1
|
Liu Y, Li Q, Wu Y, Tang H, Zou C, Dou W, Zhang G. LINC01614 enhances neural invasion and gastric cancer progression by stabilizing VCAM1 and activating the AKT/mTOR pathway. Cell Signal 2025:111860. [PMID: 40349810 DOI: 10.1016/j.cellsig.2025.111860] [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: 02/24/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Gastric cancer (GC) is among the most fatal cancers worldwide. Neural infiltration (NI) is a poorly understood metastatic pathway with limited insights into its regulation and therapeutic targets. METHODS The GEPIA algorithm and RBPsuite analyzed the LINC01614-IGF2BP3-VCAM1 axis in the TCGA-STAD dataset. GC-NI clinical samples and cell lines validated LINC01614 expression and its clinical relevance. CCK-8, colony formation, flow cytometry, Transwell migration and dorsal root ganglia (DRG)-GC co-culture assays assessed LINC01614's role in cell proliferation, apoptosis, migration, invasion and NI. RNA immunoprecipitation, RNA pull-down combined with mass spectrometry, actinomycin D stability assays, and fluorescence in situ hybridization elucidated the formation of an RNA-protein ternary complex and its regulatory effects on VCAM1 stability and the AKT/mTOR pathway. An in vivo murine model was established to examine the impact of LINC01614 knockdown on tumor growth and NI, with tumor progression tracked using IVIS spectral imaging and immunohistochemical staining of GP9.5. RESULTS LINC01614 was significantly overexpressed in GC-NI-positive patients and correlated with poor prognosis, as validated in clinical samples and TCGA-STAD datasets. Functional assays demonstrated that LINC01614 knockdown inhibited GC cell proliferation, migration, invasion, and NI potential. Mechanistically, RNA pull-down followed by mass spectrometry identified IGF2BP3 as a key binding partner of LINC01614. Subsequent RNA immunoprecipitation assays showed that LINC01614 interacts with IGF2BP3 to form an RNA-protein ternary complex with VCAM1 in the nucleus, as further validated by fluorescence in situ hybridization-immunofluorescence. This complex stabilized VCAM1 mRNA, as demonstrated by actinomycin D assays, leading to its upregulation. Enhanced VCAM1 expression activated the AKT/mTOR pathway, which drove tumor progression and NI. Knockdown of LINC01614 disrupted this axis, reduced VCAM1 levels, attenuated AKT/mTOR pathway activation, and mitigated the malignant phenotype of GC cells. CONCLUSION LINC01614 drives NI in GC by stabilizing VCAM1 and activating the AKT/mTOR pathway, making it a promising biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yujing Liu
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Qian Li
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Yongyou Wu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Hongjie Tang
- Department of Surgery, The Fifth People's Hospital of Suzhou / The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215100, China
| | - Chen Zou
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Wenhuan Dou
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215000, China
| | - Guoqiang Zhang
- Department of Surgery, The Fifth People's Hospital of Suzhou / The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215100, China.
| |
Collapse
|
2
|
Che H, Zhang X, Cao L, Huang W, Lu Q. LINC01614 Promotes Oral Squamous Cell Carcinoma by Regulating FOXC1. Genes (Basel) 2024; 15:1461. [PMID: 39596660 PMCID: PMC11593781 DOI: 10.3390/genes15111461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are pivotal mediators during the development of carcinomas; however, it remains to be investigated whether lncRNAs are implicated in oral squamous cell carcinoma (OSCC). Methods: In this study, quantitative real-time PCR was conducted for detecting the expression of LINC01614 in OSCC cell lines. The biological functions of LINC01614 were assessed by loss- and gain-of-function experiments conducted both in vivo and in vitro. Cellular proliferation, migration, and invasion were investigated herein, and dual luciferase reporter assays were additionally performed to explore the relationships among LINC01614, miR-138-5p, and Forkhead box C1 (FOXC1). Results: The research presented herein revealed that OSCC cells express high levels of LINC01614. Functional experiments employing cellular and animal models demonstrated that LINC01614 knockdown repressed the malignant phenotypes of OSCC cells, including their growth, invasiveness, and migration. Further investigation revealed that LINC01614 absorbs miR-138-5p miRNA by functioning as a competing endogenous RNA to downregulate the abundance of FOXC1. Conclusions: The findings revealed that LINC01614 contributes to the progression of OSCC by targeting the FOXC1 signaling pathway. The study provides insights into a novel mechanistic process to regulate the development of OSCC, and established a possible target for the therapeutic management of OSCC.
