle of breast cancer cells, and inhibited tumor growth in vivo. CONCLUSION LINC00466 promoted BC development via mediating the miR-4731-5p/EPHA2 axis, which has the potential value as a promising therapeutic target in BC.
Collapse
Affiliation(s)
- Xue Han
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, 233030, China
- Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, 233030, China
- Anhui Province Key Laboratory of Basic and Clinical Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233030, China
| | - Fan Shi
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004,China
| | - Shujun Guo
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, 233030, China
- Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, 233030, China
- Anhui Province Key Laboratory of Basic and Clinical Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233030, China
| | - Yao Li
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, 233030, China
- Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, 233030, China
- Anhui Province Key Laboratory of Basic and Clinical Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233030, China
| | - Hongtao Wang
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, 233030, China
- Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, 233030, China
- Anhui Province Key Laboratory of Basic and Clinical Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233030, China
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, 233030, China
- Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, 233030, China
- Anhui Province Key Laboratory of Basic and Clinical Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233030, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004,China
- Department of Pathology, Basic Medical College, Bengbu Medical University, Bengbu, 233030, China
- Department of Pathology, the Second People's Hospital of Anhui Province, Hefei, 230041, China
- Key Laboratory of Cancer Translational Medicine Center of Anhui Province, Bengbu Medical University, Bengbu, 233030, China
| |
Collapse
|
7
|
Fan Y, Ma Y, Wang R, Wang L. Prospective Study of lncRNA NORAD for Predicting Cerebrovascular Events in Asymptomatic Patients with Carotid Artery Stenosis. Clin Appl Thromb Hemost 2025; 31:10760296241299792. [PMID: 39763215 PMCID: PMC11705339 DOI: 10.1177/10760296241299792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Carotid artery stenosis (CAS) may cause many cerebrovascular diseases, and a biomarker for screening and monitoring is needed. This study focused on the clinical significance of long-chain non-coding RNA (lncRNA) non-coding RNA activated by DNA damage (NORAD) in patients with CAS and aimed to search for potential biomarkers of CAS. METHODS Eighty-six asymptomatic patients with CAS and 60 healthy individuals were enrolled, with corresponding clinical data and serum samples collected. The expression of NORAD was detected by reverse transcription-quantitive PCR (RT-qPCR). All patients were followed up for 2 years to collected the occurrence data of cerebrovascular events, and Kaplan-Meier and Cox regression were used for data analysis. Receiver operator characteristic curve was used to analyze the diagnostic value of NORAD in distinguishing CAS patients from healthy people, and to evaluate the prediction accuracy of NORAD. RESULTS NORAD is overexpressed in the serum of CAS patients, and associated with patients' hypertension, TC, LDL-C levels and stenosis degree. NORAD has high sensitivity (88.37%) and specificity (80.00%) in the identification of CAS patients (AUC = 0.917). NORAD was independently related to the occurrence of cerebrovascular events (HR = 2.435, P = .003). a logistic regression risk model for predicting cerebrovascular events was constructed with the parameters including NORAD, TC and LDL. CONCLUSION NORAD can be used as a diagnostic and prognostic biomarker for CAS, and NORAD, total cholesterol (TC), and low density lipoprotein cholesterol (LDL-C) can be independently correlated to predict cerebrovascular events.
Collapse
Affiliation(s)
- Yan Fan
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Yan Ma
- Department of Hyperbaric Oxygen, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Rui Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Lili Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| |
Collapse
|
8
|
Zhao Q, Xiong S, Cai H, He X, Shi X. Expression and Significance of the Long Non-Coding RNA APTR in the Occurrence and Development of Lung Adenocarcinoma. J Environ Pathol Toxicol Oncol 2025; 44:11-20. [PMID: 39462445 DOI: 10.1615/jenvironpatholtoxicoloncol.2024053394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
As one of the three major malignant tumors, lung adenocarcinoma (LUAD), with its rapid progression and high mortality rate, has become the most dangerous factor endangering human health. This study aims to explore new potential molecular targets, explore the regulatory role of lncRNA APTR in LUAD, and provide a more theoretical basis for the selection of LUAD therapeutic targets. The expression of APTR in LUAD was detected by PCR experiments, and the relationship between APTR and patients' clinical conditions and prognosis was analyzed by chi-square test, multifactor Cox regression, and Kaplan-Meier. The interaction between APTR and miR-298 and the regulation of LUAD cellular activities by APTR/miR-298 were explored by the luciferase reporter gene system. APTR expression was found to be upregulated in LUAD tissues and cells, and the expression of APTR was revealed to be substantially linked with lymph node metastases and TNM stage. High expression of LUAD also predicted a poor prognosis for patients. Downregulation of APTR expression significantly inhibited the activities of LUAD cells. In addition, APTR targeted miR-298 and negatively regulated miR-298 expression. The inhibitory effect of APTR knockdown on LUAD cell activity was also reversed after transfection with miR-298 inhibitor. Increasing expression of APTR is associated with patients' poor prognosis, APTR targets miR-298 and promotes LUAD cellular activity through negative regulation of miR-298.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Shi Xiong
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Hourong Cai
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Xiaofeng He
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University
| | - Xiaoming Shi
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| |
Collapse
|
9
|
Razipour M, Jamali Z, Sohrabpour S, Heidari F, Lotfi M, Ghadami E, Abtin M, Maghsudlu M, Sahebi L, Shakoori A. Dysregulated LINC01133 expression in laryngeal carcinoma: Prognostic implications and predicted ceRNA interactome. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2025; 14:93-107. [PMID: 39744510 PMCID: PMC11624609 DOI: 10.22099/mbrc.2024.50390.1996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Long non-coding RNAs (lncRNAs) have recently emerged as critical regulators of oncogenic or tumor-suppressive pathways in human cancers. LINC01133 is a lncRNA that has exhibited dichotomous roles in various malignancies but to the best of our knowledge, the role of LINC01133 in laryngeal squamous cell carcinoma (LSCC) has not been previously investigated. This study aimed to investigate the expression, clinical significance, and potential functions of the LINC01133 in LSCC. Integrative bioinformatics analysis of sequencing data obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets revealed LINC01133 as a differentially expressed lncRNA in head and neck/laryngeal cancers. Experimental validation via quantitative real-time PCR in 41 pairs of stage III and IV LSCC tissues and normal tissues adjacent to the tumor (NAT) demonstrated significant downregulation of LINC01133 in tumors (p<0.0001). Decreased LINC01133 expression associated with advanced tumor stage (p=0.0206) and lymph node metastasis (p=0.0203). The receiver operating characteristic analysis indicated potential diagnostic utility (AUC=0.7115, p=0.001). Bioinformatic predictions and literature mining suggested two potential competing endogenous RNA (ceRNA) mechanisms whereby LINC01133 may act as a tumor suppressor by sponging miR-205-5p to derepress the leucine-rich repeat kinase 2 (LRRK2) and androgen receptor, leading to dysregulation of cancer-related signaling cascades. This study provides initial evidence that loss of lncRNA LINC01133 expression may promote LSCC tumorigenesis, possibly by dysregulating microRNA interactions. Further verification of its regulatory mechanisms and diagnostic value is warranted.
Collapse
Affiliation(s)
- Masoumeh Razipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- These two authors contributed equally to this work
| | - Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- These two authors contributed equally to this work
| | - Saeed Sohrabpour
- Otorhinolaryngology Research Center, AmirAlam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farrokh Heidari
- Otorhinolaryngology Research Center, AmirAlam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Lotfi
- Department of Pathology and Otorhinolaryngology Research Center, AmirAlam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Ghadami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Abtin
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohaddese Maghsudlu
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leyla Sahebi
- Family Health Research Institute, Maternal-Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Shakoori
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, Cancer Institute of Iran, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Pain P, Tripathi A, Pillai PP. Involvement of PDGFR-integrin interactions in the regulation of anoikis resistance in glioblastoma progression. Cell Biol Int 2025; 49:3-15. [PMID: 39523497 DOI: 10.1002/cbin.12257] [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: 06/06/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 11/16/2024]
Abstract
The interactions between platelet-derived growth factor/PDGF receptor and integrin signaling are crucial for cells to respond to extracellular stimuli. Integrin interactions with PDGFR within the lipid rafts activate downstream cellular signaling pathways that regulate cell proliferation, cell migration, cell differentiation, and cell death processes. The mechanisms underlying PDGFR activation mediated receptor internalization, interactions with other cell-surface receptors, particularly extracellular matrix receptors, integrins, and how these cellular mechanisms switch on anchorage-independent cell survival, leading to anoikis resistance are discussed. The role of regulatory molecules such as noncoding RNAs, that can modulate several molecular and cellular processes, including autophagy, in the acquisition of anoikis resistance is also discussed. Overall, the review provides a new perspective on a complex interplay of regulatory cellular machineries involving autophagy, noncoding RNAs and cellular mechanisms of PDGFR activation, PDGFR-integrin interactions, and involvement of lipids rafts in the acquisition of anoikis resistance that regulates glioblastoma progression along with potential future strategies to develop novel therapeutics for glioblastoma multiforme.
