1
|
Wang J, Zhang W, Zhang R, Yang H, Li Y, Wang J, Li C. MiR-101-3p Promotes Tumor Cell Proliferation and Migration via the Wnt Signal Pathway in MNNG-Induced Esophageal Squamous Cell Carcinoma. TOXICS 2024; 12:824. [PMID: 39591002 PMCID: PMC11598764 DOI: 10.3390/toxics12110824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
N-methyl-n'-nitroso-n'-nitroso guanidine (MNNG) can induce esophageal squamous cell carcinoma (ESCC), and microRNAs are associated with the development of ESCC and may serve as potential tumor prognostic markers. Thus, the aim of this study was to evaluate the potential function of miR-101-3p in MNNG-induced ESCC. An investigation of risk factors in patients with ESCC was carried out and the concentration of nine nitrosamines in urine samples was detected by the SPE-GC-MS technique. Then, we performed cancer tissue gene sequencing analysis, and RT-qPCR verified the expression level of miR-101-3p. Subsequently, the relationship between miR-101-3p potential target genes and the ESCC patients' prognosis was predicted. Finally, we investigated the function of miR-101-3p in MNNG-induced ESCC pathogenesis and the regulatory mechanism of the signaling pathway by in vivo and in vitro experiments. The results revealed that high dietary nitrosamine levels are high-risk factors for ESCC. MiR-101-3p is down-regulated in ESCC tissues and cells, and its potential target genes are enriched in cell migration and cancer-related pathways. MiR-101-3p target genes include AXIN1, CK1, and GSK3, which are involved in the regulation of the Wnt signaling pathway. MiR-101-3p overexpression promotes apoptosis and inhibits the proliferation and migration of Eca109 cells. The Wnt pathway is activated after subchronic exposure to MNNG, and the Wnt pathway is inhibited by the overexpression of miR-101-3p in Eca109 cells. Down-regulated miR-101-3p may exert tumor suppressive effects by regulating the Wnt pathway and may be a useful biomarker for predicting ESCC progression.
Collapse
Affiliation(s)
- Jianding Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China; (J.W.); (W.Z.); (Y.L.); (J.W.)
| | - Wenwen Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China; (J.W.); (W.Z.); (Y.L.); (J.W.)
| | - Rui Zhang
- Key Laboratory for Reproductive Medicine and Embryo, The Reproductive Medicine Special Hospital of the Lanzhou University First Affiliated Hospital, Lanzhou 730000, China;
| | - Hanteng Yang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China;
| | - Yitong Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China; (J.W.); (W.Z.); (Y.L.); (J.W.)
| | - Junling Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China; (J.W.); (W.Z.); (Y.L.); (J.W.)
| | - Chengyun Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China; (J.W.); (W.Z.); (Y.L.); (J.W.)
| |
Collapse
|
2
|
Jiang T, Shen C, Ding P, Luo L. Data augmentation based on the WGAN-GP with data block to enhance the prediction of genes associated with RNA methylation pathways. Sci Rep 2024; 14:26321. [PMID: 39487188 PMCID: PMC11530642 DOI: 10.1038/s41598-024-77107-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
RNA methylation modification influences various processes in the human body and has gained increasing attention from scholars. Predicting genes associated with RNA methylation pathways can significantly aid biologists in studying RNA methylation processes. Several prediction methods have been investigated, but their performance is still limited by the scarcity of positive samples. To address the challenge of data imbalance in RNA methylation-associated gene prediction tasks, this study employed a generative adversarial network to learn the feature distribution of the original dataset. The quality of synthetic samples was controlled using the Classifier Two-Sample Test (CTST). These synthetic samples were then added to the data blocks to mitigate class distribution imbalance. Experimental results demonstrated that integrating the synthetic samples generated by our proposed model with the original data enhances the prediction performance of various classifiers, outperforming other oversampling methods. Moreover, gene ontology (GO) enrichment analyses further demonstrate the effectiveness of the predicted genes associated with RNA methylation pathways. The model generating gene samples with PyTorch is available at https://github.com/heyheyheyheyhey1/WGAN-GP_RNA_methylation.
Collapse
Affiliation(s)
- Tuo Jiang
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China
| | - Cong Shen
- Department of Mathematics, National University of Singapore, Singapore, 119076, Singapore
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China.
| | - Lingyun Luo
- School of Computer Science, University of South China, Hengyang, 421001, Hunan, China.
