1
|
Aghaei‐Zarch SM, Esmaeili A, Bagheri‐Mohammadi S. A Comprehensive Review on LncRNAs/miRNAs-DNMT1 Axis in Human Cancer: Mechanistic and Clinical Application. J Cell Mol Med 2025; 29:e70604. [PMID: 40387409 PMCID: PMC12087000 DOI: 10.1111/jcmm.70604] [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: 08/19/2024] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Cancer constitutes a significant public health concern, and addressing the challenge of cancer holds paramount importance and requires immediate attention. Epigenetic alterations, encompassing DNA methylation, have emerged as pivotal contributors to the development of diverse cancer types. These modifications exert their influence by modulating chromatin structure, gene expression patterns and other nuclear processes, thereby influencing cancer pathogenesis. Over the last two decades, an increasing body of evidence has established the involvement of DNA methyltransferase 1 (DNMT1) in various aspects of cancer development, including tumorigenesis, aggressiveness and treatment response. Furthermore, non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are increasingly recognised as significant modulators in diverse biological processes, encompassing metastasis, apoptosis, cell proliferation and differentiation. Several recent studies have elucidated the intricate relationship between epigenetic machinery, specifically DNMT1, and the expression of ncRNAs in the context of cancer. In this review, we provide a comprehensive overview of the interaction between DNMT1 and ncRNAs in cancer pathogenesis. Furthermore, we discuss the important role of the ncRNAs-DNMT1 axis in cancer stem cells and cancer therapy resistance as critical issues in cancer therapy. Finally, we demonstrate that herbal medicine and synthetic RNA molecules regulate DNMT1 activity and hold great promise in cancer treatment.
Collapse
Affiliation(s)
- Seyed Mohsen Aghaei‐Zarch
- Department of Medical Genetics, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Ali Esmaeili
- Student Research Committee, Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Saeid Bagheri‐Mohammadi
- Department of Paramedicine, Amol School of Paramedical SciencesMazandaran University of Medical SciencesSariIran
- Immunogenetic Research CenterMazandaran University of Medical SciencesSariIran
| |
Collapse
|
2
|
Zhang RX, Zhang ZX, Zhao XY, Liu YH, Zhang XM, Han Q, Wang XY. Mechanism of action of lncRNA-NEAT1 in immune diseases. Front Genet 2025; 16:1501115. [PMID: 40110044 PMCID: PMC11919857 DOI: 10.3389/fgene.2025.1501115] [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: 09/28/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
NEAT1, a long non-coding RNA (lncRNA), is involved in assembling nuclear paraspeckles that have been found to impact various immune-related diseases, such as autoimmune diseases, allergic diseases, cancer immunity, sepsis, etc. In immune-related diseases, lncRNA-NEAT1 affects the activation, proliferation, and differentiation process of immune cells by interacting with transcription factors and miRNA (MicroRNA) to regulate an expression level in immune-related genes. It can also regulate the apoptosis and autophagy processes of immune cells by regulating inflammatory responses, interacting with apoptosis-related proteins, or regulating the expression of autophagy-related genes, thereby regulating the development of immune-related diseases. In recent years, a large number of researchers have found that the abnormal expression of lncRNA-NEAT1 has a great impact on the onset and progression of immune diseases, such as innate immunity after viral infection and the humoral immunity of T lymphocytes. In this paper, the specific mechanism of action and the function of lncRNA-NEAT1 in different immune-related diseases are sorted out and analyzed, to furnish a theoretical foundation for the study of the mechanism of action of immune cells.
Collapse
Affiliation(s)
- Ruo-Xuan Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zi-Xuan Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiang-Yu Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yi-Han Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiao-Meng Zhang
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qin Han
- School of Management, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiao-Yu Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
3
|
Siqueira E, Velasco C, Tarrasón A, Soler M, Srinivas T, Setién F, Oliveira-Mateos C, Casado-Pelaez M, Martinez-Verbo L, Armstrong J, Esteller M, Alves L, Llobet A, Guil S. NEAT1-mediated regulation of proteostasis and mRNA localization impacts autophagy dysregulation in Rett syndrome. Nucleic Acids Res 2025; 53:gkaf074. [PMID: 39921568 PMCID: PMC11806351 DOI: 10.1093/nar/gkaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder primarily caused by loss-of-function mutations in the MECP2 gene, resulting in diverse cellular dysfunctions. Here, we investigated the role of the long noncoding RNA (lncRNA) NEAT1 in the context of MeCP2 deficiency using human neural cells and RTT patient samples. Through single-cell RNA sequencing and molecular analyses, we found that NEAT1 is markedly downregulated in MECP2 knockout (KO) cells at various stages of neural differentiation. NEAT1 downregulation correlated with aberrant activation of the mTOR pathway, abnormal protein metabolism, and dysregulated autophagy, contributing to the accumulation of protein aggregates and impaired mitochondrial function. Reactivation of NEAT1 in MECP2-KO cells rescued these phenotypes, indicating its critical role downstream of MECP2. Furthermore, direct RNA-RNA interaction was revealed as the key process for NEAT1 influence on autophagy genes, leading to altered subcellular localization of specific autophagy-related messenger RNAs and impaired biogenesis of autophagic complexes. Importantly, NEAT1 restoration rescued the morphological defects observed in MECP2-KO neurons, highlighting its crucial role in neuronal maturation. Overall, our findings elucidate lncRNA NEAT1 as a key mediator of MeCP2 function, regulating essential pathways involved in protein metabolism, autophagy, and neuronal morphology.