Collapse
Affiliation(s)
- Pampa Pain
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ashutosh Tripathi
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|
11
|
Liu Y, Hatano K, Nonomura N. Liquid Biomarkers in Prostate Cancer Diagnosis: Current Status and Emerging Prospects. World J Mens Health 2025; 43:8-27. [PMID: 38772530 PMCID: PMC11704174 DOI: 10.5534/wjmh.230386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 05/23/2024] Open
Abstract
Prostate cancer (PCa) is a major health concern that necessitates appropriate diagnostic approaches for timely intervention. This review critically evaluates the role of liquid biopsy techniques, focusing on blood- and urine-based biomarkers, in overcoming the limitations of conventional diagnostic methods. The 4Kscore test and Prostate Health Index have demonstrated efficacy in distinguishing PCa from benign conditions. Urinary biomarker tests such as PCa antigen 3, MyProstateScore, SelectMDx, and ExoDx Prostate IntelliScore test have revolutionized risk stratification and minimized unnecessary biopsies. Emerging biomarkers, including non-coding RNAs, circulating tumor DNA, and prostate-specific antigen (PSA) glycosylation, offer valuable insights into PCa biology, enabling personalized treatment strategies. Advancements in non-invasive liquid biomarkers for PCa diagnosis may facilitate the stratification of patients and avoid unnecessary biopsies, particularly when PSA is in the gray area of 4 to 10 ng/mL.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koji Hatano
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
12
|
Tokunaga T, Hirata H, Hitaka Y, Fujii N, Kobayashi K, Hayano T, Asai Y, Shiraishi K. Role of long non‑coding RNA leucine‑rich repeat containing 75 A‑antisense RNA1 in the invasion and progression of renal cell carcinoma. Oncol Rep 2025; 53:11. [PMID: 39575481 PMCID: PMC11603548 DOI: 10.3892/or.2024.8844] [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: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 12/01/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) serve pivotal roles in cancer biology. The present study investigated the oncogenic roles of lncRNAs in renal cell carcinoma (RCC) and their potential as prognostic biomarkers. The lncRNA leucine‑rich repeat containing 75 A‑antisense RNA1 (LRRC75A‑AS1) was identified through lncRNA microarray as a potential lncRNA that may predict the efficacy of immune checkpoint inhibitor therapy and cancer progression in RCC. The present study subsequently assessed the expression of LRRC75A‑AS1 in 212 patients with clear cell RCC (ccRCC) who underwent nephrectomy, and performed in vitro functional analysis of LRRC75A‑AS1 in RCC cell lines. Additionally, the interactions between LRRC75A‑AS1, microRNA (miR)‑370‑5p and ADAMTS5 were explored. LRRC75A‑AS1 was revealed to be significantly upregulated in ccRCC tissues compared with in adjacent normal tissues, and high LRRC75A‑AS1 expression was associated with poor prognosis, including lower progression‑free survival, in patients with RCC. The knockdown of LRRC75A‑AS1 in RCC cell lines resulted in reduced cell proliferation and invasion, highlighting its role in promoting tumorigenesis. Furthermore, the interaction among LRRC75A‑AS1, miR‑370‑5p and ADAMTS5 was suggested as a regulatory mechanism underlying RCC progression. These findings indicated that LRRC75A‑AS1 may function as an oncogene in RCC, promoting cell proliferation and invasion. Its significant upregulation in ccRCC tissues and association with poor prognosis underscore its potential as a prognostic biomarker for RCC. Understanding the regulatory interactions among LRRC75A‑AS1, miR‑370‑5p and ADAMTS5 may provide new insights into the molecular mechanisms underlying RCC and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Takanori Tokunaga
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Hiroshi Hirata
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Yukihiro Hitaka
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Nakanori Fujii
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Keita Kobayashi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Takahide Hayano
- Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- The Division of Systems Medicine and Informatics, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Yoshiyuki Asai
- Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- The Division of Systems Medicine and Informatics, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- AI Systems Medicine Research and Training Center, Graduate School of Medicine, Yamaguchi University and Yamaguchi University Hospital, Ube, Yamaguchi 755-8505, Japan
| | - Koji Shiraishi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
13
|
Cai XX, Chen GM, Zheng ZQ, Yin YX, Wang S, Qiao L, Chen XJ, Zhao BW, Duan JL, Liang CC, Zhang RP, Wei CZ, Zhang FY, Huang BW, Liu ZX, Zhou ZW, Xie D, Cai MY, Yuan SQ, Li YF, Nie RC. Transcriptional landscape and predictive potential of long noncoding RNAs in peritoneal recurrence of gastric cancer. Mol Cancer 2024; 23:284. [PMID: 39736670 DOI: 10.1186/s12943-024-02196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/08/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) play a critical role in gastric cancer (GC) progression and metastasis. However, research comprehensively exploring tissue-derived lncRNAs for predicting peritoneal recurrence in patients with GC remains limited. This study aims to investigate the transcriptional landscape of lncRNAs in GC with peritoneal metastasis (PM) and to develop an integrated lncRNA-based score to predict peritoneal recurrence in patients with GC after radical gastrectomy. METHODS We analyzed the transcriptome profile of lncRNAs in paired peritoneal, primary gastric tumor, and normal tissue specimens from 12 patients with GC in the Sun Yat-sen University Cancer Center (SYSUCC) discovery cohort. Key lncRNAs were identified via interactive analysis with the TCGA database and SYSUCC validation cohort. A score model was constructed using the LASSO regression model and nomogram COX regression and evaluated using receiver operating characteristic curves. The role of lncRNAs in the PM of GC was then examined through wound healing, Transwell, 3D multicellular tumor spheroid invasion, and peritoneal cavity xenograft tumorigenicity assays in mice. RESULT Five essential lncRNAs were screened and incorporated into the PM risk score to predict peritoneal recurrence-free survival (pRFS). We developed a comprehensive, integrated nomogram score, including the PM risk score, pT, pN, and tumor size, which could effectively predict the 5-year pRFS with an Area under the curve of 0.79 (95% CI: 0.71-0.88). Subsequently, we demonstrated that one of these lncRNAs, CASC15, promoted the invasion and migration of GC cells in vitro and facilitated the PM of GC cells in vivo, initially verifying that lncRNAs contribute to the PM of GC. Mechanistic analysis demonstrated that CASC15 promoted EMT and metastasis by activating the JNK and p38 pathways. CONCLUSION A lncRNA-based integrated score was constructed in this study to predict peritoneal recurrence in patients clinically.
Collapse
Affiliation(s)
- Xiao-Xia Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Guo-Ming Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zi-Qi Zheng
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi-Xin Yin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shuang Wang
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Li Qiao
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510060, P. R. China
| | - Xiao-Jiang Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bai-Wei Zhao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jin-Ling Duan
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ruo-Peng Zhang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Cheng-Zhi Wei
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Fei-Yang Zhang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bo-Wen Huang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Mu-Yan Cai
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Yuan-Fang Li
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Run-Cong Nie
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
14
|
Sun Y, Wu J, Sun W, Liu C, Shi X. Novel insights into the interaction between IGF2BPs and ncRNAs in cancers. Cancer Cell Int 2024; 24:437. [PMID: 39732659 DOI: 10.1186/s12935-024-03591-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/28/2024] [Indexed: 12/30/2024] Open
Abstract
Insulin-like growth factor II mRNA-binding proteins (IGF2BPs), a family of RNA-binding proteins, are pivotal in regulating RNA dynamics, encompassing processes such as localization, metabolism, stability, and translation through the formation of ribonucleoprotein complexes. First identified in 1999 for their affinity to insulin-like growth factor II mRNA, IGF2BPs have been implicated in promoting tumor malignancy behaviors, including proliferation, metastasis, and the maintenance of stemness, which are associated with unfavorable outcomes in various cancers. Additionally, non-coding RNAs (ncRNAs), particularly long non-coding RNAs, circular RNAs, and microRNAs, play critical roles in cancer progression through intricate protein-RNA interactions. Recent studies, predominantly from 2018 onward, indicate that IGF2BPs can recognize and modulate ncRNAs via N6-methyladenosine (m6A) modifications, enriching the regulatory landscape of RNA-protein interactions in the context of cancer. This review explores the latest insights into the interplay between IGF2BPs and ncRNAs, emphasizing their potential influence on cancer biology.
Collapse
Affiliation(s)
- Yaya Sun
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Junjie Wu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Weimin Sun
- Department of General Surgery, Xuyi People's Hospital, Huai'an, 211700, China.
| | - Congxing Liu
- Department of General Surgery, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China.
| | - Xin Shi
- Department of General Surgery, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China.
| |
Collapse
|
15
|
Shen M, Mao Y, Wang X, Huang J, Zhang Q, Zhang J. LncRNA DNM1P35 sponges hsa-mir-326 to promote ovarian cancer progression. Sci Rep 2024; 14:31502. [PMID: 39732940 DOI: 10.1038/s41598-024-83170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in cancer progression. We found lncRNA DNM1P35 is elevated in ovarian tumors compared to normal tissues, and demonstrated that lncRNA DNM1P35 promoted cancer cell proliferation, migration and invasion in SK-OV-3 and OVCAR-3 cell lines. Furthermore, lncRNA DNM1P35 also facilitated the epithelial-mesenchymal transition (EMT) of ovarian cancer cells. Mechanistic studies identified microRNA-326 (miR-326) as a target of lncRNA DNM1P35. Overexpression of miR-326 diminished the tumor-promoting activity of lncRNA DNM1P35, resulting in reduction of Zinc finger E-box-binding homeobox 1 (ZEB1) expression and EMT features. We further revealed that ZEB1, a master transcription factor for EMT that is negatively regulated by miR-326, was essential for lncRNA DNM1P35-mediated cancer cell progression and EMT. Loss of ZEB1 led to compromised pro-tumoral activity of lncRNA DNM1P35. In vivo studies using a xenograft mouse model of ovarian cancer revealed that tumors with higher levels of lncRNA DNM1P35 led to shorter survival, increased tumor burden, as well as elevated expression of proliferative marker Ki67 and EMT marker ZEB1. Our comprehensive study underscored the significance of lncRNA DNM1P35 in ovarian cancer progression, elucidating the underlying mechanism through miR-326/ZEB1 axis to promote ovarian cancer progression.
Collapse
Affiliation(s)
- Mei Shen
- Department of Gynaecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Yun Mao
- Department of Infection Management, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, Wuxi, 214023, Jiangsu, China
| | - Xiaoshi Wang
- Department of Gynaecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Jinsong Huang
- Department of Gynaecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Qingsong Zhang
- Department of Gynaecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Jinwei Zhang
- Department of Gynaecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University/Wuxi Medical Center, Nanjing Medical University/Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
16
|
Zhou X, Gu C, Xiao L, Hu L, Chen G, Zuo F, Shao H, Fei B. LINC01094 promotes gastric cancer through dual targeting of CDKN1A by directly binding RBMS2 and HDAC1. Biol Direct 2024; 19:137. [PMID: 39719596 DOI: 10.1186/s13062-024-00582-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Accumulating studies have focused on long noncoding RNAs (lncRNAs) because of their regulatory effects on multiple cancers. However, the biological functions and molecular mechanisms of lncRNAs in gastric cancer (GC) remain to be elucidated in depth. METHODS Long intergenic nonprotein coding RNA 1094 (LINC01094), a differentially expressed lncRNA between GC tissues and adjacent normal tissues, was identified. Moreover, gain- and loss-of-function experiments in vitro and in vivo were carried out. To understand the mechanisms underlying the regulatory effects of LINC01094, we performed RNA pull-down assays, RNA immunoprecipitation assays, chromatin immunoprecipitation assays, luciferase reporter assays, etc. RESULTS: LINC01094 was markedly upregulated in GC tissues and cell lines, and LINC01094 upregulation was positively correlated with GC malignant behaviours in vitro and in vivo. Mechanistically, LINC01094 downregulated the expression of CDKN1A by interacting with RNA binding motif single stranded interacting protein 2 (RBMS2) and histone deacetylase 1 (HDAC1). Additionally, LINC01094 was confirmed to sponge miR-128-3p and participate in the LINC01094-miR-128-3p-RUNX family transcription factor 1 (RUNX1) feedback loop. Finally, Ro 5-3335, a validated RUNX1 inhibitor, was explored for anticancer drug development in GC. CONCLUSIONS The LINC01094-miR-128-3p-RUNX1 feedback loop downregulates CDKN1A and promotes GC cooperatively with RBMS2 and HDAC1. Furthermore, Ro 5-3335 may hold promising therapeutic potential in the treatment of GC. Hence, our study found an oncogenic lncRNA, LINC01094, which could be a promising target for cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214062, Jiangsu Province, China.
| | - Cheng Gu
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Linmei Xiao
- Department of Liver Disease, Wuxi No.5 People's Hospital Affiliated to Jiangnan University, Wuxi, 214000, Jiangsu Province, China
| | - Li Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang Province, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu Province, China
| | - Guanhua Chen
- Department of Radiation Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, Jiangsu Province, China
| | - Fei Zuo
- Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu Province, China
| | - Hongan Shao
- Department of Thoracic Surgery, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing Second Hospital, Nanjing, 210003, Jiangsu Province, China.
| | - Bojian Fei
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214062, Jiangsu Province, China.
| |
Collapse
|
17
|
Oghenemaro EF, Khaleel AQ, Rizaev JA, Roopashree R, Suliman M, Kazmi SW, Hjazi A, Rajput P, Mustafa YF, Abosaoda MK. Dysregulation of GAS5-miRNA-Mediated Signaling Pathways in Cancer Pathobiology: A Comprehensive Exploration of Pathways Influenced by this Axis. Biochem Genet 2024:10.1007/s10528-024-10997-x. [PMID: 39718723 DOI: 10.1007/s10528-024-10997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024]
Abstract
The long non-coding RNA Growth Arrest-Specific 5 (GAS5) is pivotal in modulating key signaling pathways by functioning as a molecular sponge for microRNAs (miRNAs). GAS5 is notably recognized for its antitumor properties, primarily through its ability to sequester oncogenic miRNAs, thereby influencing critical pathways such as p53, Wnt/β-catenin, and PI3K/Akt, all of which are integral to cell proliferation, apoptosis, and metastasis. The disruption of GAS5-miRNA interactions has been implicated in various malignancies, reinforcing its potential as both a biomarker and a therapeutic target. This paper delves into the intricate signaling cascades affected by GAS5-miRNA interactions and thoroughly investigates the diagnosis and treatment prospects associated with GAS5. Moreover, it addresses both the challenges and opportunities for translational applicability of these findings in clinical environments. The study emphasizes GAS5's significance within the cancer molecular landscape and posits that precise modulation of GAS5-miRNA interactions could catalyze transformative developments in cancer diagnostics and therapeutic approaches. This comprehensive review not only highlights the critical role of non-coding RNAs in cancer biology but also aims to lay the groundwork for future investigations aimed at harnessing these insights for therapeutic interventions.