- Hunan Medical Big Data International Science and Technology Innovation Cooperation Base, Hengyang, 421001, Hunan, China.
| |
Collapse
|
3
|
Yuan L, Meng Y, Xiang J. SNX16 is required for hepatocellular carcinoma survival via modulating the EGFR-AKT signaling pathway. Sci Rep 2024; 14:13093. [PMID: 38849490 PMCID: PMC11161632 DOI: 10.1038/s41598-024-64015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Sorting nexin 16 (SNX16), a pivotal sorting nexin, emerges in tumor progression complexity, fueling research interest. However, SNX16's biological impact and molecular underpinnings in hepatocellular carcinoma (HCC) remain elusive. This study probes SNX16's function, clinical relevance via mRNA, and protein expression in HCC. Overexpression/knockdown assays of SNX16 were employed to elucidate impacts on HCC cell invasion, proliferation, and EMT. Additionally, the study delved into SNX16's regulation of the EGFR-AKT signaling cascade mechanism. SNX16 overexpression in HCC correlates with poor patient survival; enhancing proliferation, migration, invasion, and tumorigenicity, while SNX16 knockdown suppresses these processes. SNX16 downregulation curbs phospho-EGFR, dampening AKT signaling. EGFR suppression counters SNX16-overexpression-induced HCC proliferation, motility, and invasiveness. Our findings delineate SNX16's regulatory role in HCC, implicating it as a prospective therapeutic target.
Collapse
Affiliation(s)
- Lebin Yuan
- Department of General Surgery, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, China
| | - Yanqiu Meng
- Oncology Department, First Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jiajia Xiang
- Molecular Centre Laboratory, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
4
|
Song Z, Cao X, Wang X, Li Y, Zhang W, Wang Y, Chen L. A disulfidptosis-related lncRNA signature for predicting prognosis and evaluating the tumor immune microenvironment of lung adenocarcinoma. Sci Rep 2024; 14:4621. [PMID: 38409243 PMCID: PMC10897395 DOI: 10.1038/s41598-024-55201-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
As a novel form of regulated cell death (RCD), disulfidptosis offering a significant opportunity in better understanding of tumor pathogenesis and therapeutic strategies. Long non-coding RNAs (lncRNAs) regulate the biology functions of tumor cells by engaging with a range of targets. However, the prognostic value of disulfidptosis-related lncRNAs (DRlncRNAs) in lung adenocarcinoma (LUAD) remains unclear. Therefore, our study aimed at establishing a prognostic model for LUAD patients based on DRlncRNAs. RNA-seq data and clinical information were obtained from The Cancer Genome Atlas (TCGA) database. Subsequently, a prognostic model based on DRlncRNAs was constructed using LASSO and COX regression analysis. Patients were stratified into high- and low-risk groups based on their risk scores. Differences between the high-risk and low-risk groups were investigated in terms of overall survival (OS), functional enrichment, tumor immune microenvironment (TIME), somatic mutations, and drug sensitivity. Finally, the role of lncRNA GSEC in LUAD was validated through in vitro experiments. Using the prognostic model consists of 5 DRlncRNAs (AL365181.2, GSEC, AC093673.1, AC012615.1, AL606834.1), the low-risk group exhibited a markedly superior survival in comparison to the high-risk group. The significant differences were observed among patients from different risk groups in OS, immune cell infiltration, immune checkpoint expression, immunotherapy response, and mutation landscape. Experimental results from cellular studies demonstrate the knockdown of lncRNA GSEC leading to a significant reduction in the proliferation and migration abilities of LUAD cells. Our prognostic model, constructed using 5 DRlncRNAs, exhibited the capacity to independently predict the survival of LUAD patients, providing the potentially significant assistance in prognosis prediction, and treatment effects optimization. Moreover, our study established a foundation for further research on disulfidptosis in LUAD and proposed new perspectives for the treatment of LUAD.