Collapse
Affiliation(s)
- Hongze Che
- School of Dentistry, Beihua University, Jilin 132013, China
| | - Xun Zhang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Luo Cao
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Wenjun Huang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun 130021, China
| | - Qing Lu
- VIP Integrated Department, Stomatological Hospital, Jilin University, Changchun 130021, China
| |
Collapse
|
3
|
Ahmadi M, Morshedzadeh F, Ghaderian SMH, Ghafouri-Fard S. Emerging role of miR-520a in human diseases. Pathol Res Pract 2024; 262:155545. [PMID: 39154603 DOI: 10.1016/j.prp.2024.155545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
hsa-miR-520a is derived from MIR520A located at 19q13.42 and has a significant part in the development of various disorders, including different types of cancers, recurrent pregnancy loss, cerebral ischemia/reperfusion injury, and sciatica. In relation to cancer, numerous studies have presented diverse findings regarding the function of this particular miRNA. To summarize, it has been observed to be down-regulated in pancreatic cancer, glioma, ovarian cancer, cervical cancer, uterine corpus endometrial carcinoma, lung cancer, and acute myeloid leukemia. The purpose of this review is to offer an inclusive overview of the role of has-miR-520a in these disorders, with a specific focus on its target mRNAs in each setting and the deregulated signaling pathways involved. Additionally, we aimed to summarize the implication of miR-520a as a prognostic factor in malignancies. Finally, we performed comprehensive in-silico analyses to uncover the biological roles of this miRNA and introducing innovative concepts for future research endeavors.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firouzeh Morshedzadeh
- Department of Genetics, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Zhao JY, Yao JM, Zhang XZ, Wang KL, Jiang S, Guo SY, Sheng QQ, Liao L, Dong JJ. A New Ferroptosis-Related Long Non-Coding RNA Risk Model Predicts the Prognosis of Patients With Papillary Thyroid Cancer. World J Oncol 2024; 15:648-661. [PMID: 38993258 PMCID: PMC11236373 DOI: 10.14740/wjon1838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/01/2024] [Indexed: 07/13/2024] Open
Abstract
Background Ferroptosis is a novel form of regulated cell death that involves in cancer progression. However, the role of ferroptosis-related long non-coding RNAs (lncRNAs) in papillary thyroid cancer (PTC) remains to be elucidated. The purpose of this paper was to clarify the prognostic value of ferroptosis-related lncRNAs in PTC. Methods The transcriptome data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) database. The correlation between ferroptosis-related genes (FRGs) and lncRNA was determined using Pearson correlation analysis. Multivariate Cox regression model (P < 0.01) was performed to establish a ferroptosis-related lncRNAs risk model. Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curves, risk curve and nomograms were then performed to assess the accuracy and clinical applicability of prognostic models. The correlations between the prognosis model and clinicopathological variables, immune and m6A were analyzed. Finally, in vitro assays were performed to verify the role of LINC00900, LINC01614 and PARAL1 on the proliferation, migration and invasion in TPC-1 and BCPAP cells, as well as the relationship between three lncRNAs and ferroptosis. Results A five-ferroptosis-related lncRNAs (PARAL1, LINC00900, DPH6-DT, LINC01614, LPP-AS2) risk model was constructed. Based on the risk score, samples were divided into the high- and low-risk groups. Patients in the low-risk group had better prognosis than those in high-risk group. Compared to traditional clinicopathological features, risk score was more accurate in predicting prognosis in patients with PTC. Additionally, the difference of immune cell, function and checkpoints was observed between two groups. Moreover, experiments showed that LINC00900 promoted the proliferation, migration and invasion in TPC-1 and BCPAP cells, while LINC01614 and PARAL1 revealed opposite effects, all of which were related to ferroptosis. Conclusions In summary, we identified a five-ferroptosis-related lncRNAs risk model to predict the prognosis of PTC. Furthermore, our study also revealed that LINC00900 functioned as a tumor suppressor lncRNA, LINC01614 and PARAL1 as an oncogenic lncRNA in PTC.
Collapse
Affiliation(s)
- Jun Yu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Ji’nan 250014, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan 250014, China
- These authors contributed equally to this paper
| | - Jin Ming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Ji’nan 250014, China
- These authors contributed equally to this paper
| | - Xin Zhong Zhang
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| | - Kai Li Wang
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| | - Shan Jiang
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| | - Si Yi Guo
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| | - Qi Qi Sheng
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Ji’nan 250014, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan 250014, China
| | - Jian Jun Dong
- Department of Endocrinology and Metabology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Ji’nan 250012, China
| |
Collapse
|
5
|
Mosca N, Alessio N, Di Paola A, Marrapodi MM, Galderisi U, Russo A, Rossi F, Potenza N. Osteosarcoma in a ceRNET perspective. J Biomed Sci 2024; 31:59. [PMID: 38835012 PMCID: PMC11151680 DOI: 10.1186/s12929-024-01049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/24/2024] [Indexed: 06/06/2024] Open
Abstract
Osteosarcoma (OS) is the most prevalent and fatal type of bone tumor. It is characterized by great heterogeneity of genomic aberrations, mutated genes, and cell types contribution, making therapy and patients management particularly challenging. A unifying picture of molecular mechanisms underlying the disease could help to transform those challenges into opportunities.This review deeply explores the occurrence in OS of large-scale RNA regulatory networks, denominated "competing endogenous RNA network" (ceRNET), wherein different RNA biotypes, such as long non-coding RNAs, circular RNAs and mRNAs can functionally interact each other by competitively binding to shared microRNAs. Here, we discuss how the unbalancing of any network component can derail the entire circuit, driving OS onset and progression by impacting on cell proliferation, migration, invasion, tumor growth and metastasis, and even chemotherapeutic resistance, as distilled from many studies. Intriguingly, the aberrant expression of the networks components in OS cells can be triggered also by the surroundings, through cytokines and vesicles, with their bioactive cargo of proteins and non-coding RNAs, highlighting the relevance of tumor microenvironment. A comprehensive picture of RNA regulatory networks underlying OS could pave the way for the development of innovative RNA-targeted and RNA-based therapies and new diagnostic tools, also in the perspective of precision oncology.