Collapse
Affiliation(s)
- Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, College of Engineering, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Syeda Wajida Kazmi
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia.
| | - Pranchal Rajput
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
18
|
Pan J, Fan W, Gu C, Xi Y, Wang Y, Wang P. Long Non-Coding RNAs as Diagnostic Biomarkers for Ischemic Stroke: A Systematic Review and Meta-Analysis. Genes (Basel) 2024; 15:1620. [PMID: 39766887 PMCID: PMC11675862 DOI: 10.3390/genes15121620] [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: 10/17/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Ischemic stroke is a serious cerebrovascular disease, highlighting the urgent need for reliable biomarkers for early diagnosis. Recent reports suggest that long non-coding RNAs (lncRNAs) can be potential biomarkers for ischemic stroke. Therefore, our study seeks to investigate the potential diagnostic value of lncRNAs for ischemic stroke by analyzing existing research. A comprehensive literature search was conducted across the PubMed, ScienceDirect, Wiley Online Library, and Web of Science databases for articles published up to July 10, 2024. Statistical analyses were performed using Stata 17.0 software to calculate pooled sensitivity, specificity, positive likelihood ratio (PLR), diagnostic odds ratio (DOR), negative likelihood ratio (NLR), and area under the curve (AUC). Heterogeneity was explored with the Cochran-Q test and the I2 statistical test, and publication bias was assessed with Deeks' funnel plot. A total of 44 articles were included, involving 4302 ischemic stroke patients and 3725 healthy controls. Results demonstrated that lncRNAs H19, GAS5, PVT1, TUG1, and MALAT1 exhibited consistent trends across multiple studies. The pooled sensitivity of lncRNAs in the diagnosis of ischemic stroke was 79% (95% CI: 73-84%), specificity was 88% (95% CI: 77-94%), PLR was 6.63 (95% CI: 3.11-14.15), NLR was 0.23 (95% CI: 0.16-0.33), DOR was 28.5 (95% CI: 9.88-82.21), and AUC was 0.88 (95% CI: 0.85-0.90). Furthermore, the results of subgroup analysis indicated that lncRNA H19 had superior diagnostic performance. LncRNAs demonstrated strong diagnostic accuracy in distinguishing ischemic stroke patients from healthy controls, underscoring their potential as reliable biomarkers. Because most of the articles included in this study originate from China, large-scale, high-quality, multi-country prospective studies are required to further validate the reliability of lncRNAs as biomarkers for ischemic stroke.
Collapse
Affiliation(s)
- Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (W.F.); (C.G.)
| | - Weijian Fan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (W.F.); (C.G.)
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (W.F.); (C.G.)
| | - Yongmei Xi
- Institute of Genetic, Zhejiang University, Hangzhou 310007, China;
| | - Yu Wang
- Department of Medicine, Beijing Zhongwei Research Center, Biological and Translational Medicine, Beijing 100161, China;
| | - Peter Wang
- Department of Medicine, Beijing Zhongwei Research Center, Biological and Translational Medicine, Beijing 100161, China;
| |
Collapse
|
19
|
Chen Z, Wu Z, Wu M, Zhang Y, Hou S, Wang X, Peng Y. LncRNA SNHG16 Drives PD-L1-Mediated Immune Escape in Colorectal Cancer through Regulating miR-324-3p/ELK4 Signaling. Biochem Genet 2024:10.1007/s10528-024-11000-3. [PMID: 39688781 DOI: 10.1007/s10528-024-11000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
Colorectal cancer (CRC) is a common malignancy that claims the life of many patients. Nucleolar RNA host gene 16 (SNHG16) has been identified as an oncogene in CRC development. However, the role and mechanism of SNHG16 in CRC remain unclear. A total of 27 cases of CRC tumor tissues and adjacent tissues were collected to investigate the expression and correlation among SNHG16, miR-324-3p, ELK4 and PD-L1 using qRT-PCR, western blot and Pearson analysis. Cell proliferation, migration and invasion abilities were determined using CCK-8 and transwell assays. The cytotoxicity of CD8 + T cells and the apoptosis of CD8+ T cells was evaluated by LDH assay and flow cytometry, respectively. Dual luciferase assay, RIP and ChIP methods were performed to verify molecular interactions. Our results showed that SNHG16, ELK4 and PD-L1 expression were abnormally elevated and miR-324-3p expression was decreased in tumor tissues from CRC patients and CRC cells. SNHG16 silencing resulted in suppression of cell growth, metastasis, and immune escape of CRC cells, which was reversed by miR-324-3p inhibitor and ELK4 overexpression. Mechanistically, SNHG16 acted as a competitive endogenous RNA to enhance ELK4 expression by sponging miR-324-3p, thereby provoking the transcription of PD-L1. Our results demonstrated that SNHG16 silencing led to the suppression of cell growth, metastasis, and immune escape of CRC cells through mediating miR-324-3p/ELK4/PD-L1 axis, offering promising targets for CRC treatment.
Collapse
Affiliation(s)
- Zhiyuan Chen
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China.
| | - Zhenjuan Wu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China
| | - Minghao Wu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China
| | - Yu Zhang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China
| | - Sha Hou
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China
| | - Xiangyang Wang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China
| | - Ya Peng
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No. 61 West Jiefang Road, Changsha, 410005, Hunan, People's Republic of China.
| |
Collapse
|
20
|
Yang C, Shu J, Li Y, Zhao N, Liu X, Tian X, Sun Z, Tabish MS, Hong Y, Chen K, Sun M. Long non-coding RNAs are involved in the crosstalk between cancer-associated fibroblasts and tumor cells. Front Immunol 2024; 15:1469918. [PMID: 39717771 PMCID: PMC11663902 DOI: 10.3389/fimmu.2024.1469918] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
The proliferation of tumors is not merely self-regulated by the cancer cells but is also intrinsically connected to the tumor microenvironment (TME). Within this complex TME, cancer-associated fibroblasts (CAFs) are pivotal in the modulation of tumor onset and progression. Rich signaling interactions exist between CAFs and tumor cells, which are crucial for tumor regulation. Long non-coding RNAs (LncRNAs) emerge from cellular transcription as a class of functionally diverse RNA molecules. Recent studies have revealed that LncRNAs are integral to the crosstalk between CAFs and tumor cells, with the capacity to modify cellular transcriptional activity and secretion profiles, thus facilitating CAFs activation, tumor proliferation, metastasis, drug resistance, and other related functionalities. This comprehensive review revisits the latest research on LncRNA-mediated interactions between CAFs and tumor cells, encapsulates the biological roles of LncRNAs, and delves into the molecular pathways from a broader perspective, aspiring to offer novel perspectives for a deeper comprehension of the etiology of tumors and the enhancement of therapeutic approaches.
Collapse
Affiliation(s)
- Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yiwei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiaonan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Tian
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Zexin Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Muhammad Saud Tabish
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yichen Hong
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Xia J, Zhou C, Zhao H, Zhang J, Chai X. LINC01614 Accelerates CRC Progression via STAT1/LINC01614/miR-4443/PFKFB3-Mediated Aerobic Glycolysis. Dig Dis Sci 2024:10.1007/s10620-024-08756-4. [PMID: 39641899 DOI: 10.1007/s10620-024-08756-4] [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: 08/19/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is an aggressive malignancy among malignant tumours, with a high incidence globally. LINC01614, a long non-coding RNA, has been identified as an essential regulator in multiple cancer types. However, its biological functions and underlying molecular mechanisms in CRC remain largely unknown. METHODS In this study, we employed RT-qPCR to assess the expression levels of LINC01614 in CRC samples. In vitro, glucose metabolism experiments were conducted to evaluate glucose metabolism in cells. The binding relationship between miR-4443, PFKFB3, and LINC01614 was confirmed through fluorescence reporter gene detection. The subcellular localization of LINC01614 in CRC cells was determined using FISH and subcellular fractionation experiments. Additionally, a mouse subcutaneous tumor model was established for in vivo experiments. RESULTS Our findings reveal that LINC01614 is upregulated in CRC tissues. Silencing of LINC01614 suppresses the malignant behaviors of CRC cells, including cell proliferation, invasion, migration, and aerobic glycolysis. Furthermore, we discovered that LINC01614 promotes the expression of PFKFB3. Additional experiments demonstrated that LINC01614 binds to miR-4443, leading to the upregulation of PFKFB3 expression. Further experiments confirmed that the LINC01614/miR-4443/PFKFB3 axis promotes CRC cell malignancy by enhancing aerobic glycolysis. Additionally, we found that STAT1 promotes the transcription of LINC01614. CONCLUSION These findings uncover a novel regulatory pathway wherein STAT1-induced LINC01614 enhances PFKFB3 expression by sponging miR-4443, thereby accelerating CRC development. This understanding may lead to novel therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Jiangyan Xia
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Chenglin Zhou
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Heng Zhao
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Jun Zhang
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Xiaoming Chai
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China.
| |
Collapse
|
22
|
Elimam H, Alhamshry NAA, Hatawsh A, Elfar N, Moussa R, Radwan AF, Abd-Elmawla MA, Elkashlan AM, Zaki MB, Abdel-Reheim MA, Mohammed OA, Doghish AS. Natural products and long noncoding RNA signatures in gallbladder cancer: a review focuses on pathogenesis, diagnosis, and drug resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9549-9571. [PMID: 39028332 DOI: 10.1007/s00210-024-03279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
Gallbladder cancer (GBC) is an aggressive and lethal malignancy with a poor prognosis. Long noncoding RNAs (lncRNAs) and natural products have emerged as key orchestrators of cancer pathogenesis through widespread dysregulation across GBC transcriptomes. Functional studies have revealed that lncRNAs interact with oncoproteins and tumor suppressors to control proliferation, invasion, metastasis, angiogenesis, stemness, and drug resistance. Curcumin, baicalein, oleanolic acid, shikonin, oxymatrine, arctigenin, liensinine, fangchinoline, and dioscin are a few examples of natural compounds that have demonstrated promising anticancer activities against GBC through the regulation of important signaling pathways. The lncRNAs, i.e., SNHG6, Linc00261, GALM, OIP5-AS1, FOXD2-AS1, MINCR, DGCR5, MEG3, GATA6-AS, TUG1, and DILC, are key players in regulating the aforementioned processes. For example, the lncRNAs FOXD2-AS1, DILC, and HOTAIR activate oncogenes such as DNMT1, Wnt/β-catenin, BMI1, and c-Myc, whereas MEG3 and GATA6-AS suppress the tumor proteins NF-κB, EZH2, and miR-421. Clinically, specific lncRNAs can serve as diagnostic or prognostic biomarkers based on overexpression correlating with advanced TNM stage, metastasis, chemoresistance, and poor survival. Therapeutically, targeting aberrant lncRNAs with siRNA or antisense oligos disrupts their oncogenic signaling and inhibits GBC progression. Overall, dysfunctional lncRNA regulatory circuits offer multiple avenues for precision medicine approaches to improve early GBC detection and overcome this deadly cancer. They have the potential to serve as novel biomarkers as they are detectable in bodily fluids and tissues. These findings enhance gallbladder treatments, mitigating resistance to chemo- and radiotherapy.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Nora A A Alhamshry
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, 26th of July Corridor, Sheikh Zayed City, Nile University, Giza, 12588, Egypt
| | - Nourhan Elfar
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo, 11578, Egypt
- Egyptian Drug Authority (EDA), Ministry of Health and Population, Cairo, 11567, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Akram M Elkashlan
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, 11961, Shaqra, Saudi Arabia.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt.