Collapse
Affiliation(s)
- Zipei Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xincen Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaokun Wang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuting Li
- Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Weiran Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuheng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Zabeti Touchaei A, Vahidi S, Samadani AA. Decoding the regulatory landscape of lncRNAs as potential diagnostic and prognostic biomarkers for gastric and colorectal cancers. Clin Exp Med 2024; 24:29. [PMID: 38294554 PMCID: PMC10830721 DOI: 10.1007/s10238-023-01260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024]
Abstract
Colorectal cancer (CRC) and gastric cancer (GC) are major contributors to cancer-related mortality worldwide. Despite advancements in understanding molecular mechanisms and improved drug treatments, the overall survival rate for patients remains unsatisfactory. Metastasis and drug resistance are major challenges contributing to the high mortality rate in both CRC and GC. Recent research has shed light on the role of long noncoding RNAs (lncRNAs) in the development and progression of these cancers. LncRNAs regulate gene expression through various mechanisms, including epigenetic modifications and interactions with microRNAs (miRNAs) and proteins. They can serve as miRNA precursors or pseudogenes, modulating gene expression at transcriptional and post-transcriptional levels. Additionally, circulating lncRNAs have emerged as non-invasive biomarkers for the diagnosis, prognosis, and prediction of drug therapy response in CRC and GC. This review explores the intricate relationship between lncRNAs and CRC/GC, encompassing their roles in cancer development, progression, and chemoresistance. Furthermore, it discusses the potential of lncRNAs as therapeutic targets in these malignancies. The interplay between lncRNAs, miRNAs, and tumor microenvironment is also highlighted, emphasizing their impact on the complexity of cancer biology. Understanding the regulatory landscape and molecular mechanisms governed by lncRNAs in CRC and GC is crucial for the development of effective diagnostic and prognostic biomarkers, as well as novel therapeutic strategies. This review provides a comprehensive overview of the current knowledge and paves the way for further exploration of lncRNAs as key players in the management of CRC and GC.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
6
|
Tang Z, Li X, Zheng Y, Liu J, Liu C, Li X. The role of competing endogenous RNA network in the development of hepatocellular carcinoma: potential therapeutic targets. Front Cell Dev Biol 2024; 12:1341999. [PMID: 38357004 PMCID: PMC10864455 DOI: 10.3389/fcell.2024.1341999] [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: 11/30/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
The current situation of hepatocellular carcinoma (HCC) management is challenging due to its high incidence, mortality, recurrence and metastasis. Recent advances in gene genetic and expression regulation have unveiled the significant role of non-coding RNA (ncRNA) in various cancers. This led to the formulation of the competing endogenous RNA (ceRNA) hypothesis, which posits that both coding RNA and ncRNA, containing miRNA response elements (MRE), can share the same miRNA sequence. This results in a competitive network between ncRNAs, such as lncRNA and mRNA, allowing them to regulate each other. Extensive research has highlighted the crucial role of the ceRNA network in HCC development, impacting various cellular processes including proliferation, metastasis, cell death, angiogenesis, tumor microenvironment, organismal immunity, and chemotherapy resistance. Additionally, the ceRNA network, mediated by lncRNA or circRNA, offers potential in early diagnosis and prevention of HCC. Consequently, ceRNAs are emerging as therapeutic targets for HCC. The complexity of these gene networks aligns with the multi-target approach of traditional Chinese medicine (TCM), presenting a novel perspective for TCM in combating HCC. Research is beginning to show that TCM compounds and prescriptions can affect HCC progression through the ceRNA network, inhibiting proliferation and metastasis, and inducing apoptosis. Currently, the lncRNAs TUG1, NEAT1, and CCAT1, along with their associated ceRNA networks, are among the most promising ncRNAs for HCC research. However, this field is still in its infancy, necessitating advanced technology and extensive basic research to fully understand the ceRNA network mechanisms of TCM in HCC treatment.