Collapse
Affiliation(s)
- Edilene Siqueira
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), 70.070-010 Brasilia, Brazil
| | - Cecilia D Velasco
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907L’Hospitalet de Llobregat, Catalonia, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Catalonia, Spain
| | - Ariadna Tarrasón
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Marta Soler
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Tara Srinivas
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Fernando Setién
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Cristina Oliveira-Mateos
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Marta Casado-Pelaez
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Laura Martinez-Verbo
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Judith Armstrong
- Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, 08950 Barcelona, Catalonia, Spain
- Servei de Medicina Genètica i Molecular, Hospital Sant Joan de Déu, 08950 Barcelona, Catalonia, Spain
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08907 Barcelona, Catalonia, Spain
| | - Letícia F Alves
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
| | - Artur Llobet
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907L’Hospitalet de Llobregat, Catalonia, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Catalonia, Spain
| | - Sonia Guil
- Josep Carreras Leukaemia Research Institute (IJC), Genesis of cancer Program, 08916 Badalona,Catalonia, Spain
- Germans Trias i Pujol Health Science Research Institute, 08916 Badalona, Catalonia, Spain
| |
Collapse
|
4
|
Mitrovic K, Zivotic I, Kolic I, Zakula J, Zivkovic M, Stankovic A, Jovanovic I. A preliminary study of the miRNA restitution effect on CNV-induced miRNA downregulation in CAKUT. BMC Genomics 2024; 25:218. [PMID: 38413914 PMCID: PMC10900603 DOI: 10.1186/s12864-024-10121-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/14/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The majority of CAKUT-associated CNVs overlap at least one miRNA gene, thus affecting the cellular levels of the corresponding miRNA. We aimed to investigate the potency of restitution of CNV-affected miRNA levels to remediate the dysregulated expression of target genes involved in kidney physiology and development in vitro. METHODS Heterozygous MIR484 knockout HEK293 and homozygous MIR185 knockout HEK293 cell lines were used as models depicting the deletion of the frequently affected miRNA genes by CAKUT-associated CNVs. After treatment with the corresponding miRNA mimics, the levels of the target genes have been compared to the non-targeting control treatment. For both investigated miRNAs, MDM2 and PKD1 were evaluated as common targets, while additional 3 genes were investigated as targets of each individual miRNA (NOTCH3, FIS1 and APAF1 as hsa-miR-484 targets and RHOA, ATF6 and CDC42 as hsa-miR-185-5p targets). RESULTS Restitution of the corresponding miRNA levels in both knockout cell lines has induced a change in the mRNA levels of certain candidate target genes, thus confirming the potential to alleviate the CNV effect on miRNA expression. Intriguingly, HEK293 WT treatment with investigated miRNA mimics has triggered a more pronounced effect, thus suggesting the importance of miRNA interplay in different genomic contexts. CONCLUSIONS Dysregulation of multiple mRNA targets mediated by CNV-affected miRNAs could represent the underlying mechanism behind the unresolved CAKUT occurrence and phenotypic variability observed in CAKUT patients. Characterizing miRNAs located in CNVs and their potential to become molecular targets could eventually help in understanding and improving the management of CAKUT.
Collapse
Affiliation(s)
- Kristina Mitrovic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Ivan Zivotic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Ivana Kolic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Jelena Zakula
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Maja Zivkovic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Aleksandra Stankovic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia
| | - Ivan Jovanovic
- Department of Radiobiology and Molecular Genetics, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001, Belgrade, P.O. Box 522, Serbia.
| |
Collapse
|
5
|
Al-Imam MJ, Hussein UAR, Sead FF, Faqri AMA, Mekkey SM, Khazel AJ, Almashhadani HA. The interactions between DNA methylation machinery and long non-coding RNAs in tumor progression and drug resistance. DNA Repair (Amst) 2023; 128:103526. [PMID: 37406581 DOI: 10.1016/j.dnarep.2023.103526] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
DNA methylation is one of the main epigenetic mechanisms in cancer development and progression. Aberrant DNA methylation of CpG islands within promoter regions contributes to the dysregulation of various tumor suppressors and oncogenes; this leads to the appearance of malignant features, including rapid proliferation, metastasis, stemness, and drug resistance. The discovery of two important protein families, DNA methyltransferases (DNMTs) and Ten-eleven translocation (TET) dioxygenases, respectively, which are responsible for deregulated transcription of genes that play pivotal roles in tumorigenesis, led to further understanding of DNA methylation-related pathways. But how these enzymes can target specific genes in different malignancies; recent studies have highlighted the considerable role of Long Non-coding RNAs (LncRNAs). LncRNAs recruit these enzymes to promoter regions of genes and mediate their functions, showing great potential as therapeutic agents targeting the epigenetic regulation of various genes. Considering the importance of combining the current treatment methods, especially chemotherapies, with DNA methylation inhibitors in improving patients' outcomes, this review aimed to summarize the recent findings about the interaction between DNA methylation machinery and LncRNAs in regulating genes involved in tumorigenesis and drug resistance. So, these studies could provide insights toward developing novel strategies for cancer-targeted therapy.
Collapse
Affiliation(s)
- Mokhtar Jawad Al-Imam
- Department of Experimental Therapy, Iraqi Center for Cancer and Medical Genetics Research, Almustansiriyah University, Baghdad, Iraq
| | | | | | | | - Shereen M Mekkey
- Pharmacy Department, Al-Mustaqbal University College, 51001 Hilla, Babylon, Iraq
| | | | | |
Collapse
|
6
|
Leung DHL, Phon BWS, Sivalingam M, Radhakrishnan AK, Kamarudin MNA. Regulation of EMT Markers, Extracellular Matrix, and Associated Signalling Pathways by Long Non-Coding RNAs in Glioblastoma Mesenchymal Transition: A Scoping Review. BIOLOGY 2023; 12:818. [PMID: 37372103 PMCID: PMC10294841 DOI: 10.3390/biology12060818] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
Glioblastoma (GBM) mesenchymal (MES) transition can be regulated by long non-coding RNAs (lncRNAs) via modulation of various factors (Epithelial-to-Mesenchymal (EMT) markers, biological signalling, and the extracellular matrix (ECM)). However, understanding of these mechanisms in terms of lncRNAs is largely sparse. This review systematically analysed the mechanisms by which lncRNAs influence MES transition in GBM from a systematic search of the literature (using PRISMA) performed in five databases (PubMed, MEDLINE, EMBASE, Scopus, and Web of Science). We identified a total of 62 lncRNAs affiliated with GBM MES transition, of which 52 were upregulated and 10 were downregulated in GBM cells, where 55 lncRNAs were identified to regulate classical EMT markers in GBM (E-cadherin, N-cadherin, and vimentin) and 25 lncRNAs were reported to regulate EMT transcription factors (ZEB1, Snai1, Slug, Twist, and Notch); a total of 16 lncRNAs were found to regulate the associated signalling pathways (Wnt/β-catenin, PI3k/Akt/mTOR, TGFβ, and NF-κB) and 14 lncRNAs were reported to regulate ECM components (MMP2/9, fibronectin, CD44, and integrin-β1). A total of 25 lncRNAs were found dysregulated in clinical samples (TCGA vs. GTEx), of which 17 were upregulated and 8 were downregulated. Gene set enrichment analysis predicted the functions of HOXAS3, H19, HOTTIP, MEG3, DGCR5, and XIST at the transcriptional and translational levels based on their interacting target proteins. Our analysis observed that the MES transition is regulated by complex interplays between the signalling pathways and EMT factors. Nevertheless, further empirical studies are required to elucidate the complexity in this process between these EMT factors and the signalling involved in the GBM MES transition.