Collapse
Affiliation(s)
- Nicola Mosca
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Aniello Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicoletta Potenza
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
6
|
Tian X, Hu D, Wang N, Zhang L, Wang X. LINC01614 is a promising diagnostic and prognostic marker in HNSC linked to the tumor microenvironment and oncogenic function. Front Genet 2024; 15:1337525. [PMID: 38655053 PMCID: PMC11035733 DOI: 10.3389/fgene.2024.1337525] [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: 11/13/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Background Tumor initiation and metastasis influence tumor immune exclusion and immunosuppression. Long non-coding RNA (lncRNA) LINC01614 is associated with the prognosis and metastasis of several cancers. However, the relationship between LINC01614 and cancer immune infiltration and the biofunction of LINC01614 in head and neck squamous cell carcinoma (HNSC) remain unclear. Methods The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) datasets were used to analyze the expression difference and diagnostic value of LINC01614 in normal and tumor tissues. The correlation of pan-cancer prognosis and tumor stage of LINC01614 was analyzed based on the TCGA database. The pan-cancer association of LINC01614 expression with the tumor microenvironment (TME) including immune infiltration, expression of immune-related genes, and genomic instability parameters, was analyzed using the Spearman correlation method. The correlation between LINC01614 and tumor stemness evaluation indicators, RNA methylation-related genes, and drug resistance was also analyzed. The functional analysis of LINC01614 was performed using the clusterProfiler R package. The protein-protein interaction (PPI) network and ceRNA network of LINC01614 co-expressed genes and miRNA were constructed and visualized by STRING and Cytoscape, respectively. Finally, the cell location and influence of LINC01614 on cell proliferation and metastasis of HNSC cell lines were evaluated using FISH, CCK-8, wound-healing assay, and transwell assay. Results LINC01614 was found to be overexpressed in 23 cancers and showed a highly sensitive prediction value in nine cancers (AUC >0.85). LINC01614 dysregulation was associated with tumor stage in 12 cancers and significantly influenced the survival outcomes of 26 cancer types, with only Lymphoid Neoplasm Diffuse Large B-cell Lymphoma (DLBC), uterine corpus endometrial carcinoma (UCEC), and bladder urothelial carcinoma (BLCA) showing a benign influence. LINC01614 was also associated with immune cell infiltration, tumor heterogeneity, cancer stemness, RNA methylation modification, and drug resistance. The potential biological function of LINC01614 was verified in HNSC, and it was found to play important roles in proliferation, immune infiltration, immunotherapy response, and metastasis of HNSC. Conclusion LINC01614 may serve as a cancer diagnosis and prognosis biomarker and an immunotherapy target for specific cancers.
Collapse
Affiliation(s)
- Xiong Tian
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dali Hu
- Department of Plastic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Na Wang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Lele Zhang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xuequan Wang
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
7
|
Wasson MCD, Venkatesh J, Cahill HF, McLean ME, Dean CA, Marcato P. LncRNAs exhibit subtype-specific expression, survival associations, and cancer-promoting effects in breast cancer. Gene 2024; 901:148165. [PMID: 38219875 DOI: 10.1016/j.gene.2024.148165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/25/2023] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in cancer progression, influencing processes such as invasion, metastasis, and drug resistance. Their reported cell type-dependent expression patterns suggest the potential for specialized functions in specific contexts. In breast cancer, lncRNA expression has been associated with different subtypes, highlighting their relevance in disease heterogeneity. However, our understanding of lncRNA function within breast cancer subtypes remains limited, warranting further investigation. We conducted a comprehensive analysis using the TANRIC dataset derived from the TCGA-BRCA cohort, profiling the expression, patient survival associations and immune cell type correlations of 12,727 lncRNAs across subtypes. Our findings revealed subtype-specific associations of lncRNAs with patient survival, tumor infiltrating lymphocytes and other immune cells. Targeting of lncRNAs exhibiting subtype-specific survival associations and expression in a panel of breast cancer cells demonstrated a selective reduction in cell proliferation within their associated subtype, supporting subtype-specific functions of certain lncRNAs. Characterization of HER2 + -specific lncRNA LINC01269 and TNBC-specific lncRNA AL078604.2 showed nuclear localization and altered expression of hundreds of genes enriched in cancer-promoting processes, including apoptosis, cell proliferation and immune cell regulation. This work emphasizes the importance of considering the heterogeneity of breast cancer subtypes and the need for subtype-specific analyses to fully uncover the relevance and potential impact of lncRNAs. Collectively, these findings demonstrate the contribution of lncRNAs to the distinct molecular, prognostic, and cellular composition of breast cancer subtypes.
Collapse
Affiliation(s)
| | | | - Hannah F Cahill
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Meghan E McLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Cheryl A Dean
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada; Nova Scotia Health Authority, Halifax, NS B3H1V8, Canada.
| |
Collapse
|
8
|
Liao X, Wei R, Zhou J, Wu K, Li J. Emerging roles of long non-coding RNAs in osteosarcoma. Front Mol Biosci 2024; 11:1327459. [PMID: 38516191 PMCID: PMC10955361 DOI: 10.3389/fmolb.2024.1327459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Osteosarcoma (OS) is a highly aggressive and lethal malignant bone tumor that primarily afflicts children, adolescents, and young adults. However, the molecular mechanisms underlying OS pathogenesis remain obscure. Mounting evidence implicates dysregulated long non-coding RNAs (lncRNAs) in tumorigenesis and progression. These lncRNAs play a pivotal role in modulating gene expression at diverse epigenetic, transcriptional, and post-transcriptional levels. Uncovering the roles of aberrant lncRNAs would provide new insights into OS pathogenesis and novel tools for its early diagnosis and treatment. In this review, we summarize the significance of lncRNAs in controlling signaling pathways implicated in OS development, including the Wnt/β-catenin, PI3K/AKT/mTOR, NF-κB, Notch, Hippo, and HIF-1α. Moreover, we discuss the multifaceted contributions of lncRNAs to drug resistance in OS, as well as their potential to serve as biomarkers and therapeutic targets. This review aims to encourage further research into lncRNA field and the development of more effective therapeutic strategies for patients with OS.
