51
|
Jeon YJ, Hasan MM, Park HW, Lee KW, Manavalan B. TACOS: a novel approach for accurate prediction of cell-specific long noncoding RNAs subcellular localization. Brief Bioinform 2022; 23:6618237. [PMID: 35753698 PMCID: PMC9294414 DOI: 10.1093/bib/bbac243] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are primarily regulated by their cellular localization, which is responsible for their molecular functions, including cell cycle regulation and genome rearrangements. Accurately identifying the subcellular location of lncRNAs from sequence information is crucial for a better understanding of their biological functions and mechanisms. In contrast to traditional experimental methods, bioinformatics or computational methods can be applied for the annotation of lncRNA subcellular locations in humans more effectively. In the past, several machine learning-based methods have been developed to identify lncRNA subcellular localization, but relevant work for identifying cell-specific localization of human lncRNA remains limited. In this study, we present the first application of the tree-based stacking approach, TACOS, which allows users to identify the subcellular localization of human lncRNA in 10 different cell types. Specifically, we conducted comprehensive evaluations of six tree-based classifiers with 10 different feature descriptors, using a newly constructed balanced training dataset for each cell type. Subsequently, the strengths of the AdaBoost baseline models were integrated via a stacking approach, with an appropriate tree-based classifier for the final prediction. TACOS displayed consistent performance in both the cross-validation and independent assessments compared with the other two approaches employed in this study. The user-friendly online TACOS web server can be accessed at https://balalab-skku.org/TACOS.
Collapse
Affiliation(s)
- Young-Jun Jeon
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Md Mehedi Hasan
- Tulane Center for Biomedical Informatics and Genomics, Division of Biomedical Informatics and Genomics, John W. Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hyun Woo Park
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics laboratory, Department of Integrative Biotechnology, College of Bioengineering and Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
52
|
Identification of LOC101927355 as a Novel Biomarker for Preeclampsia. Biomedicines 2022; 10:biomedicines10061253. [PMID: 35740273 PMCID: PMC9219905 DOI: 10.3390/biomedicines10061253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Preeclampsia, a disorder with a heterogeneous physiopathology, can be attributed to maternal, fetal, and/or placental factors. Long non-coding RNAs (lncRNAs) refer to a class of non-coding RNAs, the essential regulators of biological processes; their differential expression has been associated with the pathogenesis of multiple diseases. The study aimed to identify lncRNAs, expressed in the placentas and plasma of patients who presented with preeclampsia, as potential putative biomarkers of the disease. In silico analysis was performed to determine lncRNAs differentially expressed in the placentas of patients with preeclampsia, using a previously published RNA-Seq dataset. Seven placentas and maternal plasma samples collected at delivery from preterm preeclamptic patients (≤37 gestational weeks of gestation), and controls were used to validate the expression of lncRNAs by qRT-PCR. Six lncRNAs were validated and differentially expressed (p < 0.05) in the preeclampsia and control placentas: UCA1 and HCG4 were found upregulated, and LOC101927355, LINC00551, PART1, and NRAD1 downregulated. Two of these lncRNAs, HCG4 and LOC101927355, were also detected in maternal plasma, the latter showing a significant decrease (p = 0.03) in preeclamptic patients compared to the control group. In silico analyses showed the cytoplasmic location of LOC101927355, which suggests a role in post-transcriptional gene regulation. The detection of LOC101927355 in the placenta and plasma opens new possibilities for understanding the pathogenesis of preeclampsia and for its potential use as a biomarker.