Collapse
Affiliation(s)
| | | | | | | | - Muhamad Noor Alfarizal Kamarudin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia
| |
Collapse
|
7
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
8
|
Stein RA, Thompson LM. Epigenetic changes induced by pathogenic Chlamydia spp. Pathog Dis 2023; 81:ftad034. [PMID: 38031337 DOI: 10.1093/femspd/ftad034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023] Open
Abstract
Chlamydia trachomatis, C. pneumoniae, and C. psittaci, the three Chlamydia species known to cause human disease, have been collectively linked to several pathologies, including conjunctivitis, trachoma, respiratory disease, acute and chronic urogenital infections and their complications, and psittacosis. In vitro, animal, and human studies also established additional correlations, such as between C. pneumoniae and atherosclerosis and between C. trachomatis and ovarian cancer. As part of their survival and pathogenesis strategies as obligate intracellular bacteria, Chlamydia spp. modulate all three major types of epigenetic changes, which include deoxyribonucleic acid (DNA) methylation, histone post-translational modifications, and microRNA-mediated gene silencing. Some of these epigenetic changes may be implicated in key aspects of pathogenesis, such as the ability of the Chlamydia spp. to induce epithelial-to-mesenchymal transition, interfere with DNA damage repair, suppress cholesterol efflux from infected macrophages, act as a co-factor in human papillomavirus (HPV)-mediated cervical cancer, prevent apoptosis, and preserve the integrity of mitochondrial networks in infected host cells. A better understanding of the individual and collective contribution of epigenetic changes to pathogenesis will enhance our knowledge about the biology of Chlamydia spp. and facilitate the development of novel therapies and biomarkers. Pathogenic Chlamydia spp. contribute to epigenetically-mediated gene expression changes in host cells by multiple mechanisms.
Collapse
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, United States
| | - Lily M Thompson
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, United States
| |
Collapse
|
9
|
Wang N, Gao X, Ji H, Ma S, Wu J, Dong J, Wang F, Zhao H, Liu Z, Yan X, Li B, Du J, Zhang J, Hu S. Machine learning-based screening of an epithelial-mesenchymal transition-related long non-coding RNA signature reveals lower-grade glioma prognosis and the tumor microenvironment and predicts antitumor therapy response. Front Mol Biosci 2022; 9:942966. [PMID: 36090045 PMCID: PMC9459009 DOI: 10.3389/fmolb.2022.942966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) confers high invasive and migratory capacity to cancer cells, which limits the effectiveness of tumor therapy. Long non-coding RNAs (lncRNAs) can regulate the dynamic process of EMT at different levels through various complex regulatory networks. We aimed to comprehensively analyze and screen EMT-related lncRNAs to characterize lower-grade glioma (LGG) tumor biology and provide new ideas for current therapeutic approaches. We retrieved 1065 LGG samples from the Cancer Genome Atlas and Chinese Glioma Genome Atlas by machine learning algorithms, identified three hub lncRNAs including CRNDE, LINC00665, and NEAT1, and established an EMT-related lncRNA signature (EMTrLS). This novel signature had strong prognostic value and potential clinical significance. EMTrLS described LGG genomic alterations and clinical features including gene mutations, tumor mutational burden, World Health Organization (WHO) grade, IDH status, and 1p/19q status. Notably, stratified analysis revealed activation of malignancy-related and metabolic pathways in the EMTrLS-high cohort. Moreover, the population with increased EMTrLS scores had increased cells with immune killing function. However, this antitumor immune function may be suppressed by increased Tregs and macrophages. Meanwhile, the relatively high expression of immune checkpoints explained the immunosuppressive state of patients with high EMTrLS scores. Importantly, we validated this result by quantifying the course of antitumor immunity. In particular, EMTrLS stratification enabled assessment of the responsiveness of LGG to chemotherapeutic drug efficacy and PD1 blockade. In conclusion, our findings complement the foundation of molecular studies of LGG, provide valuable insight into our understanding of EMT-related lncRNAs, and offer new strategies for LGG therapy.
Collapse
Affiliation(s)
- Nan Wang
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hang Ji
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai Ma
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Jiasheng Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Dong
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Fang Wang
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Hongtao Zhao
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Zhihui Liu
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Xiuwei Yan
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
| | - Bo Li
- Department of Neurosurgery, Taizhou First People’s Hospital, Taizhou, China
| | - Jianyang Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jiheng Zhang, ; Jianyang Du, ; Shaoshan Hu,
| | - Jiheng Zhang
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jiheng Zhang, ; Jianyang Du, ; Shaoshan Hu,
| | - Shaoshan Hu
- Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People’s Hospital, Affiliated to Hangzhou Medical College, Hangzhou, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jiheng Zhang, ; Jianyang Du, ; Shaoshan Hu,
| |
Collapse
|
10
|
Gu P, Lin L. Long Intergenic Nonprotein Coding RNA 00174 Aggravates Lung Squamous Cell Carcinoma Progression via MicroRNA-185-5p/Nuclear Factor IX axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9490827. [PMID: 36060149 PMCID: PMC9436563 DOI: 10.1155/2022/9490827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/13/2022] [Accepted: 06/25/2022] [Indexed: 12/05/2022]
Abstract
Extensive studies have presented that long noncoding RNAs (lncRNAs) are closely implicated in the pathogenesis of various human malignancies, including lung squamous cell carcinoma (LUSC). This study explored the biological role and the underlying mechanism of long intergenic nonprotein coding RNA 00174 (LINC00174) in LUSC. LINC00174 expression was measured by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR). Both in vitro and in vivo experiments were conducted to determine LINC00174 function in LUSC. Mechanical assays were performed to investigate the molecular mechanism involving LINC00174 and related genes. LINC00174 expression was high in LUSC cells. Silencing of LINC00174 could restrain LUSC cells proliferation, migration, and invasion while promoting cell apoptosis. Mechanically, LINC00174 could interact with miR-185-5p to upregulate nuclear factor IX (NFIX), which was the direct target gene of miR-185-5p. Notably, NFIX elevation could rescue the repressing effect of LINC00174 silence on LUSC cell malignant behaviors. Our data suggested that LINC00174 aggravated LUSC progression via serving as a competing endogenous RNA (ceRNA) to sponge miR-185-5p and ultimately upregulate NFIX, which offered a promising novel target for LUSC therapy.