Collapse
Affiliation(s)
- Xun Liao
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Rong Wei
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junxiu Zhou
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Ke Wu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Mierzejewski B, Pulik Ł, Grabowska I, Sibilska A, Ciemerych MA, Łęgosz P, Brzoska E. Coding and noncoding RNA profile of human heterotopic ossifications - Risk factors and biomarkers. Bone 2023; 176:116883. [PMID: 37597797 DOI: 10.1016/j.bone.2023.116883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Heterotopic ossification (HO) means the formation of bone in muscles and soft tissues, such as ligaments or tendons. HO could have a genetic history or develop after a traumatic event, as a result of muscle injury, fractures, burns, surgery, or neurological disorders. Many lines of evidence suggest that the formation of HO is related to the pathological differentiation of stem or progenitor cells present within soft tissues or mobilized from the bone marrow. The cells responsible for the initiation and progression of HO are generally called HO precursor cells. The exact mechanisms behind the development of HO are not fully understood. However, several factors have been identified as potential contributors. For example, local tissue injury and inflammation disturb soft tissue homeostasis. Inflammatory cells release growth factors and cytokines that promote osteogenic or chondrogenic differentiation of HO precursor cells. The bone morphogenetic protein (BMP) is one of the main factors involved in the development of HO. In this study, next-generation sequencing (NGS) and RT-qPCR were performed to analyze the differences in mRNA, miRNA, and lncRNA expression profiles between muscles, control bone samples, and HO samples coming from patients who underwent total hip replacement (THR). As a result, crucial changes in the level of gene expression between HO and healthy tissues were identified. The bioinformatic analysis allowed to describe the processes most severely impacted, as well as genes which level differed the most significantly between HO and control samples. Our analysis showed that the level of transcripts involved in leukocyte migration, differentiation, and activation, as well as markers of chronic inflammatory diseases, that is, miR-148, increased in HO, as compared to muscle. Furthermore, the levels of miR-195 and miR-143, which are involved in angiogenesis, were up-regulated in HO, as compared to bone. Thus, we suggested that inflammation and angiogenesis play an important role in HO formation. Importantly, we noticed that HO is characterized by a higher level of TLR3 expression, compared to muscle and bone. Thus, we suggest that infection may also be a risk factor in HO development. Furthermore, an increased level of transcripts coding proteins involved in osteogenesis and signaling pathways, such as ALPL, SP7, BGLAP, BMP8A, BMP8B, SMPD3 was noticed in HO, as compared to muscles. Interestingly, miR-99b, miR-146, miR-204, and LINC00320 were up-regulated in HO, comparing to muscles and bone. Therefore, we suggested that these molecules could be important biomarkers of HO formation and a potential target for therapies.
Collapse
Affiliation(s)
- Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Łukasz Pulik
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Aleksandra Sibilska
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland
| | - Maria Anna Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Paweł Łęgosz
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland.
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland.
| |
Collapse
|
10
|
Yang F, Wang M, Shi J, Xu G. IncRNA MALAT1 Regulates the Proliferation, Apoptosis, Migration, and Invasion of Osteosarcoma Cells by Targeting miR-873-5p/ROCK1. Crit Rev Eukaryot Gene Expr 2023; 33:67-79. [PMID: 36734858 DOI: 10.1615/critreveukaryotgeneexpr.2022044747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The malignant bone tumor osteosarcoma (OS) was one of the most aggressive tumors. Despite breakthroughs in treatment options for OS recently, the survival rate of patients with metastasis or reoccurring disease has remained unchanged over the last 25 years, at around 20%. lncRNA expression dysregulation is linked to carcinogenesis, advancement, and metastasis. Additionally, the fundamental mechanism of lncRNAs in regulating OS cell biological activity and progression is still being investigated. The expression of miR-873-5p and MALAT1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR) in OS. The relationship between the expression level of MALAT1 and the survival rate of OS individuals was evaluated by the Kaplan-Meier plotter. The tumor cell's capability of proliferation was determined using the CCK-8. Transwell was used to test the migratory and invasive properties of tumor cells. ROCK1 protein expression was analyzed by western blot, while qRT-PCR was used to detect ROCK1 mRNA expression. Targeted genes of MALAT1 or miR-873-5p were predicted by StarBase2.0. The target association among miR-873-5p and MALAT1 or ROCK1 was confirmed using the luciferase assay. The relationship between ROCK1 and MALAT1 or miR-873-5p expression in OS was investigated using Spearman's correlation analysis. MALAT1 was up-regulated and was linked to a lower survival rate of patients in OS. The malignant behaviors of cells were inhibited by down-regulated MALAT1 in vitro. Dual-luciferase gene experiments confirmed the presence of MALAT1/miR-873-5p/ROCK1 axis. The up-regulated miR-873-5p blocked the promoted effects of MALAT1 on cell behaviors. Over-expressed MALAT1 promoted the malignant behaviors of cells by miR-873-5p/ROCK1 axis in OS.
Collapse
Affiliation(s)
- Fan Yang
- Department of Bone and Soft-Tissue Tumor, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi, China
| | - Mao Wang
- Department of Bone and Soft-Tissue Tumor, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi, China
| | - Junlong Shi
- Department of Bone and Soft-Tissue Tumor, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi, China
| | - Gang Xu
- Department of Bone and Soft-Tissue Tumor, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi, China
| |
Collapse
|
11
|
Xiaotong S, Xiao L, Shiyu L, Zhiguo B, Chunyang F, Jianguo L. LncRNAs could play a vital role in osteosarcoma treatment: Inhibiting osteosarcoma progression and improving chemotherapy resistance. Front Genet 2023; 13:1022155. [PMID: 36726721 PMCID: PMC9885180 DOI: 10.3389/fgene.2022.1022155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Osteosarcoma (OS) is one of the most common primary solid malignant tumors in orthopedics, and its main clinical treatments are surgery and chemotherapy. However, a wide surgical resection range, functional reconstruction of postoperative limbs, and chemotherapy resistance remain as challenges for patients and orthopedists. To address these problems, the discovery of new effective conservative treatments is important. Long non-coding RNAs (lncRNAs) are RNAs longer than 200 nucleotides in length that do not encode proteins. Researchers have recently found that long non-coding RNAs are closely associated with the development of OS, indicating their potentially vital role in new treatment methods for OS. This review presents new findings regarding the association of lncRNAs with OS and summarizes potential clinical applications of OS with lncRNAs, including the downregulation of oncogenic lncRNAs, upregulation of tumor suppressive lncRNAs, and lncRNAs-based treatment to improve chemotherapy resistance. We hope these potential methods will be translated into clinical applications and greatly reduce patient suffering.