Collapse
Affiliation(s)
- Ziwei Tang
- The Ninth People’s Hospital of Chongqing, Chongqing, China
| | - Xue Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanfeng Zheng
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Jin Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Liu
- Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Xia Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Liu P, Zhou L, Chen H, He Y, Li G, Hu K. Identification of a novel intermittent hypoxia-related prognostic lncRNA signature and the ceRNA of lncRNA GSEC/miR-873-3p/EGLN3 regulatory axis in lung adenocarcinoma. PeerJ 2023; 11:e16242. [PMID: 37842058 PMCID: PMC10573295 DOI: 10.7717/peerj.16242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is still the most prevalent type of respiratory cancer. Intermittent hypoxia can increase the mortality and morbidity associated with lung cancer. Long non-coding RNAs (lncRNAs) are crucial in lung adenocarcinoma. However, the effects of intermittent hypoxia-related long non-coding RNAs (IHRLs) on lung adenocarcinoma are still unknown. Method In the current research, eight IHRLs were selected to create a prognostic model. The risk score of the prognostic model was evaluated using multivariate and univariate analyses, and its accuracy and reliability were validated using a nomogram and ROC. Additionally, we investigated the relationships between IHRLs and the immune microenvironment. Result Our analysis identified GSEC, AC099850.3, and AL391001.1 as risk lncRNAs, while AC010615.2, AC010654.1, AL513550.1, LINC00996, and LINC01150 were categorized as protective lncRNAs. We observed variances in the expression of seven immune cells and 15 immune-correlated pathways between the two risk groups. Furthermore, our results confirmed the ceRNA network associated with the intermittent hypoxia-related lncRNA GSEC/miR-873-3p/EGLN3 regulatory pathway. GSEC showed pronounced expression in lung adenocarcinoma tissues and specific cell lines, and its inhibition resulted in reduced proliferation and migration in A549 and PC9 cells. Intriguingly, GSEC manifested oncogenic properties by sponging miR-873-3p and demonstrated a tendency to modulate EGLN3 expression favorably. Conclusion GSEC acts as an oncogenic lncRNA by interacting with miR-873-3p, modulating EGLN3 expression. This observation underscores the potential of GSEC as a diagnostic and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Peijun Liu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Long Zhou
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China
| | - Hao Chen
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yang He
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guangcai Li
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
8
|
Li Y, Li X, Yu Z. Novel methylation-related long non-coding RNA clinical outcome prediction method: the clinical phenotype and immune infiltration research in low-grade gliomas. Front Oncol 2023; 13:1177120. [PMID: 37228500 PMCID: PMC10203515 DOI: 10.3389/fonc.2023.1177120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/11/2023] [Indexed: 05/27/2023] Open
Abstract
Background Recent studies have suggested that long non-coding RNAs (lncRNAs) may play crucial role in low-grade glioma; however, the underlying mechanisms linking them to epigenetic methylation remain unclear. Methods We downloaded expression level data for regulators associated with N1 methyladenosine (m1A), 5-methyladenine (m5C), and N6 methyladenosine (m6A) (M1A/M5C/M6A) methylation from the Cancer Genome Atlas-low-grade glioma (TCGA-LGG) database. We identified the expression patterns of lncRNAs, and selected methylation-related lncRNAs using Pearson correlation coefficient>0.4. Non-negative matrix dimensionality reduction was then used to determine the expression patterns of the methylation-associated lncRNAs. We constructed a weighted gene co-expression network analysis (WGCNA) network to explore the co-expression networks between the two expression patterns. Functional enrichment of the co-expression network was performed to identify biological differences between the expression patterns of different lncRNAs. We also constructed prognostic networks based on the methylation presence in lncRNAs in low-grade gliomas. Results We identified 44 regulators by literature review. Using a correlation coefficient greater than 0.4, we identified 2330 lncRNAs, among which 108 lncRNAs with independent prognostic values were further screened using univariate Cox regression at P< 0.05. Functional enrichment of the co-expression networks revealed that regulation of trans-synaptic signaling, modulation of chemical synaptic transmission, calmodulin binding, and SNARE binding were mostly enriched in the blue module. The calcium and CA2 signaling pathways were associated with different methylation-related long non-coding chains. Using the Least Absolute Shrinkage Selector Operator (LASSO) regression analysis, we analyzed a prognostic model containing four lncRNAs. The model's risk score was 1.12 *AC012063 + 0.74 * AC022382 + 0.32 * AL049712 + 0.16 * GSEC. Gene set variation analysis (GSVA) revealed significant differences in mismatch repair, cell cycle, WNT signaling pathway, NOTCH signaling pathway, Complement and Cascades, and cancer pathways at different GSEC expression levels. Thus, these results suggest that GSEC may be involved in the proliferation and invasion of low-grade glioma, making it a prognostic risk factor for low-grade glioma. Conclusion Our analysis identified methylation-related lncRNAs in low-grade gliomas, providing a foundation for further research on lncRNA methylation. We found that GSEC could serve as a candidate methylation marker and a prognostic risk factor for overall survival in low-grade glioma patients. These findings shed light on the underlying mechanisms of low-grade glioma development and may facilitate the development of new treatment strategies.