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
23
|
Al-Noshokaty TM, El-Sayyad GS, Abdelhamid R, Mansour A, Abdellatif N, Alaaeldien A, Reda T, Gendi D, Abdelmaksoud NM, Elshaer SS, Doghish AS, Mohammed OA, Abulsoud AI. Long non-coding RNAs and their role in breast cancer pathogenesis and drug resistance: Navigating the non-coding landscape review. Exp Cell Res 2024; 444:114365. [PMID: 39626864 DOI: 10.1016/j.yexcr.2024.114365] [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: 09/03/2024] [Revised: 10/27/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
Despite the progress made in the development of targeted therapies, breast cancer (BC) continues to pose a significant threat to the health of women. Transcriptomics has emerged due to the advancements in high-throughput sequencing technology. This provides crucial information about the role of non-coding RNAs (ncRNAs) in human cells, particularly long ncRNAs (lncRNAs), in disease development and function. When the control of these ncRNAs is disrupted, various illnesses emerge, including cancer. Numerous studies have produced empirical data on the function of lncRNAs in tumorigenesis and disease development. However, the roles and mechanisms of numerous lncRNAs remain unidentified at the molecular level because their regulatory role and the functional implications of abnormalities in cancer biology have yet to be thoroughly defined. The review gives an itemized summary of the most current developments in the role of lncRNA in BC, focusing on three main pathways, PI3K, MAPK, NF-kB, and hypoxia, and their resistance mechanisms.
Collapse
Affiliation(s)
- Tohada M Al-Noshokaty
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Gharieb S El-Sayyad
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Badr, Cairo, 11829, Egypt; Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt; Microbiology and Immunology Department, Faculty of Pharmacy, Ahram Canadian University (ACU), 6th October City, Giza, Egypt.
| | - Rehab Abdelhamid
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Abdallah Mansour
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Nourhan Abdellatif
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Ayat Alaaeldien
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Tasnim Reda
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - David Gendi
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11823, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo, 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
| |
Collapse
|
24
|
Liu M, Song X, Sun Y, Zhang T. LncRNA OIP5-AS1 Targets the miR-140-5p/UBR5 Cascade to Promote the Development of Gastric Cancer. Mol Biotechnol 2024; 66:3583-3596. [PMID: 38112962 DOI: 10.1007/s12033-023-00958-x] [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: 08/20/2023] [Accepted: 10/23/2023] [Indexed: 12/21/2023]
Abstract
Gastric cancer (GC) is a malignant tumor with the highest incidence among all kinds of malignant tumors in China. Long noncoding RNAs (LncRNAs) have been reported to act as microRNA (miRNAs) sponges and thus play key roles in biological processes and pathogenesis. Thus, this study aimed to investigate the functional effects and the regulatory mechanism of lncRNA opa interacting protein 5-antisense 1 (OIP5-AS1) in gastric cancer cells. The expression of OIP5-AS1, miR-140-5p, Ubiquitin protein ligase E3 component n-recognin 5 (UBR5) was detected using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, apoptosis, migration, and invasion were assessed using Cell-Counting Kit-8 (CCK-8), Flow cytometry, and Transwell assays. UBR5 protein level was detected by Western blot. Binding between miR-140-5p and OIP5-AS1 or UBR5 was predicted by Starbasev2.0 and TargetScan, and verified using Dual-luciferase reporter assays and RNA pull-down assay. A xenograft mice model was used to evaluate the effects of OIP5-AS1 on tumor growth in vivo. OIP5-AS1 was upregulated in GC cancer and cells. OIP5-AS1 knockdown inhibited cell proliferation, migration, invasion, but induced cell apoptosis in GC. In mechanism, OIP5-AS1 might serve as a sponge for miR-140-5p to enhance UBR5 expression. Moreover, overexpression of miR-140-5p or UBR5 partly reversed the effects of OIP5-AS1 depletion on the progression of GC cells. Furthermore, OIP5-AS1 depletion also suppressed tumor growth in vivo. OIP5-AS1 silencing might suppress proliferation, migration, invasion, and induced apoptosis in GC cells by regulating the miR-140-5p/UBR5 axis.
Collapse
Affiliation(s)
- Mei Liu
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136 Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei Province, China
| | - Xiujun Song
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136 Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei Province, China
| | - Yinyin Sun
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136 Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei Province, China.
| | - Tieshan Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136 Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei Province, China.
| |
Collapse
|
25
|
Zeng M, Chen Z, Wang Y, Yang Z, Xiang J, Wang X, Wang X. LncRNA MALAT1 to Enhance Pyroptosis in Viral Myocarditis Through UPF1-Mediated SIRT6 mRNA Decay and Wnt-β-Catenin Signal Pathway. Cardiovasc Toxicol 2024; 24:1439-1454. [PMID: 39367210 DOI: 10.1007/s12012-024-09922-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/14/2024] [Indexed: 10/06/2024]
Abstract
Viral myocarditis (VMC) is an inflammatory disease of the myocardium caused by cardioviral infection, especially coxsackievirus B3 (CVB3), and is a major contributor to acute heart failure and sudden cardiac death in children and adolescents. LncRNA MALAT1 knockdown reportedly inhibits the differentiation of Th17 cells to attenuate CVB3-induced VMC in mice. Moreover, long non-coding RNAs (lncRNAs) interact with RNA-binding proteins (RBPs) to regulate UPF1-mediated mRNA decay. However, it remains unclear whether MALAT1 can bind to UPF1 to mediate the mRNA decay of its target genes in VMC. Herein, we aimed to explore the effect of lncRNA MALAT1 on UPF1-mediated SIRT6 mRNA decay in VMC using in vivo and in vitro experiments. CVB3-infected BABL/C mice were used as VMC models, and MALAT1 interfering adenovirus was injected to achieve MALAT1 knockdown. The heart function of the VMC mice was assessed using echocardiography. Pathological changes in myocardial tissues were assessed after hematoxylin-eosin staining. Myocardial injury and inflammation were evaluated by measuring creatine kinase isoenzyme B, cardiac troponin T, interleukin (IL)-1β, and IL-18. TUNEL staining was performed to assess apoptosis in myocardial tissues. In vitro experiments were performed using H9c2 cells after transfection and CVB3 infection. The lactic dehydrogenase release, caspase-1 activity, and IL-1β and IL-18 levels in the cellular supernatant were detected. Western blotting was performed to determine the expression of pyroptosis-related proteins (GSDMD-N, NLRP3, ASC, and Cleaved-Caspase-1) and Wnt/β-catenin signal pathway-related proteins (Wnt1, β-catenin, and p-GSK-3β). RNA immunoprecipitation and RNA stability assays assessed the relationship between MALAT1, UPF1, and SIRT6. CVB3-infected mice and H9c2 cells exhibited elevated MALAT1 and reduced SIRT6 expression. MALAT1 knockdown or SIRT6 overexpression suppressed inflammation and pyroptosis and inhibited the activation of the Wnt/β-catenin signal pathway in myocardial tissues and cells. MALAT1 enhanced the enrichment of SIRT6 mRNA by UPF1 and disturbed the stability of SIRT6 mRNA to promote the development of VMC. MALAT1 can bind UPF1 to mediate SIRT6 mRNA decay and activate the Wnt/β-catenin signal pathway in VMC.
Collapse
Affiliation(s)
- Min Zeng
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China.
| | - Zhi Chen
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| | - Yefeng Wang
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| | - Zhou Yang
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| | - Jinxing Xiang
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| | - Xiang Wang
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| | - Xun Wang
- Department of Cardiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan, 410007, People's Republic of China
| |
Collapse
|
26
|
Zhang L, Li G, Jiang Y, Jin L. LINC00668 silencing retards tumorigenesis via sponging miR-518c-3p to regulating WDR1 in triple negative breast cancer. Int J Biol Macromol 2024; 282:136698. [PMID: 39427801 DOI: 10.1016/j.ijbiomac.2024.136698] [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: 08/31/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
The emergence of long noncoding RNAs (lncRNAs) was proved to be crucial to the aggravation of triple negative breast cancer (TNBC), a fatal female malignancy. LINC00668 was unveiled as an overexpressed lncRNA in TNBC previously. However, its exact function and whether it functioned in TNBC development needs to be ascertained. To explore this, qRT-PCR was used to detect its dysregulation in TNBC cells. Biological functions of LINC00668 were determined through loss-of-function experiments. Bioinformatics analysis was utilized to predict the downstream modulatory genes of LINC00668. Dual-luciferase reporter assay plus RNA immunoprecipitation analysis, quantitative PCR analysis, and rescue assays were employed for the exploration of potential action of mode in competitive endogenous RNA (ceRNA) network. It was revealed that LINC00668 was upregulated and its depletion resulted in impeded proliferation and migration of TNBC cells. Bioinformatics analysis and mechanical assays uncovered that LINC00668 sponged miR-518c-3p to facilitate WDR1 level in TNBC. Furthermore, rescue experiments demonstrated that LINC00668/miR-518c-3p pathway contributed to TNBC cell proliferation and migration in the form of WDR1 dependency. Overall our study might discover a vital clue for the cure of lncRNA-directed treatment for TNBC patients.
Collapse
Affiliation(s)
- Liaoyun Zhang
- Department of Pharmacy, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610000, China
| | - Gen Li
- Department of Pharmacy, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610000, China
| | - Yongxian Jiang
- Department of Pharmacy, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610000, China
| | - Li Jin
- School of Pharmacy, State Key Laboratory for Quality Research of Chinese Medicine, Macau University of Science and Technology, Taipa 999078, Macao; Central Laboratory, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55 Renmin South Road, Chengdu 610041, Sichuan, China.
| |
Collapse
|
27
|
Zhou J, Xu J, Cheng L, Li S, Jiang D, Zhang J, Sheng Y. Alteration of lncRNA RHPN1-AS1 predicts clinical prognosis and regulates the progression of bladder cancer via modulating miR-485-5p. Int J Biol Markers 2024; 39:284-291. [PMID: 39233606 DOI: 10.1177/03936155241281076] [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: 09/06/2024]
Abstract
BACKGROUND Exploring effect biomarkers that monitor tumor progression and predict the prognosis could benefit the clinical management of bladder cancer and improve the postoperative life of patients. This study aimed to estimate the function of long non-coding (lnc)RNA RHPN1-AS1 (RHPN1-AS1) in bladder cancer and the potential molecular mechanism. METHODS The expression of RHPN1-AS1 was evaluated in bladder cancer tissues from 115 patients and cells by polymerase chain reaction. The clinical significance of RHPN1-AS1 was assessed and its effect was also estimated in cell proliferation, migration, and invasion. The underlying molecular mechanism was explored by the dual-luciferase reporter assay. RESULTS The expression of RHPN1-AS1 was 2.91-fold elevated in bladder cancer, which showed a close correlation with advanced tumor node metastasis stage (P = 0.013) and the presence of lymph node metastasis (P = 0.018). RHPN1-AS1 also served as a poor prognostic indicator (hazard ratio = 2.563) for bladder cancer. The knockdown of RHPN1-AS1 significantly suppressed the proliferation and metastasis ability of bladder cancer cells. Moreover, miR-485-5p was found to mediate the function of RHPN1-AS1 in bladder cancer, which was considered the underlying regulatory mechanism. CONCLUSIONS RHPN1-AS1 serves as a prognostic biomarker and tumor promoter in bladder cancer via modulating miR-485-5p, which might be a reliable target of bladder cancer therapy.