Collapse
Affiliation(s)
- Shi Xiaotong
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Li Xiao
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Liao Shiyu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Bi Zhiguo
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Feng Chunyang
- Department of Obstetrics and Gynecology, Renji Hospital of Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Feng Chunyang, ; Liu Jianguo,
| | - Liu Jianguo
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China,*Correspondence: Feng Chunyang, ; Liu Jianguo,
| |
Collapse
|
12
|
Farzaneh M, Najafi S, Anbiyaee O, Azizidoost S, Khoshnam SE. LncRNA MALAT1-related signaling pathways in osteosarcoma. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:21-32. [PMID: 35790599 DOI: 10.1007/s12094-022-02876-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/10/2022] [Indexed: 01/07/2023]
Abstract
Osteosarcoma (OS) is a common and malignant form of bone cancer, which affects children and young adults. OS is identified by osteogenic differentiation and metastasis. However, the exact molecular mechanism of OS development and progression is still unclear. Recently, long non-coding RNAs (lncRNA) have been proven to regulate OS proliferation and drug resistance. LncRNAs are longer than 200 nucleotides that represent the extensive applications in the processing of pre-mRNA and the pathogenesis of human diseases. Metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) is a well-known lncRNA known as a transcriptional and translational regulator. The aberrant expression of MALAT1 has been shown in several human cancers. The high level of MALAT1 is involved in OS cell growth and tumorigenicity by targeting several signaling pathways and miRNAs. Hence, MALAT1 might be a suitable approach for OS diagnosis and treatment. In this review, we will summarize the role of lncRNA MALAT1 in the pathophysiology of OS.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Anbiyaee
- School of Medicine, Cardiovascular Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
13
|
Pei S, Zhang P, Chen H, Zhao S, Dai Y, Yang L, Kang Y, Zheng M, Xia Y, Xie H. Integrating single-cell RNA-seq and bulk RNA-seq to construct prognostic signatures to explore the role of glutamine metabolism in breast cancer. Front Endocrinol (Lausanne) 2023; 14:1135297. [PMID: 36843602 PMCID: PMC9950399 DOI: 10.3389/fendo.2023.1135297] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Although breast cancer (BC) treatment has entered the era of precision therapy, the prognosis is good in the case of comprehensive multimodal treatment such as neoadjuvant, endocrine, and targeted therapy. However, due to its high heterogeneity, some patients still cannot benefit from conventional treatment and have poor survival prognoses. Amino acids and their metabolites affect tumor development, alter the tumor microenvironment, play an increasingly obvious role in immune response and regulation of immune cell function, and are involved in acquired and innate immune regulation; therefore, amino acid metabolism is receiving increasing attention. METHODS Based on public datasets, we carried out a comprehensive transcriptome and single-cell sequencing investigation. Then we used 2.5 Weighted Co-Expression Network Analysis (WGCNA) and Cox to evaluate glutamine metabolism-related genes (GRGs) in BC and constructed a prognostic model for BC patients. Finally, the expression and function of the signature key gene SNX3 were examined by in vitro experiments. RESULTS In this study, we constituted a risk signature to predict overall survival (OS) in BC patients by glutamine-related genes. According to our risk signature, BC patients can obtain a Prognostic Risk Signature (PRS), and the response to immunotherapy can be further stratified according to PRS. Compared with traditional clinicopathological features, PRS demonstrated robust prognostic power and accurate survival prediction. In addition, altered pathways and mutational patterns were analyzed in PRS subgroups. Our study sheds some light on the immune status of BC. In in vitro experiments, the knockdown of SNX3, an essential gene in the signature, resulted in a dramatic reduction in proliferation, invasion, and migration of MDA-MB-231 and MCF-7 cell lines. CONCLUSION We established a brand-new PRS consisting of genes associated with glutamine metabolism. It expands unique ideas for the diagnosis, treatment, and prognosis of BC.
Collapse
Affiliation(s)
- Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huilin Chen
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuhan Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuhan Dai
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Yang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yakun Kang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingjie Zheng
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiqin Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Hui Xie, ; Yiqin Xia,
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Hui Xie, ; Yiqin Xia,
| |
Collapse
|
14
|
Fan L, Zhong Z, Lin Y, Li J. Non-coding RNAs as potential biomarkers in osteosarcoma. Front Genet 2022; 13:1028477. [PMID: 36338952 PMCID: PMC9627036 DOI: 10.3389/fgene.2022.1028477] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma (OS) is a primary solid malignant tumor that occurs most frequently in the metaphysis of long bones. More likely to happen to children and adolescents. OS has high mortality and disability rate. However, the etiology and pathogenesis of OS have not been fully understood till now. Due to the lack of effective biomarkers, OS cannot be precisely detected in the early stage. With the application of next-generation and high-throughput sequencing, more and more abnormally expressed non-coding RNAs(ncRNAs) have been identified in OS. Growing evidences have suggested the ncRNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), have played an important role in the tumorigenesis and progression of OS. Thus, they can be served as novel biomarkers for diagnosis, treatment and prognosis. This review summarized the application of ncRNA as biomarkers in OS in detail, and discussed the limitation and future improvement of the potential biomarkers.