Collapse
Affiliation(s)
- Peipei Gu
- Department of Thoracic Surgery, The No. 4 Hospital, No. 50 Shifan Road, Tianqiao District, Jinan, 250000 Shandong, China
| | - Lin Lin
- Department of Thoracic Surgery, The No. 4 Hospital, No. 50 Shifan Road, Tianqiao District, Jinan, 250000 Shandong, China
| |
Collapse
|
11
|
Molecular Interactions of the Long Noncoding RNA NEAT1 in Cancer. Cancers (Basel) 2022; 14:cancers14164009. [PMID: 36011001 PMCID: PMC9406559 DOI: 10.3390/cancers14164009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
As one of the best-studied long noncoding RNAs, nuclear paraspeckle assembly transcript 1 (NEAT1) plays a pivotal role in the progression of cancers. NEAT1, especially its isoform NEAT1-1, facilitates the growth and metastasis of various cancers, excluding acute promyelocytic leukemia. NEAT1 can be elevated via transcriptional activation or stability alteration in cancers changing the aggressive phenotype of cancer cells. NEAT1 can also be secreted from other cells and be delivered to cancer cells through exosomes. Hence, elucidating the molecular interaction of NEAT1 may shed light on the future treatment of cancer. Herein, we review the molecular function of NEAT1 in cancer progression, and explain how NEAT1 interacts with RNAs, proteins, and DNA promoter regions to upregulate tumorigenic factors.
Collapse
|
12
|
Zeng C, Hu J, Chen F, Huang T, Zhang L. The Coordination of mTOR Signaling and Non-Coding RNA in Regulating Epileptic Neuroinflammation. Front Immunol 2022; 13:924642. [PMID: 35898503 PMCID: PMC9310657 DOI: 10.3389/fimmu.2022.924642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy accounts for a significant proportion of the burden of neurological disorders. Neuroinflammation acting as the inflammatory response to epileptic seizures is characterized by aberrant regulation of inflammatory cells and molecules, and has been regarded as a key process in epilepsy where mTOR signaling serves as a pivotal modulator. Meanwhile, accumulating evidence has revealed that non-coding RNAs (ncRNAs) interfering with mTOR signaling are involved in neuroinflammation and therefore articipate in the development and progression of epilepsy. In this review, we highlight recent advances in the regulation of mTOR on neuroinflammatory cells and mediators, and feature the progresses of the interaction between ncRNAs and mTOR in epileptic neuroinflammation.
Collapse
Affiliation(s)
- Chudai Zeng
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jason Hu
- Department of Neonatology, Yale School of Medicine, New Haven, CT, United States
| | - Fenghua Chen
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| | - Tianxiang Huang
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| | - Longbo Zhang
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| |
Collapse
|
13
|
Lee WJ, Ji H, Jeong SD, Pandey PR, Gorospe M, Kim HH. LINC00162 regulates cell proliferation and apoptosis by sponging PAQR4-targeting miR-485-5p. J Cell Physiol 2022; 237:2943-2960. [PMID: 35491694 PMCID: PMC9846112 DOI: 10.1002/jcp.30758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 01/21/2023]
Abstract
Growing evidence indicates that long intergenic noncoding RNAs play an important role in cancer progression by affecting gene regulation at the transcriptional and posttranscriptional levels. Recent studies have shown that long intergenic noncoding RNA functions as a competitive endogenous RNA, which can interact with and mitigate the function of microRNA. In this study, we investigated the molecular mechanism by which LINC00162 regulates cell proliferation and apoptotic cell death. By analyzing RNA sequencing data, LINC00162 was identified to be a target of heterogeneous nuclear ribonucleoprotein K (hnRNPK). HnRNPK positively regulated LINC00162 expression through p38 mitogen-activated protein kinase. Lowering the level of either hnRNPK or LINC00162 decreased proliferation and colony formation while it increased apoptotic cell death. Small RNA sequencing followed by the antisense oligonucleotide pulldown, revealed that LINC00162 interacts directly with miR-485-5p which exhibited tumor-suppressing effects by suppressing cell proliferation and colony formation, and increasing apoptotic cell death. Through the bioinformatic approaches, progestin and adipoQ receptor 4 (PAQR4) was selected as a common target of LINC00162 and miR-485-5p. miR-485-5p decreased the expression of PAQR4 by directly binding to the 3'-untranslated region of PAQR4 messenger RNA. Knockdown of hnRNPK and LINC00162 increased the level of functional miR-485-5p, indicating that LINC00162 may compete for miR-485-5p, thereby derepressing PAQR4 expression. Overexpression of either hnRNPK or LINC00162, or inhibition of miR-485-5p, protected cells against etoposide-induced apoptotic death. Our findings demonstrate that a regulatory paradigm implicating hnRNPK, LINC00162, miR-485-5p, and PAQR4 plays an important role in cell proliferation and apoptosis, and is a promising target for cancer therapeutics.
Collapse
Affiliation(s)
- Woo Joo Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Haein Ji
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Seong Dong Jeong
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea,Department of Biopharmaceutical Convergence, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Poonam R Pandey
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hyeon Ho Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea,Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea,Correspondence: Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea. Phone: +82-2-3410-1039; Fax: +82-2-3410-0534;
| |
Collapse
|
14
|
Wang S, Sun Y, Hu S, Lou C, Pan YB. Construction of a lncRNA-associated competing endogenous RNA regulatory network after traumatic brain injury in mouse. Mol Brain 2022; 15:40. [PMID: 35501920 PMCID: PMC9063179 DOI: 10.1186/s13041-022-00925-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/19/2022] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem worldwide which causes high mortality and disability. Functioning as microRNA (miRNA) sponges, long non-coding RNA (lncRNA) regulates the expression of protein-coding genes in a competing endogenous RNA (ceRNA) network. However, the lncRNA-associated ceRNA in TBI remains unclear. In this study, we processed the raw SRR files of mice cortex samples of sham injury (n = 3) and TBI groups (n = 3) to count files. Then, the expression profiles of lncRNAs and mRNAs were identified, and 86 differentially expressed (DE) lncRNAs and 1201 DEmRNAs between sham and TBI groups were identified. The DEmRNAs were used to perform enrichment analyses. Next, a lncRNA-miRNA-mRNA regulatory ceRNA network was constructed. The network consisted of 23 mRNAs, 5 miRNAs and 2 lncRNAs. The expression alternations of the 5 miRNAs were validated via qRT-PCR. The subnetwork of hub lncRNA Neat1 was extracted. We identified a potential inflammatory associated regulatory axis: Neat1/miR-31-5p/Myd88 axis. The PPI network based on DEmRNA involved in ceRNA network was constructed PPI networks to identify the hub genes. Finally, DElncRNAs and DEmRNAs were selected randomly and validated by qRT-PCR. In conclusion, with the lncRNA-miRNA-mRNA ceRNA network provided above, we can improve our understanding of the regulatory mechanisms and interaction among lncRNAs, miRNAs and mRNAs in TBI process.