Collapse
|
9
|
Qin H, Sheng W, Zhang G, Yang Q, Yao S, Yue Y, Zhang P, Zhu Y, Wang Q, Chen Y, Zeng H, Weng J, Yu F, Yang J. Comprehensive analysis of cuproptosis-related prognostic gene signature and tumor immune microenvironment in HCC. Front Genet 2023; 14:1094793. [PMID: 36891150 PMCID: PMC9986498 DOI: 10.3389/fgene.2023.1094793] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Background: Copper is an indispensable mineral element involved in many physiological metabolic processes. Cuproptosis is associated with a variety of cancer such as hepatocellular carcinoma (HCC). The objective of this study was to examine the relationships between the expression of cuproptosis-related genes (CRGs) and tumor characteristics, including prognosis and microenvironment of HCC. Methods: The differentially expressed genes (DEGs) between high and low CRGs expression groups in HCC samples were identified, and further were analyzed for functional enrichment analysis. Then, CRGs signature of HCC was constructed and analyzed utilizing LASSO and univariate and multivariate Cox regression analysis. Prognostic values of CRGs signature were evaluated by Kaplan-Meier analysis, independent prognostic analysis and nomograph. The expression of prognostic CRGs was verified by Real-time quantitative PCR (RT-qPCR) in HCC cell lines. In addition, the relationships between prognostic CRGs expression and the immune infiltration, tumor microenvironment, antitumor drugs response and m6A modifications were further explored using a series of algorithms in HCC. Finally, ceRNA regulatory network based on prognostic CRGs was constructed. Results: The DEGs between high and low CRG expression groups in HCC were mainly enriched in focal adhesion and extracellular matrix organization. Besides, we constructed a prognostic model that consists of CDKN2A, DLAT, DLST, GLS, and PDHA1 CRGs for predicting the survival likelihood of HCC patients. And the elevated expression of these five prognostic CRGs was substantially in HCC cell lines and associated with poor prognosis. Moreover, immune score and m6A gene expression were higher in the high CRG expression group of HCC patients. Furthermore, prognostic CRGs have higher mutation rates in HCC, and are significantly correlated with immune cell infiltration, tumor mutational burden, microsatellite instability, and anti-tumor drug sensitivity. Then, eight lncRNA-miRNA-mRNA regulatory axes that affected the progression of HCC were predicted. Conclusion: This study demonstrated that the CRGs signature could effectively evaluate prognosis, tumor immune microenvironment, immunotherapy response and predict lncRNA-miRNA-mRNA regulatory axes in HCC. These findings extend our knowledge of cuproptosis in HCC and may inform novel therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Haotian Qin
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weibei Sheng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | | | - Qi Yang
- Department of Medical Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Sen Yao
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yaohang Yue
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peng Zhang
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuanchao Zhu
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qichang Wang
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yixiao Chen
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jian Weng
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
10
|
Liu N, Yang C, Gao A, Sun M, Lv D. MiR-101: An Important Regulator of Gene Expression and Tumor Ecosystem. Cancers (Basel) 2022; 14:cancers14235861. [PMID: 36497343 PMCID: PMC9739992 DOI: 10.3390/cancers14235861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
MiRNAs are small single-stranded non-coding RNAs. MiRNA contributes to the transcriptional and post-transcriptional regulation of mRNA in different cell types, including mRNA transcription inhibition and mRNA decay and phenotypes via the effect of several essential oncogenic processes and tumor microenvironment. MiR-101 is a highly conserved miRNA that was found to alter the expression in various human cancers. MiR-101 has been reported to have tumor oncogenic and suppressive effects to regulate tumorigenesis and tumor progression. In this review, we summarize the new findings about the roles of miR-101 in cancers and the underlying mechanisms of targeting genes degradation and microenvironment regulation, which will improve biological understanding and design of novel therapeutics.
Collapse
Affiliation(s)
- Ning Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Chunsheng Yang
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Ang Gao
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Meili Sun
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
- Correspondence: (M.S.); (D.L.)
| | - Deguan Lv
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Correspondence: (M.S.); (D.L.)