Collapse
Affiliation(s)
- Lijuan Fan
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhenhao Zhong
- Department of Spinal Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yubo Lin
- School of Clinical Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First College for Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- *Correspondence: Jitian Li,
| |
Collapse
|
15
|
Chen LJ, Wu L, Wang W, Zhai LL, Xiang F, Li WB, Tang ZG. Long non‑coding RNA 01614 hyperactivates WNT/β‑catenin signaling to promote pancreatic cancer progression by suppressing GSK‑3β. Int J Oncol 2022; 61:116. [PMID: 35929518 PMCID: PMC9387559 DOI: 10.3892/ijo.2022.5406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/24/2022] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal type of cancer for which effective therapies are limited. Long non-coding RNAs (lncRNAs) represent a critical type of regulator category, mediating the tumorigenesis and development of various tumor types, including PC. However, the expression patterns and functions of numerous lncRNAs in PC remain poorly understood. In the present study, linc01614 was identified as a PC-related lncRNA. linc01614 was notably upregulated in PC tissues and cell lines and was associated with the poor disease-free survival of patients with PC according to the analysis of The Cancer Genome Atlas-derived datasets. Functionally, linc01614 knockdown suppressed PC cell proliferation, migration and invasion in vitro, and inhibited tumor proliferation in vitro and in vivo. Mechanistically, linc01614 overexpression stabilized the level of β-catenin protein to hyperactivate the WNT/β-catenin signaling pathway in PC cells. Further analyses revealed that linc01614 bound to GSK-3β and perturbed the interaction between GSK-3β and AXIN1, thereby preventing the formation of the β-catenin degradation complex and reducing the degradation of β-catenin. In summary, the present findings reveal that linc01614 may function as an oncogene and promote the progression of PC and may thus be considered as a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Long-Jiang Chen
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lun Wu
- Department of Breast and Τhyroid Surgery, Experiment Center of Medicine, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008, P.R. China
| | - Wei Wang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lu-Lu Zhai
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Feng Xiang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Bo Li
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Gang Tang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
16
|
A Newly Established Cuproptosis-Associated Long Non-Coding RNA Signature for Predicting Prognosis and Indicating Immune Microenvironment Features in Soft Tissue Sarcoma. JOURNAL OF ONCOLOGY 2022; 2022:8489387. [PMID: 35847354 PMCID: PMC9279026 DOI: 10.1155/2022/8489387] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/16/2022]
Abstract
Cuproptosis, a new type of programmed cell death, is involved in the development and progression of malignancies. The study of cuproptosis-associated long non-coding RNAs (lncRNAs) in soft tissue sarcomas (STSs) is however limited. There is also uncertainty regarding the prognostic accuracy of cuproptosis-associated lncRNAs in STSs and their relationship to the tumor immune microenvironment. The aim of this study was to determine the prognostic significance of cuprotosis-associated lncRNAs in STSs and their relationship to the tumor immune microenvironment. Transcriptomic and clinical data from patients with STSs were obtained through The Cancer Genome Atlas (TCGA). Overall, 259 patients were randomly allocated to a training group or a testing group. In the training group, a cuproptosis-associated lncRNA signature was constructed, and the signature was verified in the testing group. On the basis of risk scores and clinical features, we later developed a hybrid nomogram. We also performed functional and tumor immune microenvironment analysis based on the cuproptosis-associated lncRNA signature. A signature of 5 cuproptosis-associated lncRNAs was created. Based on this signature, we categorized STS patients into high-risk and low-risk groups. The study showed that patients at high risk had a worse prognosis than those at low risk. A nomogram was then constructed combining clinical characteristics with the risk scores, and it was shown to have credible predictive power. Functional enrichment and tumor immune microenvironmental analyses showed that high-risk STSs tend to be immunologically sensitive tumors. In our study, we found a cuproptosis-associated lncRNAs signature, which serves as an independent prognostic indicator. Cuproptosis-associated lncRNAs may play a role in the tumor immune microenvironment, which might be a therapeutic target for patients with STSs.
Collapse
|
17
|
Liu H, Zong C, Sun J, Li H, Qin G, Wang X, Zhu J, Yang Y, Xue Q, Liu X. Bioinformatics analysis of lncRNAs in the occurrence and development of osteosarcoma. Transl Pediatr 2022; 11:1182-1198. [PMID: 35958002 PMCID: PMC9360822 DOI: 10.21037/tp-22-253] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/04/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a disease with high mortality in children and adolescents, and metastasis is one of its important clinical features. However, the molecular mechanism of OS occurrence is not completely clear. Thus, we screened potential biomarkers of OS and analyze their prognostic value. METHODS The Cancer Genome Atlas (TCGA) datasets were used to analyze the differential lncRNAs in patients with OS of different immune score and the lncRNAs expressed by immune cells. Cox regression was used to develop the prognosis prediction model and specify the prognosis outcomes. Risk-proportional regression model was constructed, and the samples were divided into high and low groups based on the risk scores for the survival analysis. The areas under the receiver operating characteristic (ROC) curve were calculated and the risk-score model was verified. Finally, using 4 gene sets (comprising chemokines, immune checkpoint blockades, immune activity-related genes, and immune cells), and 4 analysis tools (CIBERSORT, TIMER, XCELL and MCP) to evaluated tumor immune infiltration. RESULTS Twenty-nine long non-coding ribonucleic acids (lncRNAs) were obtained from the intersection of the screened lncRNAs. Caspase recruitment domain-containing protein 8-antisense RNA 1 (CARD8-AS1), lncRNA five prime to Xist (FTX), KAT8 regulatory NSL complex unit 1-antisense RNA 1 (KANSL1-AS1), Neuroplastin Intronic Transcript 1 (NPTN-IT1), oligodendrocyte maturation-associated long intervening non-coding RNA (OLMALINC) and RPARP Antisense RNA 1 (RPARP-AS1) were found to be correlated with survival. Univariate and multivariate regression analysis showed risk score [HR (hazard ratio) 3.5, P value 0.0043; HR 3.7, P value 0.0033] and metastasis (HR 4.7, P value 6.60E-05; HR 4.8, P value 8.36E-05) were the key factors of patients with OS. The areas under curves (AUCs) of the 1-, 3-, and 5-year ROC curves of the prognostic model were 0.715, 0.729, and 0.771. The low-risk patients tended to have a high abundance of immune cells. CONCLUSIONS This study showed that a risk score based on 6 lncRNAs has potential value in the prognosis of OS, and patients with low-risk scores have high immune cell infiltration and good prognosis. This study may enrich understandings of underlying mechanisms related to the occurrence and development of OS.