Collapse
|
53
|
Downregulation of lncRNA Miat contributes to the protective effect of electroacupuncture against myocardial fibrosis. Chin Med 2022; 17:57. [PMID: 35578250 PMCID: PMC9112552 DOI: 10.1186/s13020-022-00615-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Background Myocardial fibrosis changes the structure of myocardium, leads to cardiac dysfunction and induces arrhythmia and cardiac ischemia, threatening patients’ lives. Electroacupuncture at PC6 (Neiguan) was previously found to inhibit myocardial fibrosis. Long non-coding RNAs (lncRNAs) play a variety of regulatory functions in myocardial fibrosis, but whether electroacupuncture can inhibit myocardial fibrosis by regulating lncRNA has rarely been reported. Methods In this study, we constructed myocardial fibrosis rat models using isoproterenol (ISO) and treated rats with electroacupuncture at PC6 point and non-point as control. Hematoxylin–eosin, Masson and Sirius Red staining were performed to assess the pathological changes and collagen deposition. The expression of fibrosis-related markers in rat myocardial tissue were detected by RT-qPCR and Western blot. Miat, an important long non-coding RNA, was selected to study the regulation of myocardial fibrosis by electroacupuncture at the transcriptional and post-transcriptional levels. In post-transcriptional level, we explored the myocardial fibrosis regulation effect of Miat on the sponge effect of miR-133a-3p. At the transcriptional level, we studied the formation of heterodimer PPARG–RXRA complex and promotion of the TGF-β1 transcription. Results Miat was overexpressed by ISO injection in rats. We found that Miat can play a dual regulatory role in myocardial fibrosis. Miat can sponge miR-133a-3p in an Ago2-dependent manner, reduce the binding of miR-133a-3p target to the 3ʹUTR region of CTGF mRNA and improve the protein expression level of CTGF. In addition, it can also directly bind with PPARG protein, inhibit the formation of heterodimer PPARG–RXRA complex and then promote the transcription of TGF-β1. Electroacupuncture at PC6 point, but not at non-points, can reduce the expression of Miat, thus inhibiting the expression of CTGF and TGF-β1 and inhibiting myocardial fibrosis. Conclusion We revealed that electroacupuncture at PC6 point can inhibit the process of myocardial fibrosis by reducing the expression of lncRNA Miat, which is a potential therapeutic method for myocardial fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00615-6.
Collapse
|
54
|
Cao J, Xiao C, Fong CJTH, Gong J, Li D, Li X, Jie Y, Chong Y. Expression and Regulatory Network Analysis of Function of Small Nucleolar RNA Host Gene 4 in Hepatocellular Carcinoma. J Clin Transl Hepatol 2022; 10:297-307. [PMID: 35528985 PMCID: PMC9039712 DOI: 10.14218/jcth.2020.00175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/30/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND AIMS Long non-coding RNA small nucleolar RNA host genes (SNHGs) play a critical role in the occurrence and development of tumors. In this study, we aimed to investigate the role of SNHG4 in hepatocellular carcinoma (HCC) and its underlining mechanism. METHODS Datasets were acquired from The Cancer Genome Atlas (TCGA) database. lncLocator 2.0 was used to identify the distribution of SNHG4 in HCC cells. Gene expression, Kaplan-Meier survival, microRNA and transcription factor target analyses were performed with the University of Alabama Cancer (UALCAN) Database, Kaplan-Meier Plotter, LinkedOmics, WebGestalt and gene set enrichment analysis, respectively. Gene Ontology and pathway enrichment analyses and assessment of RNA binding proteins were performed by R software, circlncRNAnet and Encyclopedia of RNA Interactomes (ENCORI). In addition, CirclncRNAnet and ENCORI were used to find the correlation between SNHG4 and important proteins, while the prognostic value was assessed with the Human Protein Atlas database and Kaplan-Meier Plotter. RESULTS Expression of SNHG4 in HCC is higher in HCC tissue than in normal healthy liver tissues and is mainly distributed in the nucleus. SNHG4 positively correlated with poor prognosis (p<0.01 for overall survival and recurrence-free survival). Functional enrichment analysis revealed SNHG4 involvement with regulation of ribosomal RNA synthesis and the RNA processing and surveillance pathway. SNHG4 is closely associated with miR-154 and miR-206, transcription factor target E2F family and the signaling pathway for MAPK/ERK and mTOR. U2 auxiliary factor 2 (U2AF2) showed strong correlation with SNHG4, while low-expression of U2AF2 showed good prognosis. CONCLUSIONS Based on our findings, we infer SNHG4 may play a role in the formation of HCC via regulation of tumor-related pathways.