Collapse
|
15
|
Liu RJ, Xu ZP, Li SY, Yu JJ, Feng NH, Xu B, Chen M. BAP1-Related ceRNA (NEAT1/miR-10a-5p/SERPINE1) Promotes Proliferation and Migration of Kidney Cancer Cells. Front Oncol 2022; 12:852515. [PMID: 35425712 PMCID: PMC9004599 DOI: 10.3389/fonc.2022.852515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/25/2022] [Indexed: 01/09/2023] Open
Abstract
Background BAP1 is an important tumor suppressor involved in various biological processes and is commonly lost or inactivated in clear-cell renal cell carcinoma (ccRCC). However, the role of the BAP1-deficient tumor competing endogenous RNA (ceRNA) network involved in ccRCC remains unclear. Thus, this study aims to investigate the prognostic BAP1-related ceRNA in ccRCC. Methods Raw data was obtained from the TCGA and the differentially expressed genes were screened to establish a BAP1-related ceRNA network. Subsequently, the role of the ceRNA axis was validated using phenotypic experiments. Dual-luciferase reporter assays and fluorescence in situ hybridization (FISH) assays were used to confirm the ceRNA network. Results Nuclear enriched abundant transcript 1 (NEAT1) expression was significantly increased in kidney cancer cell lines. NEAT1 knockdown significantly inhibited cell proliferation and migration, which could be reversed by miR-10a-5p inhibitor. Dual-luciferase reporter assay confirmed miR-10a-5p as a common target of NEAT1 and Serine protease inhibitor family E member 1 (SERPINE1). FISH assays revealed the co-localization of NEAT1 and miR-10a-5p in the cytoplasm. Additionally, the methylation level of SERPINE1 in ccRCC was significantly lower than that in normal tissues. Furthermore, SERPINE1 expression was positively correlated with multiple immune cell infiltration levels. Conclusions In BAP1-deficient ccRCC, NEAT1 competitively binds to miR-10a-5p, indirectly upregulating SERPINE1 expression to promote kidney cancer cell proliferation. Furthermore, NEAT1/miR-10a-5p/SERPINE1 were found to be independent prognostic factors of ccRCC.
Collapse
Affiliation(s)
- Rui-Ji Liu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Zhi-Peng Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Shu-Ying Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital affiliate to School of Medicine, UESTC, Chengdu, China
| | - Jun-Jie Yu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Ning-Han Feng
- Department of Urology, Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.,Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
16
|
LncRNA: a new perspective on the study of neurological diseases. Biochem Soc Trans 2022; 50:951-963. [PMID: 35383841 DOI: 10.1042/bst20211181] [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: 10/15/2021] [Revised: 02/21/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022]
Abstract
Long non-coding RNAs (lncRNAs) are a class of non-coding RNA with a length greater than 200 nt. It has a mRNA-like structure, formed by splicing after transcription, and contains a polyA tail and a promoter, of whom promoter plays a role by binding transcription factors. LncRNAs' sequences are low in conservation, and other species can only find a handful of the same lncRNAs as humans, and there are different splicing ways during the differentiation of identical species, with spatiotemporal expression specificity. With developing high-throughput sequencing and bioinformatics, found that more and more lncRNAs associated with nervous system disease. This article deals with the regulation of certain lncRNAs in the nervous system disease, by mean of to understand its mechanism of action, and the pathogenesis of some neurological diseases have a fresh understanding, deposit a foundation for resulting research and clinical treatment of disease.
Collapse
|
17
|
Lin S, Wen Z, Li S, Chen Z, Li C, Ouyang Z, Lin C, Kuang M, Xue C, Ding Y. LncRNA Neat1 promotes the macrophage inflammatory response and acts as a therapeutic target in titanium particle-induced osteolysis. Acta Biomater 2022; 142:345-360. [PMID: 35151924 DOI: 10.1016/j.actbio.2022.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
Aseptic loosening (AL), secondary to particle-caused periprosthetic osteolysis, is one of the main reasons of artificial joint failure. Suppressing the macrophage inflammatory response caused by wear particles extends the life of prosthesis, and the long noncoding RNAs (lncRNAs) may play a predominant part in it. Here, titanium particles' (TiPs') stimulation increases both the cytoplasmic and nuclear levels of lncRNA Neat1 in bone marrow derived macrophages (BMDMs), which further induces the inflammatory response. Mechanically, Neat1 facilitates Bruton's tyrosine kinase (BTK) transcription by reducing the transcriptional factor KLF4, which further activates the NF-κB pathway, NLRP3 inflammation, and M1 polarization in BMDMs. Cytoplasmic Neat1 also works as an miRNA sponge in miR-188-5p-regulated BTK expression in the post-transcriptional stage. In vivo, Neat1 downregulation can reduce the TiP-induced pro-inflammatory factors and reverse the osteolysis induced by BTK overexpression. In addition, the PLGA-based microparticles loaded with si-Neat1 are developed for the treatment of the mouse calvarial osteolysis model via local injection, presenting satisfactory anti-osteolysis efficacy. These findings indicate that Neat1 is a key regulator of AL. STATEMENT OF SIGNIFICANCE: Due to released particles, aseptic loosening (AL) is the most common reason for prosthesis failure and surgical revision and represents a substantial economic burden worldwide. Herein, we reported that lncRNA Neat1 is a key regulator in regulating wear particles-induced osteolysis by activating NF-κB pathway, NLRP3 inflammation and M1 polarization via BTK, and the underlying mechanisms of Neat1-BTK interaction were further portrayed. For potential clinical application, the microparticles are developed for effective si-Neat1 delivery, leading to a dramatically enhanced effect for the treatment of osteolysis, which might be a novel strategy to extend the life of the implant.