Collapse
Affiliation(s)
- Jingmin Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinshan Xu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lin Cheng
- Department of Urology Surgery, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Shuhui Li
- Department of Joint Surgery, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Deqi Jiang
- Department of Urology Surgery, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Jianchao Zhang
- Department of Urology Surgery, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Yulong Sheng
- Department of Interventional Medicine, The Affiliated Hospital of Qingdao University (Pingdu), Qingdao, Shandong, China
| |
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
|
Wu T, Zeng L, Peng C, Zhao Z, Mu W, Wang S, Tan D. SNHG12 in cancer-associated fibroblast-derived extracellular vesicle induces macrophage-myofibroblast transition. Epigenomics 2024; 16:1415-1427. [PMID: 39568326 PMCID: PMC11622769 DOI: 10.1080/17501911.2024.2430166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024] Open
Abstract
AIMS To investigate mechanism of lncRNA SNHG12 induced macrophage-myofibroblast transition (MMT) in cancer-associated fibroblasts (CAFs)-derived extracellular vesicles (EVs) in non-small cell lung cancer (NSCLC). METHOD CAFs EVs were isolated from human NSCLC tissue and adjacent cancerous tissue (n = 3), and their morphology and particle size were evaluated. Macrophages and MMT cells with different phenotypes were detected, and the binding relationship of lncRNA SNHG12, miR-181a-5p, and Smad3 was verified. RESULT LncRNA SNHG12 derived from CAFs-EVs promoted the transformation of M2 macrophages into MMT. In addition, lncRNA-SNHG12 increased the expression of Smad3 which was significantly upregulated in MMT through sponge of miR-181a-5p. CONCLUSION LncRNA SNHG12 derived from CAFs-EV induced MMT in NSCLC.
Collapse
Affiliation(s)
- Tao Wu
- Cardiothoracic surgery department, Banan Hospital of Chongqing medical university, Chongqing, China
| | - Li Zeng
- Department of Pathology, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Chao Peng
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Zheng Zhao
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Weihao Mu
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Song Wang
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| | - Deli Tan
- Department of Thoracic surgery, Chongqing Ninth People’s Hospital, Chongqing, China
| |
Collapse
|
30
|
Liu Y, Li K, Gao Y, Feng Y, Zhao X, Hou R. lncRNA WAC-AS1 promotes the progression of gastric cancer through miR-204-5p/HOXC8 axis. Transl Oncol 2024; 50:102139. [PMID: 39395273 DOI: 10.1016/j.tranon.2024.102139] [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: 05/07/2024] [Revised: 08/29/2024] [Accepted: 09/21/2024] [Indexed: 10/14/2024] Open
Abstract
LncRNAs affect tumorigenesis, and although the genesis, regulation and physiological mechanism of lncRNAs in gastric cancer (GC) have been reported, the research of lncRNAs still have a lot of value. Through comprehensive bioinformatics analysis, we screened the candidate lncRNA WAC-AS1(WAC-AS1). We analyzed WAC-AS1 expression in GC related tissues and cells using qRT-PCR. WAC-AS1's impact on GC growth and metastasis was investigated. LncRNA WC-AS-miR-204-5p-HOXC8 interaction was established through dual-luciferase reporter, FISH, RIP and RNA pull-down assay. We observed substantial upregulation in WAC-AS1 expression in cells and tissues of GC. WAC-AS1 through miR-204-5p/HOXC8 axis promoted GC proliferation, invasion, and migration. WAC-AS1 plays a cancer-promoting role for promoting the progression of GC.
Collapse
Affiliation(s)
- Yan Liu
- Department of Ultrasonography, the third Norman Bethune Hospital of Jilin university, Changchun, Jilin, China
| | - Kaixuan Li
- Department of Gastrointestinal surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, China
| | - Yongjian Gao
- Department of Gastrointestinal and Colonretal Surgery, the third Norman Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Ye Feng
- Department of Gastrointestinal and Colonretal Surgery, the third Norman Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoling Zhao
- Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Hebei Key Laboratory of Infectious Disease Pathogenesis and Precise Diagnosis and Treatment, Baoding, Hebei, China.
| | - Ruizhi Hou
- Department of Gastrointestinal and Colonretal Surgery, the third Norman Bethune Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
31
|
Wang J, Xu B, Liang L, Chen Q. Long Non-coding RNA 02298 Promotes the Malignancy of HCC by Targeting the miR-28-5p/CCDC6 Pathway. Biochem Genet 2024; 62:4967-4986. [PMID: 38381357 DOI: 10.1007/s10528-023-10662-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/30/2023] [Indexed: 02/22/2024]
Abstract
Hepatocellular carcinoma (HCC) is a malignancy characterized by a high fatality rate. Increasing evidence indicating that long non-coding RNAs (lncRNAs) play a regulatory role in hepatocellular carcinoma (HCC). Among them, the correlation between LINC02298 and HCC remains unknown. The expression and subcellular localization of LINC02298 in HCC tissues and cell lines were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Furthermore, the correlation between the expression of LINC02298 and clinicopathological features of HCC patients was analyzed. The regulatory effects of LINC02298 in HCC were investigated using colony formation, cell count Kit-8(CCK8), Transwell, EDU, cell cycle and apoptosis analysis. In addition, the expression of EMT-related proteins were detected by western blotting. Dual-luciferase reporter, RT-qPCR and rescue assays were employed to validate the involvement of the LINC02298/miR-28-5p/CCDC6 axis in the progression of HCC. The up-regulation of LINC02298 was observed in hepatocellular carcinoma (HCC) tissues and cells, and it was found to be correlated with a negative prognosis in patients with HCC. Overexpression of LINC02298 enhanced the proliferation, migration, invasion, and induction of Epithelial-Mesenchymal Transition (EMT) while suppressing apoptosis in HCC cells. LINC02298 bind to miR-28-5p to regulate the expression of CCDC6. Inhibition of miR-28-5p saved the inhibitory effect of shLINC02298, and knockdown of CCDC6 also saved the inhibitory effect of miR-28-5p on HCC in vitro and in vivo. LINC02298 regulates the expression of CCDC6 by sponging of miR-28-5p, thereby facilitating the the malignancy and EMT of HCC.
Collapse
Affiliation(s)
- Jinyi Wang
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210019, Jiangsu, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bin Xu
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210019, Jiangsu, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Litao Liang
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210019, Jiangsu, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Chen
- Department of General Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China.
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
32
|
Guha P, Chini A, Rishi A, Mandal SS. Long noncoding RNAs in ubiquitination, protein degradation, and human diseases. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195061. [PMID: 39341591 DOI: 10.1016/j.bbagrm.2024.195061] [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: 04/14/2024] [Revised: 08/07/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Protein stability and turnover is critical in normal cellular and physiological process and their misregulation may contribute to accumulation of unwanted proteins causing cellular malfunction, neurodegeneration, mitochondrial malfunction, and disrupted metabolism. Signaling mechanism associated with protein degradation is complex and is extensively studied. Many protein and enzyme machineries have been implicated in regulation of protein degradation. Despite these insights, our understanding of protein degradation mechanisms remains limited. Emerging studies suggest that long non-coding RNAs (lncRNAs) play critical roles in various cellular and physiological processes including metabolism, cellular homeostasis, and protein turnover. LncRNAs, being large nucleic acids (>200 nt long) can interact with various proteins and other nucleic acids and modulate protein structure and function leading to regulation of cell signaling processes. LncRNAs are widely distributed across cell types and may exhibit tissue specific expression. They are detected in body fluids including blood and urine. Their expressions are also altered in various human diseases including cancer, neurological disorders, immune disorder, and others. LncRNAs are being recognized as novel biomarkers and therapeutic targets. This review article focuses on the emerging role of noncoding RNAs (ncRNAs), particularly long noncoding RNAs (lncRNAs), in the regulation of protein polyubiquitination and proteasomal degradation.
Collapse
Affiliation(s)
- Prarthana Guha
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Avisankar Chini
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Ashcharya Rishi
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Subhrangsu S Mandal
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, United States of America.
| |
Collapse
|
33
|
Nishiyama H, Niinuma T, Kitajima H, Ishiguro K, Yamamoto E, Sudo G, Sasaki H, Yorozu A, Aoki H, Toyota M, Kai M, Suzuki H. HOXA11-As Promotes Lymph Node Metastasis Through Regulation of IFNL and HMGB Family Genes in Pancreatic Cancer. Int J Mol Sci 2024; 25:12920. [PMID: 39684631 DOI: 10.3390/ijms252312920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have shown that long noncoding RNAs (lncRNAs) play pivotal roles in the development and progression of cancer. In the present study, we aimed to identify lncRNAs associated with lymph node metastasis in pancreatic ductal adenocarcinoma (PDAC). We analyzed data from The Cancer Genome Atlas (TCGA) database to screen for genes overexpressed in primary PDAC tumors with lymph node metastasis. Our screen revealed 740 genes potentially associated with lymph node metastasis, among which were multiple lncRNA genes located in the HOXA locus, including HOXA11-AS. Elevated expression of HOXA11-AS was associated with more advanced tumor stages and shorter overall survival in PDAC patients. HOXA11-AS knockdown suppressed proliferation and migration of PDAC cells. RNA-sequencing analysis revealed that HOXA11-AS knockdown upregulated interferon lambda (IFNL) family genes and downregulated high-mobility group box (HMGB) family genes in PDAC cells. Moreover, HMGB3 knockdown suppressed proliferation and migration by PDAC cells. These results suggest that HOXA11-AS contributes to PDAC progression, at least in part, through regulation of IFNL and HMGB family genes and that HOXA11 AS is a potential therapeutic target in PDAC.
Collapse
Affiliation(s)
- Hayato Nishiyama
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Hiroshi Kitajima
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kazuya Ishiguro
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Eiichiro Yamamoto
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Gota Sudo
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan
| | - Hajime Sasaki
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan
| | - Akira Yorozu
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
- Department of Otolaryngology-Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan
| | - Hironori Aoki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Mutsumi Toyota
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masahiro Kai
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|
34
|
Huang J, Yang P, Pan W, Wu F, Qiu J, Ma Z. The role of polypeptides encoded by ncRNAs in cancer. Gene 2024; 928:148817. [PMID: 39098512 DOI: 10.1016/j.gene.2024.148817] [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: 04/12/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
It was previously thought that ncRNA could not encode polypeptides, but recent reports have challenged this notion. As research into ncRNA progresses, it is increasingly clear that it serves roles beyond traditional mechanisms, playing significant regulatory roles in various diseases, notably cancer, which is responsible for 70% of human deaths. Numerous studies have highlighted the diverse regulatory mechanisms of ncRNA that are pivotal in cancer initiation and progression. The role of ncRNA-encoded polypeptides in cancer regulation has gained prominence. This article explores the newly identified regulatory functions of these polypeptides in three types of ncRNA-lncRNA, pri-miRNA, and circRNA. These polypeptides can interact with proteins, influence signaling pathways, enhance miRNA stability, and regulate cancer progression, malignancy, resistance, and other clinical challenges. Furthermore, we discuss the evolutionary significance of these polypeptides in the transition from RNA to protein, examining their emergence and conservation throughout evolution.