Collapse
Affiliation(s)
- Jing Cao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cuicui Xiao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Christ-Jonathan Tsia Hin Fong
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiao Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Danyang Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangyong Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yusheng Jie
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Correspondence to: Yusheng Jie and Yutian Chong, Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China. ORCID: https://orcid.org/0000-0003-3756-0653 (YJ), https://orcid.org/0000-0001-8215-4393 (YC). Tel: +86-20-8525-2372, Fax: +86-20-8525-2250, E-mail: (YJ), (YC)
| | - Yutian Chong
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Correspondence to: Yusheng Jie and Yutian Chong, Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China. ORCID: https://orcid.org/0000-0003-3756-0653 (YJ), https://orcid.org/0000-0001-8215-4393 (YC). Tel: +86-20-8525-2372, Fax: +86-20-8525-2250, E-mail: (YJ), (YC)
| |
Collapse
|
55
|
Li J, Momen-Heravi F, Wu X, He K. Mechanism of METTL14 and m6A modification of lncRNA MALAT1 in the proliferation of oral squamous cell carcinoma cells. Oral Dis 2022. [PMID: 35467063 DOI: 10.1111/odi.14220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Methyltransferase-like 14 (METTL14) plays an epigenetic role in various cancer through N6-methyladenosine (m6A) modification. This study sought to analyze the mechanism of METTL14 in oral squamous cell carcinoma (OSCC) cell proliferation. METHODS Expression levels of METTL14, lncRNA metastasis associated with lung adenocarcinoma transcript 1 (lncRNA MALAT1), microRNA (miR)-224-5p, and histone lysine demethylase 2A (KDM2A) in OSCC tissues (N = 40), and cell lines (FaDu, SCC-25, CAL-27, and SCC-15) were detected. Cell viability and colony formation capacity were assessed. m6A level, stability, and subcellular localization of lncRNA MALAT1 were determined. Nude mouse xenograft tumor assay was performed to confirm the role of METTL14 in vivo. RESULTS METTL14 and lncRNA MALAT1 were upregulated, and miR-224-5p was downregulated in OSCC tissues and cells. Silencing METTL14 repressed OSCC cell viability and colony formation. Overexpression of MALAT1 and KDM2A or miR-224-5p downregulation reversed the inhibition of silencing METTL14 on OSCC cell proliferation. METTL14 induced m6A modification of MALAT1 to upregulate MALAT1. MALAT1 is comparatively bound to miR-224-5p to promote KDM2A transcription. In vivo, METTL14 promoted tumor growth via regulating MALAT1/miR-224-5p/ KDM2A. CONCLUSIONS Overall, our findings verified the therapeutic role of silencing METTL14 in OSCC treatment through the MALAT1/miR-224-5p/KDM2A axis.
Collapse
Affiliation(s)
- Jinli Li
- Department of Gastroenterology, 923 Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Nanning City, Guangxi Province, China
| | - Fatemeh Momen-Heravi
- Cancer Biology and Immunology Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, Columbia University College of Dental Medicine, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Xun Wu
- Department of Maxillofacial Surgery, Southern Medical University Shenzhen Stomatology Hospital (Pingshan), Shenzhen City, Guangdong Province, China
| | - Kaili He
- Department of Stomatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China
| |
Collapse
|
56
|
Yin S, Li X, Xiong Z, Xie M, Jin L, Chen H, Mao C, Zhang F, Lian L. A novel ceRNA-immunoregulatory axis based on immune cell infiltration in ulcerative colitis-associated colorectal carcinoma by integrated weighted gene co-expression network analysis. BMC Gastroenterol 2022; 22:188. [PMID: 35428188 PMCID: PMC9013140 DOI: 10.1186/s12876-022-02252-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 03/27/2022] [Indexed: 11/22/2022] Open
Abstract