Collapse
|
18
|
Zhou H, Wang Y, Liu Z, Zhang Z, Xiong L, Wen Y. Recent advances of NEAT1-miRNA interactions in cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:153-162. [PMID: 35538025 PMCID: PMC9827865 DOI: 10.3724/abbs.2021022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022] Open
Abstract
With high incidence rate, cancer is the main cause of death in humans. Non-coding RNAs, as novel master regulators, especially long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), play important roles in the regulation of tumorigenesis. lncRNA NEAT1 has recently gained much attention, as it is dysregulated in a broad spectrum of cancers, where it acts as either an oncogene or a tumor suppressor gene. Accumulating evidence shows that NEAT1 is correlated with the process of carcinogenesis, including proliferation, invasion, survival, drug resistance, and metastasis. NEAT1 is considered to be a biomarker and a novel therapeutic target for the diagnosis and prognosis of different cancer types. The mechanisms by which NEAT1 plays a critical role in cancers are mainly via interactions with miRNAs. NEAT1-miRNA regulatory networks play significant roles in tumorigenesis, which has attracted much attention from researchers around the world. In this review, we summarize the interaction of NEAT1 with miRNAs in the regulation of protein-coding genes in cancer. A better understanding of the NEAT1-miRNA interactions in cancer will help develop new diagnostic biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Hui Zhou
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Yongxiang Wang
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Zhongtao Liu
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Zijian Zhang
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Li Xiong
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Yu Wen
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| |
Collapse
|
19
|
Liang J, Liu C, Xu D, Xie K, Li A. LncRNA NEAT1 facilitates glioma progression via stabilizing PGK1. J Transl Med 2022; 20:80. [PMID: 35123484 PMCID: PMC8818213 DOI: 10.1186/s12967-022-03273-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/23/2022] [Indexed: 01/09/2023] Open
Abstract
Background Long noncoding RNA NEAT1 has been implicated in glioma progression. However, the effect of NEAT1 on glycolysis of glioma cell and the potential mechanism remain unclear. Methods In vitro experiments, including CCK-8, colony formation, ECAR, and lactate detection assays were performed to evaluate the effect of NEAT1 on proliferation and glycolysis of glioma cell. RNA pulldown and RIP assays were performed to identify the interaction between NEAT1 and PGK1. Truncated mutation of NEAT1 and PGK1 was used to confirm the specific interactive domains between NEAT1 and PGK1. Animal studies were performed to analyze the effect of NEAT1/PGK1 on glioma progression. Results NEAT1 knockdown significantly suppressed the proliferation and glycolysis of glioma cells. NEAT1 could specifically interact with PGK1, which promotes PGK1 stability. Hairpin A of NEAT1 is essential for interaction with M1 domain of PGK1. Depletion of NEAT1 markedly inhibited tumor growth in mice, while PGK1 could reverse this effect. Higher expression of NEAT1 was associated with poor overall survival of GBM patients. Conclusions NEAT1 over expression promotes glioma progression through stabilizing PGK1. NEAT1/PGK1 axis is a candidate therapeutic target for glioma treatment.
Collapse
|
20
|
Guan Y, Yang W, Zhang F, Zhang L, Wang L. CircPOSTN competes with KIF1B for miR-185-5p binding sites to promote the tumorigenesis of glioma. Brain Res Bull 2021; 180:86-96. [PMID: 34974134 DOI: 10.1016/j.brainresbull.2021.12.014] [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: 05/17/2021] [Revised: 10/24/2021] [Accepted: 12/28/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND The involvement of certain circular RNAs (circRNAs) in the development of glioma has been revealed. CircRNA periostin (circPOSTN) was validated to be positively associated with glioma cell growth and metastasis. However, the mechanism underlying circPOSTN in glioma tumorigenesis remain vague. METHODS The expression of circPOSTN, KIF1B (Kinesin Family Member 1B) and miR-185-5p was detected using quantitative real-time polymerase chain reaction and Western blot. In vitro assays were conducted using cell counting kit-8 assay, colony formation assay, EdU assay, flow cytometry, Western blot, and transwell assay, respectively. The direct interactions between miR-185-5p and circPOSTN or KIF1B was confirmed by using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. RESULTS CircPOSTN was highly expressed in glioma tissues and cells. Knockdown of circPOSTN restrained glioma cell proliferation, migration and invasion in vitro, as well as hindered glioma xenograft growth in vivo. Mechanistically, circPOSTN acted as miR-185-5p sponge to up-regulate the expression of its target KIF1B. Moreover, miR-185-5p inhibition reversed the anticancer effects of circPOSTN knockdown on glioma tumorigenesis, and miR-185-5p re-expression suppressed the malignant phenotype of glioma cells via targeting KIF1B. CONCLUSION CircPOSTN acted as an oncogene to expedite glioma tumorigenesis via targeting miR-185-5p/KIF1B axis, indicating a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Yongchang Guan
- Department of Neurosurgery, the Fourth Affiliated Hospital of China Medical University.
| | - Wenjin Yang
- Department of Neurosurgery, Pudong New Area People's Hospital
| | - Feng Zhang
- Department of Neurosurgery, the Fourth Affiliated Hospital of China Medical University
| | - Liming Zhang
- Department of Neurosurgery, the Fourth Affiliated Hospital of China Medical University
| | - Liang Wang
- Department of Neurosurgery, the Fourth Affiliated Hospital of China Medical University
| |
Collapse
|
21
|
Construction of a ceRNA network in glioma and analysis of its clinical significance. BMC Genomics 2021; 22:722. [PMID: 34615480 PMCID: PMC8496082 DOI: 10.1186/s12864-021-08035-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 09/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background Glioma is the most common central nervous system tumor with a poor survival rate and prognosis. Previous studies have found that long non-coding RNA (lncRNA) and competitive endogenous RNA (ceRNA) play important roles in regulating various tumor mechanisms. We obtained RNA-Seq data of glioma and normal brain tissue samples from TCGA and GTEx databases and extracted the lncRNA and mRNA expression data. Further, we analyzed these data using weighted gene co-expression network analysis and differential expression analysis, respectively. Differential expression analysis was also carried out on the mRNA data from the GEO database. Further, we predicted the interactions between lncRNA, miRNA, and targeted mRNA. Using the CGGA data to perform univariate and multivariate Cox regression analysis on mRNA. Results We constructed a Cox proportional hazard regression model containing four mRNAs and performed immune infiltration analysis. Moreover, we also constructed a ceRNA network including 21 lncRNAs, two miRNAs, and four mRNAs, and identified seven lncRNAs related to survival that have not been previously studied in gliomas. Through the gene set enrichment analysis, we found four lncRNAs that may have a significant role in tumors and should be explored further in the context of gliomas. Conclusions In short, we identified four lncRNAs with research value for gliomas, constructed a ceRNA network in gliomas, and developed a prognostic prediction model. Our research enhances our understanding of the molecular mechanisms underlying gliomas, providing new insights for developing targeted therapies and efficiently evaluating the prognosis of gliomas. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08035-w.