Collapse
Affiliation(s)
- Hua Liu
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Haian, China
| | - Chenyu Zong
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiacheng Sun
- Xinglin College, Nantong University, Nantong, China
| | - Haiyang Li
- Department of Oncology, Binhai County People's Hospital, Yancheng, China
| | - Guangzhen Qin
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Haian, China
| | - Xiaojian Wang
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Haian, China
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Yang Yang
- Department of Trauma Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Qiang Xue
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xianchen Liu
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
18
|
Hou Y, Zhou M, Li Y, Tian T, Sun X, Chen M, Xu W, Lu M. Risk SNP-mediated LINC01614 upregulation drives head and neck squamous cell carcinoma progression via PI3K/AKT signaling pathway. Mol Carcinog 2022; 61:797-811. [PMID: 35687049 DOI: 10.1002/mc.23422] [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: 02/04/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/11/2022]
Abstract
As potential biomarkers and therapeutic targets, long noncoding RNAs (lncRNAs) are involved in the tumorigenesis of various tumors. Genetic variation in long noncoding regions can lead to lncRNA dysfunction and even cancer. Nevertheless, studies on the association between lncRNA-associated single-nucleotide polymorphisms (SNPs) and the risk of head and neck squamous cell carcinoma (HNSCC) remain inadequate. Here, we aimed to explore the association between SNPs in LINC01614 and HNSCC risk, and the potential role of LINC01614 in tumorigenesis. In this study, we found that rs16854802 A > G (odds ratio [OR] = 1.42, 95% confidence interval [CI]: 1.22-1.77, p < 0.001) and rs3113503 G > C (OR = 1.38, 95% CI: 1.15-1.64, p < 0.001) in LINC01614 increased the risk of HNSCC in the Chinese population. Functional bioinformatic analysis and luciferase reporter assay revealed that rs3113503 G > C variant disrupted the binding of miRNA-616-3p to LINC01614, which resulted in the increased expression of LINC01614. Further analysis of the TCGA database demonstrated that the upregulated LINC01614 in HNSCC cancer tissues was associated with poor prognostic in HNSCC patients. In vitro experiments showed that knockdown of LINC01614 inhibited the proliferation, invasion, and migration ability of HNSCC cells. Mechanistically, allele C of rs3113503 in LINC01614 was more effective than allele G in activating the PI3K/AKT signaling pathway. Moreover, the reduced expression of LINC01614 also inhibited the activation of the PI3K/AKT signaling pathway. In summary, our findings revealed that the risk SNP rs3113503 G > C in LINC01614 altered the binding to miR-616-3p, which led to increased LINC01614 expression and promoted HNSCC progression by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yaxuan Hou
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Zhou
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuncheng Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Tian
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xun Sun
- Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Guidance Center for Social Psychological Service, Wuhan Mental Health Center, Huazhong University of Science and Technology, Wuhan, China
| | - Wenmao Xu
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Public Health, Wuhan No. 1 Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Meixia Lu
- Department of Epidemiology and Biostatistics, and The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Wang G, Sun L, Wang S, Guo J, Xiao R, Li W, Qi W, Qiu W. Ferroptosis‑related long non‑coding RNAs and the roles of LASTR in stomach adenocarcinoma. Mol Med Rep 2022; 25:118. [PMID: 35137922 PMCID: PMC8855154 DOI: 10.3892/mmr.2022.12634] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
Ferroptosis is a form of programmed cell death that participates in diverse physiological processes. Increasing evidence suggests that long noncoding RNAs (lncRNAs) regulate ferroptosis in tumors, including stomach adenocarcinoma (STAD). In the present study, RNA-sequencing data from The Cancer Genome Atlas database and ferroptosis-related markers from the FerrDb data resource were analyzed to select differentially expressed lncRNAs. Univariate and multivariate Cox regression analyses were performed on these differentially expressed lncRNAs to screen 12 lncRNAs linked with overall survival (OS) and 13 associated with progression-free survival (PFS). Subsequently, two signatures for predicting OS and PFS were established based on these lncRNAs. Kaplan-Meier analyses indicated that the high-risk group of patients with STAD had relatively poor prognosis. The areas under the receiver operating characteristic curves of the two signatures indicated their excellent efficacy in predicting STAD prognosis. In addition, the effect of the lncRNA LASTR on proliferation and migration in gastric cancer was confirmed and the relationship between LASTR and ferroptosis was initially explored through experiments. These results provide potential novel targets for tumor treatment and promote personalized medicine.