Background Patients with ulcerative colitis are at an increased risk of developing colorectal cancer with a prolonged disease course. Many studies have shown that alterations in the immune microenvironment play a key role in ulcerative colitis-associated colorectal cancer. Additionally, competing endogenous RNAs have important functions in immunoregulation, affecting inflammation and tumorigenesis. However, the complexity and behavioral characteristics of the competing endogenous RNA immunoregulatory network in ulcerative colitis-associated colorectal cancer remain unclear. We constructed a competing endogenous RNA immunoregulatory network to discover and validate a novel competing endogenous RNA immunoregulatory axis to provide insight into ulcerative colitis-associated colorectal cancer progression. Methods The competing endogenous RNA immunoregulatory network was constructed using differential expression analysis, weighted gene co-expression network analysis, and immune-related genes. Cmap was used to identify small-molecule drugs with therapeutic potential in ulcerative colitis-associated colorectal cancer. The ulcerative colitis-associated colorectal cancer-related pathways were identified by gene set variation and enrichment analysis. CIBERSORT, single-sample Gene Set Enrichment Analysis, and xCell were used to evaluate the infiltration of immune cells and screen hub immunocytes. The competing endogenous RNA immunoregulatory axis was identified by correlation analysis. Results We identified 130 hub immune genes and constructed a competing endogenous RNA immunoregulatory network consisting of 56 long non-coding RNAs, four microRNAs, and six targeted hub immune genes. Four small-molecule drugs exerted potential therapeutic effects by reversing the expression of hub immune genes. Pathway analysis showed that the NF-κB pathway was significantly enriched. Neutrophils were identified as hub immunocytes, and IL6ST was significantly positively correlated with the neutrophil count. In addition, NEAT1 may serve as a competing endogenous RNA to sponge miR-1-3p and promote IL6ST expression. Conclusions The competing endogenous RNA immunoregulatory axis may regulate neutrophil infiltration, affecting the occurrence of ulcerative colitis-associated colorectal cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02252-7.
Collapse
|
57
|
Peng S, Zhang T, Zhang S, Tang Q, Yan Y, Feng H. Integrated Bioinformatics and Validation Reveal IL1B and Its Related Molecules as Potential Biomarkers in Chronic Spontaneous Urticaria. Front Immunol 2022; 13:850993. [PMID: 35371000 PMCID: PMC8975268 DOI: 10.3389/fimmu.2022.850993] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Background The etiopathogenesis of chronic spontaneous urticaria (CSU) has not been fully understood, and there has been extensive interest in the interaction between inflammatory dermatosis and pyroptosis. This study intends to investigate the molecular mechanism of pyroptosis-related genes in CSU via bioinformatic ways, aiming at identifying the potential key biomarker. Methods GSE72540, the RNA expression profile dataset of CSU, was utilized as the training set, and GSE57178 as the validation set. Differently expressed pyroptosis-related genes (DEPRGs), GO, KEGG, and DO analyses were performed. The hub genes were explored by the protein–protein interaction analysis. Moreover, CIBERSORT was employed for estimating immune cell types and proportions. Then, we constructed a DEmRNA–miRNA–DElncRNA ceRNA network and a drug–gene interaction network. Finally, ELISA was used for gene expression analysis. Results We recognized 17 DEPRGs, whose enrichment analyses showed that they were mostly enriched in inflammatory response and immunomodulation. Moreover, 5 hub genes (IL1B, TNF, and IRF1 are upregulated, HMGB1 and P2RX7 are downregulated) were identified via the PPI network and verified by a validation set. Then immune infiltration analysis displayed that compared with normal tissue, CSU owned a significantly higher proportion of mast cells activated, but a lower proportion of T cells CD4 naive and so on. Furthermore, IL1B was statistically and positively associated with mast cells activated in CSU, and SNHG3, the upstream factor of IL1B in the ceRNA we constructed, also related with mast cells in CSU. Further analysis exhibited that the protein subcellular localization of IL1B was extracellular, according with its intercellular regulation role; IL1B was significantly correlated with key immune checkpoints; and the NOD-like receptor signaling pathway was the mainly involved pathway of IL1B based on the couple databases. What is more, the result of ELISA of CSU patients was the same as the above analyses about IL1B. In addition, the drug–gene interaction network contained 15 potential therapeutic drugs targeting IL1B, and molecular docking might make this relationship viable. Conclusion IL1B and its related molecules might play a key role in the development of CSU and could be potential biomarkers in CSU.