Collapse
|
22
|
Jia G, Wang Y, Lin C, Lai S, Dai H, Wang Z, Dai L, Su H, Song Y, Zhang N, Feng Y, Tang B. LNCAROD enhances hepatocellular carcinoma malignancy by activating glycolysis through induction of pyruvate kinase isoform PKM2. J Exp Clin Cancer Res 2021; 40:299. [PMID: 34551796 PMCID: PMC8459495 DOI: 10.1186/s13046-021-02090-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mounting evidence has suggested the essential role of long non-coding RNAs (lncRNAs) in a plethora of malignant tumors, including hepatocellular carcinoma. However, the underlyling mechanisms of lncRNAs remain unidentified in HCC. The present work was aimed to explore the regulatory functions and mechanisms of LncRNA LNCAROD in HCC progression and chemotherapeutic response. METHODS The expression of LNCAROD in HCC tissues and cell lines were detected by quantitative reverse transcription PCR (qPCR). Cancer cell proliferation, migration, invasion, and chemoresistance were evaluated by cell counting kit 8 (CCK8), colony formation, transwell, and chemosensitivity assays. Methylated RNA immunoprecipitation qRCR (MeRIP-qPCR) was used to determine N6-methyladenosine (m6A) modification level. RNA immunoprecipitation (RIP) and RNA pull down were applied to identify the molecular sponge role of LNCAROD for modulation of miR-145-5p via the competing endogenous RNA (ceRNA) mechanism, as well as the interaction between LNCAROD and serine-and arginine-rich splicing factor 3 (SRSF3). The interaction between insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and LNCAROD was also identified by RIP assay. Gain- or-loss-of-function assays were used to identify the function and underlying mechanisms of LNCAROD in HCC. RESULTS We found that LNCAROD was significantly upregulated and predicted a poorer prognosis in HCC patients. LNCAROD upregulation was maintained by increased m6A methylation-mediated RNA stability. LNCAROD significantly promoted HCC cell proliferation, migration, invasion, and chemoresistance both in vitro and in vivo. Furthermore, mechanistic studies revealed that pyruvate kinase isoform M2 (PKM2)-mediated glycolysis enhancement is critical for the role of LNACROD in HCC. According to bioinformatics prediction and our experimental data, LNCAROD directly binds to SRSF3 to induce PKM switching towards PKM2 and maintains PKM2 levels in HCC by acting as a ceRNA against miR-145-5p. The oncogenic effects of LNCAROD in HCC were more prominent under hypoxia than normoxia due to the upregulation of hypoxia-triggered hypoxia-inducible factor 1α. CONCLUSIONS In summary, our present study suggests that LNCAROD induces PKM2 upregulation via simultaneously enhancing SRSF3-mediated PKM switching to PKM2 and sponging miR-145-5p to increase PKM2 level, eventually increasing cancer cell aerobic glycolysis to participate in tumor malignancy and chemoresistance, especially under hypoxic microenvironment. This study provides a promising diagnostic marker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Guizhi Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Yan Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Chengjie Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Shihui Lai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Hongliang Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Zhiqian Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Luo Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Huizhao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China
| | - Yanjie Song
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, People's Republic of China
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, School of Basic Medicine, Mudanjiang Medical University, 157011, Mudanjiang, People's Republic of China
| | - Naiwen Zhang
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, People's Republic of China
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, School of Basic Medicine, Mudanjiang Medical University, 157011, Mudanjiang, People's Republic of China
| | - Yukuan Feng
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, 300060, Tianjin, People's Republic of China.
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, School of Basic Medicine, Mudanjiang Medical University, 157011, Mudanjiang, People's Republic of China.
| | - Bo Tang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, People's Republic of China.
- Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases of Guangxi, 530021, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
23
|
Li K, Yao T, Zhang Y, Li W, Wang Z. NEAT1 as a competing endogenous RNA in tumorigenesis of various cancers: Role, mechanism and therapeutic potential. Int J Biol Sci 2021; 17:3428-3440. [PMID: 34512157 PMCID: PMC8416723 DOI: 10.7150/ijbs.62728] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/24/2021] [Indexed: 12/24/2022] Open
Abstract
The nuclear paraspeckle assembly transcript 1 (NEAT1) is a long non-coding RNA (lncRNA) that is upregulated in a variety of human cancer types. Increasing evidence has shown that the elevation of NEAT1 in cancer cells promotes cell growth, migration, and invasion and inhibits cell apoptosis. It is also known that lncRNAs act as a competing endogenous RNA (ceRNA) by sponging microRNAs (miRNAs) to alter the expression levels of their target genes in the development of cancers. Therefore, it is important to understand the molecular mechanisms underlying this observation. In this review, specific emphasis was placed on NEAT1's role in tumor development. We also summarize and discuss the feedback roles of NEAT1/miRNA/target network in the progression of various cancers. As our understanding of the role of NEAT1 during tumorigenesis improves, its therapeutic potential as a biomarker and/or target for cancer also becomes clearer.
Collapse
Affiliation(s)
- Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Tongyue Yao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yu Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wen Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Ziqiang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China.,Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| |
Collapse
|
24
|
Chen J, Wang H, Wang J, Niu W, Deng C, Zhou M. LncRNA NEAT1 Enhances Glioma Progression via Regulating the miR-128-3p/ITGA5 Axis. Mol Neurobiol 2021; 58:5163-5177. [PMID: 34263426 PMCID: PMC8497354 DOI: 10.1007/s12035-021-02474-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022]
Abstract
Accumulating evidences indicate that long non-coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) promotes the progression of glioma. In this study, we postulated that NEAT1 may act as a miR-128-3p sponge. Relative levels of NEAT1 and miR-128-3p expression in human glioma samples and GBM cells were detected using quantitative real-time PCR. By means of CCK-8 assays, transwell assays, and flow cytometric analysis, the biological functions of miR-128-3p and NEAT1 were investigated in U87MG and U251MG human GBM cell lines with stable miR-128-3p and NEAT1 knockdown or overexpression. The luciferase reports, RNA pull-down assay, and RNA immunoprecipitation assay were conducted to determine the relevance of NEAT1 and miR-128-3p in glioma. As a result, high expression of NEAT1 and lack of miR-128-3p were observed in glioma specimens and cells. By binding to anti-oncogene miR-128-3p in the nucleus, NEAT1 enhanced tumorigenesis and glioma development. Further experiments suggested that ITGA5 expression was increased in glioma tissues and was found to be connected with miR-128-3p. Additionally, NEAT1 facilitated ITGA5 expression via competitively binding to miR-128-3p. For this reason, ITGA5 would not be decomposed by miR-128-3p and could activate FAK signaling pathway, thereby promoting cell growth. Collectively, these results indicated that the NEAT1/miR-128-3p/ITGA5 axis was involved in glioma initiation and progression, and might offer a potential novel strategy for treatment of glioma.
Collapse
Affiliation(s)
- Jiakai Chen
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China.