Collapse
Affiliation(s)
- Jiayuan Huang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Ping Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118,China
| | - Wei Pan
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Fan Wu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jianhua Qiu
- Department of Anesthesiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201800, China.
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
35
|
Ye C, Xu C, Tang Y, Qi Y, Peng X, Wei G, Jiang L. A novel disulfidptosis-related LncRNA prognostic risk model: predicts the prognosis, tumor microenvironment and drug sensitivity in esophageal squamous cell carcinoma. BMC Gastroenterol 2024; 24:437. [PMID: 39604874 PMCID: PMC11603746 DOI: 10.1186/s12876-024-03530-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Disulfidptosis is a newly discovered type of cell death that differs from apoptosis, necrosis, ferroptosis and other death modes and is closely related to the occurrence and progression of tumors. However, the predictive potential and biological characteristics of disulfidptosis-related lncRNAs (DRGs-lncRNAs) in esophageal squamous cell carcinoma (ESCC) are unclear. METHODS RNA transcriptome data, clinical information and mutation data for ESCC patients were obtained from The Cancer Genome Atlas (TCGA) database. Pearson correlation and Cox regression analyses were used to identify the DRGs-lncRNAs associated with overall survival (OS). LASSO regression analysis was used to construct the prognostic model. A nomogram was created to predict the prognosis of patients with ESCC. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were used to identify the signaling pathways associated with the model. TIMER, CIBERSORT, ESTIMATE and other methods were used to analyze immune infiltration, immune function, immune checkpoints and drug sensitivity. The tumor mutation burden (TMB) were assessed between different risk groups. Real-time polymerase chain reaction (RT‒PCR) was used to detect the expression of DRGs-lncRNAs in ESCC cell lines. RESULTS A total of 155 lncRNAs significantly associated with disulfidptosis were identified. Through univariate Cox regression analysis, LASSO regression analysis and multivariate Cox regression analysis, 9 lncRNAs with independent prognostic significance were selected, and a prognosis model was established. Survival analysis with the prognostic model revealed that there were obvious differences in survival between the high- and low-risk groups. Further analysis revealed that the immune microenvironment, immune infiltration, immune function, immune checkpoints, and drug sensitivity significantly differed between the high-risk and low-risk groups. Patients who exhibited both high risk and high tumor mutation burden (TMB) survived shorter, while those who fell into the low risk and low TMB categories survived longer. In addition, RT‒PCR analysis revealed differential expression of DRG lncRNAs between ESCC cell lines and esophageal epithelial cell lines. CONCLUSIONS We established a DRG-lncRNA prognostic model that can be used to predict the prognosis, tumor mutation burden, immune cell infiltration, and drug sensitivity of ECSS patients. The results of this study provide valuable insights into the understanding of ESCC and provide valuable assistance for the individualized treatment of ESCC patients.
Collapse
Affiliation(s)
- Chunlin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Chuan Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yongchao Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yingcheng Qi
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Xiaoyue Peng
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| | - Lei Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| |
Collapse
|
36
|
Liu X, Jiang D, Liu Y, Xie K, Zhao Y, Liu F. Crispr-Cas9-based long non-coding RNA interference and activation identified that the aberrant expression of Myc-regulated ST8SIA6 antisense RNA 1 promotes tumorigenesis and metastasis in hepatocellular carcinoma. Cytojournal 2024; 21:53. [PMID: 39737136 PMCID: PMC11683396 DOI: 10.25259/cytojournal_109_2024] [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: 06/18/2024] [Accepted: 09/26/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Long non-coding RNAs (lncRNAs) participate in the formation, progression, and metastasis of cancer. This study aimed to explore the roles of the lncRNA ST8SIA6 antisense RNA 1 (ST8SIA6-AS1) in tumorigenesis and elucidate the underlying molecular mechanism of its upregulation in hepatocellular carcinoma (HCC). Material and Methods A total of 56 in-house pairs of HCC tissues were examined, and ST8SIA6-AS1 levels were determined through real-time polymerase chain reaction (PCR). The biological behavior of ST8SIA6-AS1 by Crispr-Cas9-based gene repression and activation was determined in vitro and in vivo. The binding sites and biological behavior of Myc proto-oncogene and forkhead box A on chromatin were investigated through luciferase reporter assays, chromatin immunoprecipitation-quantitative PCR, and co-immunoprecipitation (co-IP) assays. The regulatory mechanisms of ST8SIA6-AS1 expression were analyzed with encyclopedia of DNA elements and gene expression profiling interactive analysis. Results The expression of ST8SIA6-AS1 significantly increased in multiple HCC cell lines and the 56 in-house pairs of HCC tissues (P = 0.0018). Functionally, high-efficiency Crispr-Cas9-based knockdown of ST8SIA6-AS1 revealed that ST8SIA6-AS1 knockdown attenuated the proliferation, migration, and infiltration of HCC cells and considerably reduced the growth rate of subcutaneous and orthotopic HCC tumors. Conversely, ST8SIA6-AS1 overexpression considerably improved the oncogenic characteristics of the HCC cells. Furthermore, ST8SIA6-AS1 upregulation was regulated by the direct binding of transcription factor Myc to the -260 bp to+155 bp and +1003 bp to +1312 bp regions of the ST8SIA6-AS1 transcription start site, which is a segment with high level of H3K27 acetylation. Myc knockdown or treatment with the BET bromodomain inhibitor JQ-1 considerably reduced ST8SIA6-AS1 RNA expression in the HCC cells. Conclusion Our study has established the oncogenic role of ST8SIA6-AS1 and elucidated the Myc-dependent upregulation mechanism of ST8SIA6-AS1 in HCC, providing a profound theoretical molecular basis for the carcinogenic function of ST8SIA6-AS1 in HCC.
Collapse
Affiliation(s)
- Xueqian Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dong Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yang Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kun Xie
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yijun Zhao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fubao Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
37
|
Zhu Z, Xuan W, Wang C, Li C. Long noncoding RNA mediates enzalutamide resistance and transformation in neuroendocrine prostate cancer. Front Oncol 2024; 14:1481777. [PMID: 39655078 PMCID: PMC11625809 DOI: 10.3389/fonc.2024.1481777] [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: 08/16/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Prostate cancer is a malignant tumor caused by the malignant proliferation of epithelial cells, which is highly heterogeneous and drug-resistant, and neuroendocrine prostate cancer (NEPC) is an essential cause of drug resistance in its late stage. Elucidating the evolution of NEPC and the resistance process of enzalutamide, a novel antiandrogen, will be of great help in improving the prognosis of patients. As a research hotspot in the field of molecular biology in recent years, the wide range of biological functions of long noncoding RNAs (lncRNAs) has demonstrated their position in the therapeutic process of many diseases, and a large number of studies have revealed their critical roles in tumor progression and drug resistance. Therefore, elucidating the involvement of lncRNAs in the formation of NEPCs and their interrelationship with enzalutamide resistance may provide new ideas for a deeper understanding of the development of this disease and the occurrence of enzalutamide resistance and give a new direction for reversing the therapeutic dilemma of advanced prostate cancer. This article focuses on lncRNAs that regulate enzalutamide resistance and the neuroendocrine transition of prostate cancer through epigenetic, androgen receptor (AR) signaling, and non-AR pathways that act as "molecular sponges" interacting with miRNAs. Some insights into these mechanisms are used to provide some help for subsequent research in this area.
Collapse
Affiliation(s)
- Zhe Zhu
- Department of Urology, Anhui No.2 Provincial People’s Hospital, HeFei, China
| | - Wenjing Xuan
- Department of Obstetrics, Anhui No.2 Provincial People’s Hospital, HeFei, China
| | - Chaohui Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chancan Li
- Department of Urology, Anhui No.2 Provincial People’s Hospital, HeFei, China
| |
Collapse
|
38
|
Huang Z, Lou K, Qi M, Wang J, Li L, Sun B, Wang C, Zhou X, Chen D, Liu H. RERE-AS1 enhances the effect of CDK4/6 inhibitor Ribociclib and suppresses malignant phenotype in breast cancer via MEK/ERK pathway. J Transl Med 2024; 22:1052. [PMID: 39574120 PMCID: PMC11583401 DOI: 10.1186/s12967-024-05828-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Currently, there is a lack of biomarkers to identify breast cancer (BC) patients who would benefit from CDK4/6 inhibitors. This study combined machine learning (ML) algorithms based on transcriptomic data with both in vivo and in vitro experiments to identify therapeutic efficacy-related biomarkers of the CDK4/6 inhibitor ribociclib from the perspective of long non-coding RNA (lncRNA). METHODS We used the Genomics of Drug Sensitivity in Cancer database along with the "oncoPredict" algorithm to calculate the half maximal inhibitory concentration (IC50) values for ribociclib based on transcriptome data. ML algorithms were utilized to select key lncRNAs related to ribociclib and to establish a model which could be used for selection of potential beneficiaries of ribociclib. Cellular experiments were conducted to validate the ML analysis and explore the potential biological mechanisms by which RERE-AS1 influences ribociclib efficacy and malignant phenotype of BC cells. Correlation analysis with clinical pathological factors, RT-qPCR experiments on tissue specimens, and pan-cancer analysis were carried out to explore the expression pattern, and the prognostic and diagnostic potential of RERE-AS1 in cancers. RESULTS We have identified 11 key ribociclib-related lncRNAs and constructed an artificial neural network model (ANNM) based on lncRNA. Cellular experiments demonstrated that overexpression of RERE-AS1 promoted the anti-tumor activity of ribociclib in BC cells. Furthermore, RERE-AS1 is crucial in suppressing the malignant traits of BC cells through the reduction of MEK and ERK phosphorylation levels. Patients with smaller primary tumors and lower pathological stage exhibited higher levels of RERE-AS1 expression. Lastly, a pan-cancer analysis revealed that RERE-AS1 exhibits distinctly abnormal expression patterns, prognostic significance, and clinical diagnostic value in BC, compared to other cancers. CONCLUSIONS The ANNM established through ML algorithms can serve as predictive indicators for the efficacy of ribociclib in BC patients. LncRNA RERE-AS1, a newly discovered biomarker, holds significant promise for diagnosis, treatment, and enhancing the therapeutic response to ribociclib in BC.
Collapse
Affiliation(s)
- Zhidong Huang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kaixin Lou
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Mengyang Qi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinhui Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Linwei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bo Sun
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chen Wang
- The School of Basic Medicine, Tianjin Medical University, Tianjin, China
| | - Xirui Zhou
- The School of Basic Medicine, Tianjin Medical University, Tianjin, China
| | - Debo Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China.
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China.