Collapse
Affiliation(s)
- Shixiong Peng
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial People’s Hospital, Changsha, China
| | - Teng Zhang
- Department of Dermatology, Chinese Traditional Hospital of Changsha, Changsha, China
| | - Sisi Zhang
- Nursing Department, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Qian Tang
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial People’s Hospital, Changsha, China
| | - Yang Yan
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial People’s Hospital, Changsha, China
| | - Hao Feng
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial People’s Hospital, Changsha, China
- *Correspondence: Hao Feng,
| |
Collapse
|
58
|
Qian HG, Wu Q, Wu JH, Tian XY, Xu W, Hao CY. Long non‑coding RNA LINC00238 suppresses the malignant phenotype of liver cancer by sponging miR‑522. Mol Med Rep 2022; 25:71. [PMID: 35014686 PMCID: PMC8767542 DOI: 10.3892/mmr.2022.12587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 11/05/2022] Open
Abstract
Long non‑coding RNAs can regulate the malignant tumor phenotype either as tumor suppressors or oncogenes. The present study investigated the underlying mechanism of LINC00238 in liver cancer. LINC00238 was identified as a downregulated molecule in The Cancer Genome Atlas liver hepatocellular carcinoma dataset through Gene Expression Profiling Interactive Analysis software. Through gain‑ and loss‑of‑function experiments, LINC00238 was confirmed as a tumor suppressor that could not only decrease cell viability, migration and invasion in vitro, but also tumorigenesis and tumor metastasis in vivo. By cytoplasmic and nuclear RNA isolation, LINC00238 was confirmed to be predominantly cytoplasmic. Mechanistically, RNA pull‑down assays showed that LINC00238 sponged microRNA (miR)‑522 and then reversed the inhibitory effects on two downstream targets, secreted frizzled related protein 2 and dickkopf1. Collectively, LINC00238 was identified as a tumor suppressor that acts via sponging miR‑522 followed by silencing of downstream targets, suggesting that LINC00238 may have a key role in suppressing the malignant phenotype of liver cancer cells.
Collapse
Affiliation(s)
- Hong-Gang Qian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Qiong Wu
- MOE Key Lab, Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Jian-Hui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xiu-Yun Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Wei Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Chun-Yi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
59
|
Yang L, Dong X, Liu Z, Tan J, Huang X, Wen T, Qu H, Wang Z. VPS9D1-AS1 overexpression amplifies intratumoral TGF-β signaling and promotes tumor cell escape from CD8 + T cell killing in colorectal cancer. eLife 2022; 11:79811. [PMID: 36458816 PMCID: PMC9744440 DOI: 10.7554/elife.79811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 12/01/2022] [Indexed: 12/05/2022] Open
Abstract
Efficacy of immunotherapy is limited in patients with colorectal cancer (CRC) because high expression of tumor-derived transforming growth factor (TGF)-β pathway molecules and interferon (IFN)-stimulated genes (ISGs) promotes tumor immune evasion. Here, we identified a long noncoding RNA (lncRNA), VPS9D1-AS1, which was located in ribosomes and amplified TGF-β signaling and ISG expression. We show that high expression of VPS9D1-AS1 was negatively associated with T lymphocyte infiltration in two independent cohorts of CRC. VPS9D1-AS1 served as a scaffolding lncRNA by binding with ribosome protein S3 (RPS3) to increase the translation of TGF-β, TGFBR1, and SMAD1/5/9. VPS9D1-AS1 knockout downregulated OAS1, an ISG gene, which further reduced IFNAR1 levels in tumor cells. Conversely, tumor cells overexpressing VPS9D1-AS1 were resistant to CD8+ T cell killing and lowered IFNAR1 expression in CD8+ T cells. In a conditional overexpression mouse model, VPS9D1-AS1 enhanced tumorigenesis and suppressed the infiltration of CD8+ T cells. Treating tumor-bearing mice with antisense oligonucleotide drugs targeting VPS9D1-AS1 significantly suppressed tumor growth. Our findings indicate that the tumor-derived VPS9D1-AS1/TGF-β/ISG signaling cascade promotes tumor growth and enhances immune evasion and may thus serve as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Lei Yang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina,Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Xichen Dong
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Zheng Liu
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Jinjing Tan
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Xiaoxi Huang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Tao Wen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Hao Qu
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| | - Zhenjun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
60
|