| |
Collapse
|
11
|
Involvement of plasma lncRNA GSEC in sepsis discrimination and prognosis, and its correlation with macrophage cell inflammation and proliferation. Immunobiology 2022; 227:152264. [PMID: 36049364 DOI: 10.1016/j.imbio.2022.152264] [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: 06/27/2022] [Revised: 08/08/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Despite the dysregulation and function of G-quadruplex-forming sequence containing lncRNA (GSEC) have been widely reported in human cancers, there are few available data revealing its role in sepsis. OBJECTIVE To assess the expression and function of GSEC in the development of sepsis and its potential molecular mechanism. MATERIALS AND METHODS A total of 78 sepsis patients, 55 non-sepsis intensive care unit patients, and 42 healthy individuals were enrolled in this study. The expression of GSEC was evaluated in plasma and macrophage cells with polymerase chain reaction. The inflammation response of sepsis patients and macrophage cells was analyzed with an enzyme-linked immunosorbent assay. The diagnostic and prognostic value of GSEC in sepsis patients were estimated by receiver operator curve (ROC) and Cox analysis. The molecular mechanism underlying the function of GSEC was investigated in RAW264.7 cell with luciferase reporter assay and cell transfection. RESULTS Significant upregulation of GSEC was observed in sepsis patients' plasma, which could discriminate sepsis patients from healthy and non-sepsis individuals. Upregulation of GSEC was positively correlated with inflammation cytokine levels and adverse prognosis of sepsis patients. In vitro, GSEC was found to modulate the expression level of miR-873-3p, which mediated the regulatory effect of GSEC on the inflammation and proliferation of RAW264.7. CONCLUSION Upregulated GSEC could serve as a biomarker of sepsis pathogenesis and development. GSEC regulates the inflammation and proliferation of macrophage cells through modulating miR-873-3p.
Collapse
|
12
|
de la Cruz-Ojeda P, Schmid T, Boix L, Moreno M, Sapena V, Praena-Fernández JM, Castell FJ, Falcón-Pérez JM, Reig M, Brüne B, Gómez-Bravo MA, Giráldez Á, Bruix J, Ferrer MT, Muntané J. miR-200c-3p, miR-222-5p, and miR-512-3p Constitute a Biomarker Signature of Sorafenib Effectiveness in Advanced Hepatocellular Carcinoma. Cells 2022; 11:cells11172673. [PMID: 36078082 PMCID: PMC9454520 DOI: 10.3390/cells11172673] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Sorafenib constitutes a suitable treatment alternative for patients with advanced hepatocellular carcinoma (HCC) in whom atezolizumab + bevacizumab therapy is contraindicated. The aim of the study was the identification of a miRNA signature in liquid biopsy related to sorafenib response. Methods: miRNAs were profiled in hepatoblastoma HepG2 cells and tested in animal models, extracellular vesicles (EVs), and plasma from HCC patients. Results: Sorafenib altered the expression of 11 miRNAs in HepG2 cells. miR-200c-3p and miR-27a-3p exerted an anti-tumoral activity by decreasing cell migration and invasion, whereas miR-122-5p, miR-148b-3p, miR-194-5p, miR-222-5p, and miR-512-3p exerted pro-tumoral properties by increasing cell proliferation, migration, or invasion, or decreasing apoptosis. Sorafenib induced a change in EVs population with an increased number of larger EVs, and promoted an accumulation of miR-27a-3p, miR-122-5p, miR-148b-3p, miR-193b-3p, miR-194-5p, miR-200c-3p, and miR-375 into exosomes. In HCC patients, circulating miR-200c-3p baseline levels were associated with increased survival, whereas high levels of miR-222-5p and miR-512-3p after 1 month of sorafenib treatment were related to poor prognosis. The RNA sequencing revealed that miR-200c-3p was related to the regulation of cell growth and death, whereas miR-222-5p and miR-512-3p were related to metabolic control. Conclusions: The study showed that Sorafenib regulates a specific miRNA signature in which miR-200c-3p, miR-222-5p, and miR-512-3p bear prognostic value and contribute to treatment response.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60528 Frankfurt, Germany
| | - Loreto Boix
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - Manuela Moreno
- Department of General Surgery, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Víctor Sapena
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | | | - Francisco J. Castell
- Department of Radiology, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Juan Manuel Falcón-Pérez
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Exosomes Lab, CIC bioGUNE, 48160 Derio, Spain
| | - María Reig
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60528 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60528 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60528 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60528 Frankfurt, Germany
| | - Miguel A. Gómez-Bravo
- Department of General Surgery, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Álvaro Giráldez
- Unit for the Clinical Management of Digestive Diseases, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Jordi Bruix
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, 08036 Barcelona, Spain
| | - María T. Ferrer
- Unit for the Clinical Management of Digestive Diseases, Hospital University “Virgen del Rocío”/CSIC/University of Seville/IBIS, 41013 Seville, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain
- Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain
- Correspondence: ; Tel.: +34-955-923-122; Fax: +34-955-923-002
| |
Collapse
|