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
39
|
Tsai KW, Liao JB, Tseng HW. Metformin regulates the proliferation and motility of melanoma cells by modulating the LINC00094/miR-1270 axis. Cancer Cell Int 2024; 24:384. [PMID: 39563323 PMCID: PMC11575040 DOI: 10.1186/s12935-024-03545-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Melanoma is an aggressive tumor with a high mortality rate. Metformin, a commonly prescribed diabetes medication, has shown promise in cancer prevention and treatment. Long noncoding RNAs (lncRNAs) are non-protein-coding RNA molecules that play a key role in tumor development by interacting with cellular chromatins. Despite the benefits of metformin, the anticancer mechanism underlying its effect on the regulation of lncRNAs in melanoma remains unclear. METHODS We investigated the lncRNA profiles of human melanoma cells with and without metformin treatment using a next-generation sequencing approach (NGS). Utilizing public databases, we analyzed the expression levels and clinical impacts of LINC00094 and miR-1270 in melanoma. The expression levels of LINC00094 and miR-1270 were verified in human cell lines and clinical samples by real-time PCR and in situ hybridization. The biological roles of LINC00094 and miR-1270 in cell growth, proliferation, cell cycle, apoptosis, and motility were studied using in vitro assays. RESULTS We identify a novel long noncoding RNA, namely LINC00094, whose expression considerably decreased in melanoma cells after metformin treatment. In situ hybridization analysis revealed substantially higher expression of LINC00094 in cutaneous melanoma tissue compared with adjacent normal epidermis and normal control tissues (P < 0.001). In nondiabetic patients with melanoma, the overall survival of high LINC00094 expression group was shorter than the low LINC00094 expression group with borderline statistical significance (log-rank test, P = 0.057). Coexpression analysis of LINC00094 indicated its involvement in the mitochondrial respiratory pathway, with its knockdown suppressing genes associated with mitochondrial oxidative phosphorylation, glycolysis, antioxidant production, and metabolite levels. Functional analysis revealed that silencing-LINC00094 inhibited the proliferation, colony formation, invasion, and migration of melanoma cells. Cell cycle analysis following LINC00094 knockdown revealed G1 phase arrest with reduced cell cycle protein expression. Combined TargetScan and reporter assays revealed a direct link between miR-1270 and LINC00094. Ectopic miR-1270 expression inhibited melanoma cell growth and motility while inducing apoptosis. Finally, through in silico analysis, we identified two miR-1270 target genes, CD276 and centromere protein M (CENPM), which may be involved in the biological functions of LINC00094. CONCLUSIONS Overall, LINC00094 expression may regulate melanoma cell growth and motility by modulating the expression of miR-1270, and targeting genes of CD276 and CENPM indicating its therapeutic potential in melanoma treatment.
Collapse
Affiliation(s)
- Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City, Taiwan
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Shu Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hui-Wen Tseng
- Department of Dermatology, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan.
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- Department of Nursing, College of Nursing, Meiho University, Neipu, Pingtung, Taiwan.
| |
Collapse
|
40
|
Zhou M, Tang J, Huang G, Hong L. Prognostic Significance and Immune Landscape of a Cuproptosis-Related LncRNA Signature in Ovarian Cancer. Biomedicines 2024; 12:2640. [PMID: 39595204 PMCID: PMC11592286 DOI: 10.3390/biomedicines12112640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Cuproptosis is a copper-induced mitochondrial cell death, and regulating cuproptosis is becoming a rising cancer treatment modality. Here, we attempted to establish a cuproptosis-associated lncRNAs (CRLs) signature (CRlncSig) to predict the survival, immune landscape, and treatment response in ovarian cancer (OC) patients. Methods: A series of statistical analyses were used to identify the key CRLs that are closely related to the prognosis, and a prognostic CRlncSig was constructed. The predictive accuracy of the CRlncSig was further validated in an independent Gene Expression Omnibus (GEO) set. Then, we compared the immune cell infiltration, immune checkpoints, tumor microenvironment (TME), tumor mutational burden (TMB), drug sensitivity, and efficacy of immunotherapy between the two subgroups. We further built a nomogram integrating the CRlncSig and different clinical traits to enhance the clinical application of the CRlncSig. Results: Nine hub CRLs, namely RGMB-AS1, TYMSOS, DANCR, LINC00702, LINC00240, LINC00996, DNM1P35, LINC00892, and TMEM254-AS1, were correlated with the overall survival (OS) of OC and a prognostic CRlncSig was established. The CRlncSig classified OC patients into two risk groups with strikingly different survival probabilities. The time-dependent ROC (tdROC) curves demonstrated good predictive ability in both the training cohort and an independent validation cohort. Multivariate analysis confirmed the independent predictive performance of the CRlncSig. We constructed a nomogram based on the CRlncSig, which can predict the prognosis of OC patients. The high-risk score was characterized by decreased immune cell infiltration and activation of stroma, while activation of immunity was observed in the low-risk subgroup. Moreover, patients in low-risk subgroups had more Immunophenoscore (IPS) and fewer immune escapes compared to high-risk subgroups. Finally, an immunotherapeutic cohort confirmed the value of the CRlncSig in predicting immunotherapy outcomes. Conclusions: The developed CRlncSig may be promising for the clinical prediction of OC patient outcomes and immunotherapeutic responses.
Collapse
Affiliation(s)
| | | | | | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (M.Z.)
| |
Collapse
|
41
|
Yang TF, Li XR, Kong MW. Molecular mechanisms underlying roles of long non-coding RNA small nucleolar RNA host gene 16 in digestive system cancers. World J Gastrointest Oncol 2024; 16:4300-4308. [PMID: 39554746 PMCID: PMC11551640 DOI: 10.4251/wjgo.v16.i11.4300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 10/25/2024] Open
Abstract
This editorial reviews the molecular mechanisms underlying the roles of the long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in digestive system cancers based on two recent studies on lncRNAs in digestive system tumors. The first study, by Zhao et al, explored how hBD-1 affects colon cancer, via the lncRNA TCONS_00014506, by inhibiting mTOR and promoting autophagy. The second one, by Li et al, identified the lncRNA prion protein testis specific (PRNT) as a factor in oxaliplatin resistance by sponging ZNF184 to regulate HIPK2 and influence colorectal cancer progression and chemoresistance, suggesting PRNT as a potential therapeutic target for colorectal cancer. Both of these two articles discuss the mechanisms by which lncRNAs contribute to the development and progression of digestive system cancers. As a recent research hotspot, SNHG16 is a typical lncRNA that has been extensively studied for its association with digestive system cancers. The prevailing hypothesis is that SNHG16 participates in the development and progression of digestive system tumors by acting as a competing endogenous RNA, interacting with other proteins, regulating various genes, and affecting downstream target molecules. This review systematically examines the recently reported biological functions, related molecular mechanisms, and potential clinical significance of SNHG16 in various digestive system cancers, and explores the relationship between SNHG16 and digestive system cancers. The findings suggest that SNHG16 may serve as a potential biomarker and therapeutic target for human digestive system cancers.
Collapse
Affiliation(s)
- Ting-Fang Yang
- Department of Oncology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| | - Xin-Rui Li
- Department of Cardiology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| | - Mo-Wei Kong
- Department of Cardiology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| |
Collapse
|
42
|
Che H, Zhang X, Cao L, Huang W, Lu Q. LINC01614 Promotes Oral Squamous Cell Carcinoma by Regulating FOXC1. Genes (Basel) 2024; 15:1461. [PMID: 39596660 PMCID: PMC11593781 DOI: 10.3390/genes15111461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are pivotal mediators during the development of carcinomas; however, it remains to be investigated whether lncRNAs are implicated in oral squamous cell carcinoma (OSCC). Methods: In this study, quantitative real-time PCR was conducted for detecting the expression of LINC01614 in OSCC cell lines. The biological functions of LINC01614 were assessed by loss- and gain-of-function experiments conducted both in vivo and in vitro. Cellular proliferation, migration, and invasion were investigated herein, and dual luciferase reporter assays were additionally performed to explore the relationships among LINC01614, miR-138-5p, and Forkhead box C1 (FOXC1). Results: The research presented herein revealed that OSCC cells express high levels of LINC01614. Functional experiments employing cellular and animal models demonstrated that LINC01614 knockdown repressed the malignant phenotypes of OSCC cells, including their growth, invasiveness, and migration. Further investigation revealed that LINC01614 absorbs miR-138-5p miRNA by functioning as a competing endogenous RNA to downregulate the abundance of FOXC1. Conclusions: The findings revealed that LINC01614 contributes to the progression of OSCC by targeting the FOXC1 signaling pathway. The study provides insights into a novel mechanistic process to regulate the development of OSCC, and established a possible target for the therapeutic management of OSCC.
Collapse
Affiliation(s)
- Hongze Che
- School of Dentistry, Beihua University, Jilin 132013, China
| | - Xun Zhang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Luo Cao
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Wenjun Huang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Qing Lu
- VIP Integrated Department, Stomatological Hospital, Jilin University, Changchun 130021, China
| |
Collapse
|
43
|
Hussain MA, Elemam NM, Talaat IM. Androgen Receptor and Non-Coding RNAs' Interaction in Renal Cell Carcinoma. Noncoding RNA 2024; 10:56. [PMID: 39585048 PMCID: PMC11587015 DOI: 10.3390/ncrna10060056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Renal cell carcinoma (RCC), the most prevalent among the urogenital cancers, accounts for around 3% of new cancer cases worldwide. Significantly, the incidence of RCC has doubled in developed world countries, ranking it as the sixth most common cancer in males, who represent two-thirds of RCC cases. Males with RCC exhibit a higher mortality rate and tend to develop a more aggressive form of the disease than females. Sex-related risk factors, including lifestyle and biological variations, explain this difference. The androgen receptor (AR) oncogenic signaling pathway has been extensively studied among the biological factors that affect RCC. Recent advancements in high-throughput RNA sequencing techniques have underscored the significant roles played by noncoding-RNAs (ncRNAs), previously dismissed as "junk". The oncogenic potential of AR is manifested through its dysregulation of the ncRNAs' availability and function, promoting RCC tumorigenesis. This review offers a summary of the most recent findings on the role and molecular mechanisms of the AR in dysregulating the ncRNAs that play a role in the progression of RCC and the possibility of utilizing ncRNAs to target AR as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Manal A. Hussain
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Pure Lab North, Purelab, Abu Dhabi 134808, United Arab Emirates
| | - Noha M. Elemam
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Iman M. Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
| |
Collapse
|
44
|
Wang Y, Shen K, Cheng Q, Zhou X, Liu K, Xiao J, Hu L. The long noncoding RNA ELFN1-AS1 promotes gastric cancer growth and metastasis by interacting with TAOK1 to inhibit the Hippo signaling pathway. Cell Death Discov 2024; 10:465. [PMID: 39528458 PMCID: PMC11555383 DOI: 10.1038/s41420-024-02235-5] [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: 10/26/2023] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is a common digestive malignancy that causes numerous cancer-related deaths. Long noncoding RNAs (lncRNAs) play a crucial role in the development of various tumors, including GC. In this study, we revealed that ELFN1-AS1, a lncRNA with aberrantly high expression, contributes to the proliferation and metastasis of GC. Mechanically, ELFN1-AS1 plays an oncogenic role by binding to the protein kinase domain of thousand and one amino acid protein kinase (TAOK1), a tumor suppressor in GC, and disrupting the TAOK1-STK3 interaction, leading to decreased STK3 phosphorylation. This decrease is accompanied by attenuation of the Hippo kinase cascade, resulting in reduced YAP1 phosphorylation, a crucial effector of the Hippo signaling pathway. Subsequently, the reduced YAP1 phosphorylation promotes its nuclear translocation, thereby enhancing the expression of MYC, a downstream target of the pathway and well-known oncogene. Taken together, the ELFN1-AS1/TAOK1/STK3/YAP1 axis may promote GC progression and is a promising target for GC treatment.
Collapse
Affiliation(s)
- Yuanhang Wang
- Department of General Surgery, Yancheng Third People's Hospital, Affiliated Yancheng Hospital, School of Medicine, Southeast University, Yancheng, Jiangsu Province, China.