Li X, Xie B, Lu Y, Yang H, Wang J, Yu F, Zhang L, Cong B, Wen D, Ma C. Transcriptomic Analysis of Long Non-coding RNA-MicroRNA-mRNA Interactions in the Nucleus Accumbens Related to Morphine Addiction in Mice. Front Psychiatry 2022; 13:915398. [PMID: 35722589 PMCID: PMC9201067 DOI: 10.3389/fpsyt.2022.915398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Recent research suggest that some non-coding RNAs (ncRNAs) are important regulators of chromatin dynamics and gene expression in nervous system development and neurological diseases. Nevertheless, the molecular mechanisms of long non-coding RNAs (lncRNAs), acting as competing endogenous RNAs (ceRNAs), underlying morphine addiction are still unknown. In this research, RNA sequencing (RNA-seq) was used to examine the expression profiles of lncRNAs, miRNAs and mRNAs on the nucleus accumbens (NAc) tissues of mice trained with morphine or saline conditioned place preference (CPP), with differential expression of 31 lncRNAs, 393 miRNAs, and 371 mRNAs found. A ceRNA network was established for reciprocal interactions for 9 differentially expressed lncRNAs (DElncRNAs), 10 differentially expressed miRNAs (DEmiRNAs) and 12 differentially expressed mRNAs (DEmRNAs) based on predicted miRNAs shared by lncRNAs and mRNAs. KEGG pathway enrichment analyses were conducted to explore the potential functions of DEmRNAs interacting with lncRNAs in the ceRNA network. These DEmRNAs were enriched in synaptic plasticity-related pathways, including pyrimidine metabolism, ECM-receptor interaction, and focal adhesion. The correlation between the relative expression of lncRNAs, miRNAs and mRNAs was analyzed to further validate predicted ceRNA networks, and the Lnc15qD3-miR-139-3p-Lrp2 ceRNA regulatory interaction was determined. These results suggest that the comprehensive network represents a new insight into the lncRNA-mediated ceRNA regulatory mechanisms underlying morphine addiction and provide new potential diagnostic and prognostic biomarkers for morphine addiction.
Collapse
Affiliation(s)
- Xiaojie Li
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Bing Xie
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Yun Lu
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Hongyu Yang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Jian Wang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Feng Yu
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Ludi Zhang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Chunling Ma
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
61
|
He Y, Jiang X, Duan L, Xiong Q, Yuan Y, Liu P, Jiang L, Shen Q, Zhao S, Yang C, Chen Y. LncRNA PKMYT1AR promotes cancer stem cell maintenance in non-small cell lung cancer via activating Wnt signaling pathway. Mol Cancer 2021; 20:156. [PMID: 34856993 PMCID: PMC8638142 DOI: 10.1186/s12943-021-01469-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common type of human lung cancers, which has diverse pathological features. Although many signaling pathways and therapeutic targets have been defined to play important roles in NSCLC, limiting efficacies have been achieved. Methods Bioinformatics methods were used to identify differential long non-coding RNA expression in NSCLC. Real-time RT-PCR experiments were used to examine the expression pattern of lncRNA PKMYT1AR, miR-485-5p. Both in vitro and in vivo functional assays were performed to investigate the functional role of PKMYT1AR/miR-485-5p/PKMYT1 axis on regulating cell proliferation, migration and tumor growth. Dual luciferase reporter assay, fluorescent in situ hybridization (FISH), immunoblot, co-immunoprecipitation experiments were used to verify the molecular mechanism. Result Here, we identify a human-specific long non-coding RNA (lncRNA, ENST00000595422), termed PKMYT1AR (PKMYT1 associated lncRNA), that is induced in NSCLC by Yin Yang 1 (YY1) factor, especially in cancerous cell lines (H358, H1975, H1299, H1650, A549 and SPC-A1) compared to that in normal human bronchial epithelium cell line (BEAS-2B). We show that PKMYT1AR high expression correlates with worse clinical outcome, and knockdown of PKMYT1AR inhibits tumor cell proliferation, migration and xenograft tumor formation abilities. Bioinformatic analysis and a luciferase assay demonstrate that PKMYT1AR directly interacts with miR-485-5p to attenuate the inhibitory role on its downstream oncogenic factor PKMYT1 (the protein kinase, membrane-associated tyrosine/threonine 1) in NSCLC. Furthermore, we uncover that miR-485-5p is downregulated in both cancerous cell lines and peripheral blood serum isolated from NSCLC patients compared to reciprocal control groups. Consistently, forced expression of miR-485-5p inhibits the proliferation and migration abilities of tumor cells. Moreover, we provide evidence showing that PKMYT1AR targeting antisense oligonucleotide (ASO) dramatically inhibit tumor growth in vivo. Mechanistic study shows that PKMYT1AR/ miR-485-5p /PKMYT1 axis promotes cancer stem cells (CSCs) maintenance in NSCLC via inhibiting β-TrCP1 mediated ubiquitin degradation of β-catenin proteins, which in turn causes enhanced tumorigenesis. Conclusions Our findings reveal the critical role of PKMYT1AR/miR-485-5p /PKMYT1 axis during NSCLC progression, which could be used as novel therapeutic targets in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01469-6.