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China.
| | - Wenhao Niu
- Department of Neurosurgery, Jinling Hospital, Medical School of Southeast University, 305 East Zhongshan Road, 210002, Nanjing, Jiangsu, People's Republic of China
| | - Chulei Deng
- Department of Neurosurgery, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Mengliang Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| |
Collapse
|
25
|
Momtazmanesh S, Rezaei N. Long Non-Coding RNAs in Diagnosis, Treatment, Prognosis, and Progression of Glioma: A State-of-the-Art Review. Front Oncol 2021; 11:712786. [PMID: 34322395 PMCID: PMC8311560 DOI: 10.3389/fonc.2021.712786] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioma is the most common malignant central nervous system tumor with significant mortality and morbidity. Despite considerable advances, the exact molecular pathways involved in tumor progression are not fully elucidated, and patients commonly face a poor prognosis. Long non-coding RNAs (lncRNAs) have recently drawn extra attention for their potential roles in different types of cancer as well as non-malignant diseases. More than 200 lncRNAs have been reported to be associated with glioma. We aimed to assess the roles of the most investigated lncRNAs in different stages of tumor progression and the mediating molecular pathways in addition to their clinical applications. lncRNAs are involved in different stages of tumor formation, invasion, and progression, including regulating the cell cycle, apoptosis, autophagy, epithelial-to-mesenchymal transition, tumor stemness, angiogenesis, the integrity of the blood-tumor-brain barrier, tumor metabolism, and immunological responses. The well-known oncogenic lncRNAs, which are upregulated in glioma, are H19, HOTAIR, PVT1, UCA1, XIST, CRNDE, FOXD2-AS1, ANRIL, HOXA11-AS, TP73-AS1, and DANCR. On the other hand, MEG3, GAS5, CCASC2, and TUSC7 are tumor suppressor lncRNAs, which are downregulated. While most studies reported oncogenic effects for MALAT1, TUG1, and NEAT1, there are some controversies regarding these lncRNAs. Expression levels of lncRNAs can be associated with tumor grade, survival, treatment response (chemotherapy drugs or radiotherapy), and overall prognosis. Moreover, circulatory levels of lncRNAs, such as MALAT1, H19, HOTAIR, NEAT1, TUG1, GAS5, LINK-A, and TUSC7, can provide non-invasive diagnostic and prognostic tools. Modulation of expression of lncRNAs using antisense oligonucleotides can lead to novel therapeutics. Notably, a profound understanding of the underlying molecular pathways involved in the function of lncRNAs is required to develop novel therapeutic targets. More investigations with large sample sizes and increased focus on in-vivo models are required to expand our understanding of the potential roles and application of lncRNAs in glioma.
Collapse
Affiliation(s)
- Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Wang F, Zhang L, Luo Y, Zhang Q, Zhang Y, Shao Y, Yuan L. The LncRNA RP11-279C4.1 Enhances the Malignant Behaviour of Glioma Cells and Glioma Stem-Like Cells by Regulating the miR-1273g-3p/CBX3 Axis. Mol Neurobiol 2021; 58:3362-3373. [PMID: 33694060 DOI: 10.1007/s12035-021-02337-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/17/2021] [Indexed: 12/19/2022]
Abstract
Glioma is the most common type of solid tumour affecting the central nervous system, and the survival rate of patients with glioma is low. However, the mechanism associated with glioma progression remains unclear. Growing evidence suggests that lncRNAs play essential roles in the initiation and progression of tumours, including gliomas. In the present study, we identified and verified the expression of the novel lncRNA RP11-279C4.1 by analyzing the TANRIC database and performing qRT-PCR assays, the results of which revealed its upregulation in glioma tissues and cell lines. The results of multiple functional experiments demonstrated that RP11-279C4.1 knockdown inhibited glioma malignant phenotypes, including cell proliferation, migration, invasion and cell self-renew ability in vitro. In addition, RP11-279C4.1 downregulation suppressed tumour growth in vivo. Mechanistically, RP11-279C4.1 induced CBX3 activation via competitively sponging miR-1273g-3p, and rescue assay results confirmed the importance of the RP11-279C4.1/miR-1273g-3p/CBX3 axis. Overall, the results of our present study demonstrated that RP11-279C4.1 functions as an oncogene that promotes tumour progression by modulating the miR-1273g-3p/CBX3 axis in glioma, suggesting that RP11-279C4.1 may be a novel therapeutic target for glioma.
Collapse
Affiliation(s)
- Faming Wang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao, Nanjing, 210009, China
| | - Le Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Yao Luo
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao, Nanjing, 210009, China
| | - Qingyun Zhang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yueling Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Yingying Shao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Liudi Yuan
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, # 87 Dingjiaqiao, Nanjing, 210009, China. .,Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
27
|
Zhan WL, Gao N, Tu GL, Tang H, Gao L, Xia Y. LncRNA LINC00689 Promotes the Tumorigenesis of Glioma via Mediation of miR-526b-3p/IGF2BP1 Axis. Neuromolecular Med 2021; 23:383-394. [PMID: 33389570 DOI: 10.1007/s12017-020-08635-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/22/2020] [Indexed: 12/14/2022]
Abstract
Glioma ranks first among the aggressive brain tumors all over the world. LncRNA LINC00689 has been confirmed to play key roles in the progression of cancers, and LINC00689 was upregulated in glioma. However, the biological function of LINC00689 in glioma is unclear. qRT-PCR was applied to detect the expressions of LINC00689 and miR-526b-3p in glioma cells. Dual-luciferase report was performed to examine the relation among LINC00689, miR-526b-3p, and insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1). Then, the growth, migration, and invasion of glioma cells were detected by colony formation, flow cytometry, and transwell assay, respectively. The expressions of p21, cleaved caspase 3, and MAPK signaling-related proteins in glioma cells were tested by western blotting. Finally, xenograft mice model was established to detect the effect of LINC00689 on tumor growth of glioma in vivo. LINC00689 was upregulated in glioma cells, while miR-526b-3p was downregulated. In addition, LINC00689 bound to miR-526b-3p, and IGFBP1 was targeted by miR-526b-3p. Moreover, LINC00689 knockdown or upregulation of miR-526b-3p inhibited the proliferation of glioma cells and induced the apoptosis. Consistently, the migration and invasion of glioma cells were notably reduced by LINC00689 shRNA/miR-526-3p mimics. miR-526b-3p inhibitor or IGF2BP1 upregulation could reverse the effect of LINC00689 knockdown or miR-526b-3p mimics. Finally, knockdown of LINC00689 inhibited the tumor growth of glioma in vivo through regulating miR-526b-3p/IGF2BP1/MAPK axis. In conclusion, silencing of LINC00689 could inhibit the tumorigenesis of glioma via mediation of miR-526b-3p/IGF2BP1 axis. LINC00689 may serve as a new target for the treatment of glioma.
Collapse
Affiliation(s)
- Wen-Liang Zhan
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China
| | - Ning Gao
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China
| | - Guo-Long Tu
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China
| | - Hong Tang
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China
| | - Ling Gao
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, No.43, People's Avenue, Haidian Island, Haikou, 570208, Hainan Province, People's Republic of China.
| |
Collapse
|