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Kuan Shen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of General Surgery, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Liyang, Jiangsu Province, China
| | - Quan Cheng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xinyi Zhou
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kanghui Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jian Xiao
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Hu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
45
|
Sun M, Zhan N, Yang Z, Zhang X, Zhang J, Peng L, Luo Y, Lin L, Lou Y, You D, Qiu T, Liu Z, Wang Q, Liu Y, Sun P, Yu M, Wang H. Cuproptosis-related lncRNA JPX regulates malignant cell behavior and epithelial-immune interaction in head and neck squamous cell carcinoma via miR-193b-3p/PLAU axis. Int J Oral Sci 2024; 16:63. [PMID: 39511134 PMCID: PMC11543849 DOI: 10.1038/s41368-024-00314-y] [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: 01/26/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 11/15/2024] Open
Abstract
The development, progression, and curative efficacy of head and neck squamous cell carcinoma (HNSCC) are influenced by complex interactions between epithelial and immune cells. Nevertheless, the specific changes in the nature of these interactions and their underlying molecular mechanisms in HNSCC are not yet fully understood. Cuproptosis, a form of programmed cell death that is dependent on copper, has been implicated in cancer pathogenesis. However, the understanding of cuproptosis in the context of HNSCC remains limited. In this study, we have discovered that cuproptosis-related long non-coding RNAs (CRLs) known as JPX play a role in promoting the expression of the oncogene urokinase-type plasminogen activator (PLAU) by competitively binding to miR-193b-3p in HNSCC. The increased activity of the JPX/miR-193b-3p/PLAU axis in malignant epithelial cells leads to enhanced cell proliferation, migration, and invasion in HNSCC. Moreover, the overexpression of PLAU in tumor epithelial cells facilitates its interaction with the receptor PLAUR, predominantly expressed on macrophages, thereby influencing the abnormal epithelial-immune interactome in HNSCC. Notably, the JPX inhibitor Axitinib and the PLAU inhibitor Palbociclib may not only exert their effects on the JPX/miR-193b-3p/PLAU axis that impacts the malignant tumor behaviors and the epithelial-immune cell interactions but also exhibit synergistic effects in terms of suppressing tumor cell growth and arresting cell cycle by targeting epidermal growth factor receptor (EGFR) and cyclin-dependent kinase (CDK4/6) for the treatment of HNSCC.
Collapse
Affiliation(s)
- Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Ning Zhan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Zhan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Xiaoting Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jingyu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Lianjie Peng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yaxian Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Lining Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yiting Lou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Dongqi You
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Tao Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Zhichao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Qianting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yu Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Ping Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| |
Collapse
|
46
|
Al-Noshokaty TM, Abdelhamid R, Reda T, Alaaeldien A, Abdellatif N, Mansour A, Gendi D, Abdelmaksoud NM, Elshaer SS, Doghish AS, Sobhy MH, Mohammed OA, Abulsoud AI. Exploring the clinical potential of circulating LncRNAs in breast cancer: insights into primary signaling pathways and therapeutic interventions. Funct Integr Genomics 2024; 24:209. [PMID: 39508907 DOI: 10.1007/s10142-024-01476-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: 08/18/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024]
Abstract
Breast cancer (BC) occupies the top spot among women on a global scale. The tumor has a significant degree of heterogeneity, displaying a notable prevalence of medication resistance, recurrence, and metastasis, rendering it one of the most lethal forms of malignant neoplasms. The timely identification, ongoing evaluation of therapeutic interventions, and accurate prediction of outcomes play crucial roles in determining the overall survival rates of women with BC. Nevertheless, the absence of precise biomarkers remains a significant determinant impacting the overall well-being and both the physical and emotional health of BC patients. Long noncoding RNA (lncRNA) exerts regulatory control over several genes and signaling pathways, hence assuming crucial roles in the development of neoplastic growth. Recently, research has indicated that the atypical expression of circulating lncRNAs in various biological bodily fluids has a noteworthy impact on the early detection, pathological categorization, staging, monitoring of therapy outcomes, and evaluation of prognosis in cases of BC. This article aims to assess the potential clinical utility of circulating lncRNAs in the context of BC focusing on specific primary signaling pathways; Wnt/β-catenin, Notch, TGF-β, and hedgehog (Hh), in addition to some therapeutic interventions.
Collapse
Affiliation(s)
- Tohada M Al-Noshokaty
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Rehab Abdelhamid
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Tasnim Reda
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Ayat Alaaeldien
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Nourhan Abdellatif
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Abdallah Mansour
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - David Gendi
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11823, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo, 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Mohamed Hossam Sobhy
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Department of Chemistry, University of Tennessee, Knoxville, TN, 37996, USA
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
| |
Collapse
|
47
|
Tan W, Xiao C, Ma M, Cao Y, Huang Z, Wang X, Kang R, Li Z, Li E. Role of non-coding RNA in lineage plasticity of prostate cancer. Cancer Gene Ther 2024:10.1038/s41417-024-00834-z. [PMID: 39496938 DOI: 10.1038/s41417-024-00834-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 11/06/2024]
Abstract
The treatment of prostate cancer (PCa) has made great progress in recent years, but treatment resistance always develops and can even lead to fatal disease. Exploring the mechanism of drug resistance is of great significance for improving treatment outcomes and developing biomarkers with predictive value. It is increasingly recognized that mechanism of drug resistance in advanced PCa is related to lineage plasticity and tissue differentiation. Specifically, one of the mechanisms by which castration-resistant prostate cancer (CRPC) cells acquire drug resistance and transform into neuroendocrine prostate cancer (NEPC) cells is lineage plasticity. NEPC is a subtype of PCa that is highly aggressive and lethal, with a median survival of only 7 months. With the development of high-throughput RNA sequencing technology, more and more non-coding RNAs have been identified, which play important roles in different diseases through different mechanisms. Several ncRNAs have shown great potential in PCa lineage plasticity and as biomarkers. In the review, the role of ncRNA in PCa lineage plasticity and its use as biomarkers were reviewed.
Collapse
Affiliation(s)
- Wenhui Tan
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Changkai Xiao
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Min Ma
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Youhan Cao
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhenguo Huang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaolan Wang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ran Kang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhenfa Li
- Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China.
| | - Ermao Li
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
48
|
Cen J, Wang L, Zhang H, Guo Y. Analysis of the prognostic significance and potential mechanisms of lncRNAs related to m6A methylation in laryngeal cancer. Biotechnol Genet Eng Rev 2024; 40:2129-2154. [PMID: 37053494 DOI: 10.1080/02648725.2023.2198630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
Objective to investigate the prognostic significance and potential mechanism analysis of m6A methylation-associated lncRNAs in laryngeal cancer. Methods based on the expression of m6A-associated lncRNAs, the samples were divided into two clusters and least absolute value and selection operator (LASSO) regression analysis was performed to build and validate prognostic models. In addition, the relationships between risk scores, clusters, arginine synthase (SMS), tumor microenvironment, clinicopathological features, immune infiltration, immune checkpoints, and tumor mutation burden were analyzed. Finally, the relationship between SMS and m6A-associated IncRNAs was analyzed and SMS-associated pathways were enriched by gene set enrichment analysis (GSEA). Results a total of 95 lncRNAs were associated with the expression of 22 m6A methylation regulators in laryngeal cancer, 14 of which were prognostic lncRNAs. These lncRNAs were divided into two clusters and evaluated. Clinicopathological features did not show significant differences. However, the two clusters differed significantly in terms of naive B cells, memory B cells, naive CD4 T cells, T helper cells and immune score. lASSO regression analysis showed that risk score was a significant predictor of progression-free survival. Conclusion low expression of m6A-related lncRNAs involved in laryngeal cancer development in laryngeal cancer tissues can be used as an indicator to diagnose patients with laryngeal cancer, reduce patient prognosis, be an independent risk factor affecting patient prognosis and be able to assess patient prognosis.
Collapse
Affiliation(s)
- Jingtu Cen
- Shanghai Municipal Hospital of Traditional Chinese Medicine,ear-nose-throat department, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihua Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine,ear-nose-throat department, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haopeng Zhang
- Shanghai Municipal Hospital of Traditional Chinese Medicine,ear-nose-throat department, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Guo
- Shanghai Municipal Hospital of Traditional Chinese Medicine,ear-nose-throat department, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
49
|
Duan R, Zhai Y, Wang Q, Zhao L, Wang Y, Yu N, Zhang J, Guo W. LINC01764 promotes colorectal cancer cells proliferation, metastasis, and 5-fluorouracil resistance by regulating glucose and glutamine metabolism via promoting c-MYC translation. MedComm (Beijing) 2024; 5:e70003. [PMID: 39534558 PMCID: PMC11555016 DOI: 10.1002/mco2.70003] [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: 11/25/2023] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 11/16/2024] Open
Abstract
Few biomarkers are available for predicting chemotherapeutic response and prognosis in colorectal cancer (CRC). Long-noncoding RNAs (lncRNAs) are essential for CRC development and growth. Therefore, studying lncRNAs may reveal potential predictors of chemotherapy response and prognosis in CRC. LINC01764 was analyzed using datasets from Fudan University Shanghai Cancer Center's advanced CRC patients' RNA-seq and The Cancer Genome Atlas datasets. Gene set enrichment analysis was employed to detect related pathways. Cotransfection experiments, RNA pulldown assays, RNA-binding protein immunoprecipitation, protein synthesis activity, and dual-luciferase reporter assays were performed to determine interactions among LINC01764, hnRNPK, and c-MYC. High LINC01764 expression correlates with metastasis, a poor response to FOLFOX/XELOX chemotherapy, and a poor prognosis in CRC. LINC01764 enhance glycolysis and glutamine metabolism to promote CRC cells proliferation, metastasis, and 5-fluorouracil (5-FU) resistance. LINC01764 specifically binds to hnRNPK, facilitating its interaction with c-MYC mRNA and promoting internal ribosome entry site (IRES)-dependent translation of c-MYC, thereby exerting oncogenic effects. LINC01764 induced 5-FU chemoresistance by upregulating the c-MYC, glucose, and glutamine metabolism pathways, which downregulated UPP1, crucial for activating 5-FU. Conclusively, LINC01764 promotes CRC progression and 5-FU resistance through hnRNPK-mediated-c-MYC IRES-dependent translational regulation, which suggests its potential as a predictor of CRC chemotherapy response and prognosis.
Collapse
Affiliation(s)
- Ran Duan
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Medical OncologyFujian Cancer Hospital and Fujian Medical University Cancer HospitalFujian Medical UniversityFuzhouChina
| | - Yujia Zhai
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qiushuang Wang
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Liqin Zhao
- Department of OncologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yixuan Wang
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Nuoya Yu
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jieyun Zhang
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Weijian Guo
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
50
|
Luo S, Yue M, Wang D, Lu Y, Wu Q, Jiang J. Breaking the barrier: Epigenetic strategies to combat platinum resistance in colorectal cancer. Drug Resist Updat 2024; 77:101152. [PMID: 39369466 DOI: 10.1016/j.drup.2024.101152] [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: 05/22/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Platinum-based drugs, such as cisplatin and oxaliplatin, are frontline chemotherapy for CRC, effective in both monotherapy and combination regimens. However, the clinical efficacy of these treatments is often undermined by the development of drug resistance, a significant obstacle in cancer therapy. In recent years, epigenetic alterations have been recognized as key players in the acquisition of resistance to platinum drugs. Targeting these dysregulated epigenetic mechanisms with small molecules represents a promising therapeutic strategy. This review explores the complex relationship between epigenetic changes and platinum resistance in CRC, highlighting current epigenetic therapies and their effectiveness in countering resistance mechanisms. By elucidating the epigenetic underpinnings of platinum resistance, this review aims to contribute to ongoing efforts to improve treatment outcomes for CRC patients.
Collapse
Affiliation(s)
- Shiwen Luo
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ming Yue
- Department of Pharmacy, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Dequan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yukang Lu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jue Jiang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|