Collapse
Affiliation(s)
- Yaomei He
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiulin Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lincan Duan
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, Yunnan, China
| | - Qiuxia Xiong
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yixiao Yuan
- Department of Thoracic Surgery, the Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, Yunnan, China
| | - Peishen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liping Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Qiushuo Shen
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Cuiping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.
| |
Collapse
|
62
|
Decoding LncRNAs. Cancers (Basel) 2021; 13:cancers13112643. [PMID: 34072257 PMCID: PMC8199187 DOI: 10.3390/cancers13112643] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
Non-coding RNAs (ncRNAs) have been considered as unimportant additions to the transcriptome. Yet, in light of numerous studies, it has become clear that ncRNAs play important roles in development, health and disease. Long-ignored, long non-coding RNAs (lncRNAs), ncRNAs made of more than 200 nucleotides have gained attention due to their involvement as drivers or suppressors of a myriad of tumours. The detailed understanding of some of their functions, structures and interactomes has been the result of interdisciplinary efforts, as in many cases, new methods need to be created or adapted to characterise these molecules. Unlike most reviews on lncRNAs, we summarize the achievements on lncRNA studies by taking into consideration the approaches for identification of lncRNA functions, interactomes, and structural arrangements. We also provide information about the recent data on the involvement of lncRNAs in diseases and present applications of these molecules, especially in medicine.
Collapse
|
63
|
Gu S, Zhang G, Si Q, Dai J, Song Z, Wang Y. Web tools to perform long non-coding RNAs analysis in oncology research. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2021; 2021:6326500. [PMID: 34296748 PMCID: PMC8299716 DOI: 10.1093/database/baab047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/21/2021] [Accepted: 07/11/2021] [Indexed: 11/14/2022]
Abstract
Accumulated evidence suggests that the widely expressed long-non-coding RNAs (lncRNAs) are involved in biogenesis. Some aberrant lncRNAs are closely related to pathological changes, for instance, in cancer. Both in tumorigenesis and cancer progression, depending on the interplay with cellular molecules, lncRNAs can modulate transcriptional interference, chromatin remodeling, post-translational regulation and protein modification, and further interfere with signaling pathways. Aiming to the diagnosis/ prognosis markers or potential therapeutical targets, it is important to figure out the specific mechanism and the tissue-specific expressing patterns of lncRNAs. Generally, the bioinformatics analysis is the first step. More and more in silico databases are increasing. But the existing integrative online platforms’ functions are not only having their unique features but also share some common features, which may lead to a waste of time for researchers. Here, we reviewed these web tools according to the functions. For each database, we clarified the data source, analysis method and the evidence that the analysis result is derived from. This review also illustrated examples in practical use for a specific lncRNA by these web tools. It will provide convenience for researchers to quickly choose the appropriate bioinformatics web tools in oncology studies.
Collapse
Affiliation(s)
- Shixing Gu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China
| | - Guangjie Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China.,Department of Clinical Laboratory, Chengdu Fifth People's Hospital, No.33 Mashi Street, Chengdu, Sichuan 611130, China
| | - Qin Si
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China
| | - Jiawen Dai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China
| | - Zhen Song
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China
| | - Yingshuang Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Road, Chengdu, Sichuan 611137, China
| |
Collapse
|