Collapse
Affiliation(s)
- Gongjun Wang
- Department of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Libin Sun
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shasha Wang
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jing Guo
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Ruoxi Xiao
- Department of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wenqian Li
- Department of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Weiwei Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Wensheng Qiu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
20
|
Zou J, Gu Y, Zhu Q, Li X, Qin L. Identifying Glycolysis-related LncRNAs for predicting prognosis in breast cancer patients. Cancer Biomark 2022; 34:393-401. [PMID: 35068448 PMCID: PMC9198763 DOI: 10.3233/cbm-210446] [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] [Indexed: 01/01/2023]
Abstract
PURPOSE: Functions associated with glycolysis could serve as targets or biomarkers for therapy cancer. Our purpose was to establish a prognostic model that could evaluate the importance of Glycolysis-related lncRNAs in breast cancer. METHODS: Gene expressions were evaluated for breast cancer through The Cancer Genome Atlas (TCGA) database, and we calculated Pearson correlations to discover potential related lncRNAs. Differentially expressed genes were identified via criteria of FDR < 0.05 and |FC|> 2. Total samples were separated into training and validating sets randomly. Univariate Cox regression identified 14 prognostic lncRNAs in training set. A prognostic model was constructed to evaluate the accuracy in predicting prognosis. The univariate and multivariate Cox analysis were performed to verify whether lncRNA signature could be an independent prognostic factor The signature was validated in validating set. Immune infiltration levels were assessed. RESULTS: Eighty-nine differentially expressed lncRNAs were identified from 420 Glycolysis-related lncRNAs. 14 lncRNAs were correlated with prognosis in training set and were selected to establish the prognostic model. Low risk group had better prognosis in both training (p= 9.025 e -10) and validating (p= 4.272 e -3) sets. The univariate and multivariate Cox analysis revealed that risk score of glycolysis-related lncRNAs (P< 0.001) was an independent prognostic factor in both training and validating sets. The neutrophils (p= 4.214 e -13, r=-0.223), CD4+ T cells (p= 1.833 e -20, r=-0.283), CD8+ T cells (p= 7.641 e -12, r=-0.211), B cells (p= 2.502 e -10, r=-0.195) and dendritic cells (p= 5.14 e -18, r=-0.265) were negatively correlated with risk score of prognostic model. The Macrophage (p= 0.016, r= 0.0755) was positively correlated with the risk score. CONCLUSION: Our study indicated that glycolysis-related lncRNAs had a significant role to facilitate the individualized survival prediction in breast cancer patients, which would be a potential therapeutic target.
Collapse
Affiliation(s)
- Jiayue Zou
- Department of Hepatobiliary Surgery, General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanlin Gu
- Department of Thyroid and Breast Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Zhu
- Department of Thyroid and Breast Surgery, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Xiaohua Li
- Department of Thyroid and Breast Surgery, Wuzhong People’s Hospital of Suzhou City, Suzhou, Jiangsu, China
| | - Lei Qin
- Department of Hepatobiliary Surgery, General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
21
|
Sheng W, Zhou W, Cao Y, Zhong Y. Revealing the Role of lncRNA CCDC144NL-AS1 and LINC01614 in Gastric Cancer via Integrative Bioinformatics Analysis and Experimental Validation. Front Oncol 2022; 11:769563. [PMID: 35083139 PMCID: PMC8784853 DOI: 10.3389/fonc.2021.769563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are key regulators in the pathophysiology of gastric cancer, and lncRNAs have been regarded as potential biomarkers and therapeutic targets for gastric cancer. The present study performed the WGCNA analysis of the GSE70880 dataset and aimed to identify novel lncRNAs associated with gastric cancer progression. Based on the WGCNA, the lncRNAs and mRNA co-expression network were constructed. A total of four modules were identified and the eigengenes in different modules were involved in various key signaling pathways. Furthermore, the co-expression networks were constructed between the lncRNAs and mRNA; this leads to the identification of 6 modules, which participated in various cellular pathways. The survival analysis showed that high expression of CCDC144NL antisense RNA 1 (CCDC144NL-AS1) and LINC01614 was positively correlated with the poor prognosis of patients with gastric cancer. The in vitro validation results showed that CCDC144NL-AS1 and LINC01614 were both up-regulated in the gastric cancer cells. Silence of CCDC144NL-AS1 and LINC01614 both significantly suppressed the cell proliferation and migration of gastric cancer cells, and also promoted the chemosensitivity of gastric cancer cells to 5-fluorouracil. Collectively, our results suggested that the newly identified two lncRNAs (CCDC144NL-AS1 and LINC01614) may act as oncogenes in gastric cancer.
Collapse
Affiliation(s)
- Weiwei Sheng
- Physical Examination Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Weihong Zhou
- Physical Examination Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yundi Cao
- Department of Oncology, Affiliated Taikang Xianlin Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuejiao Zhong
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Wu H, Zhou J, Chen S, Zhu L, Jiang M, Liu A. Survival-Related lncRNA Landscape Analysis Identifies LINC01614 as an Oncogenic lncRNA in Gastric Cancer. Front Genet 2021; 12:698947. [PMID: 34691143 PMCID: PMC8526963 DOI: 10.3389/fgene.2021.698947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) reportedly play important roles in biomarker and tumorigenesis of gastric cancer (GC). This study aimed to determine the potential application of prognostic lncRNA signature and identified the role of LINC01614 in carcinogenesis in GC. Material and Methods: Data accessed from the Cancer Genome Atlas database was used to construct a lncRNA signature. Joint effect analysis of the signature and clinical parameters was performed to verify the clinical value of the signature. Co-expression analysis was conducted for prognostic lncRNAs and protein-coding genes. Moreover, the relative expression of LINC01614 was validated in GC tissues and cell lines. In vitro and in vivo experiments were conducted to analyze the biological functions of the newly identified gene in GC cells. Results: A seven-lncRNA (LINC01614, LINC01537, LINC01210, OVAAL, LINC01446, CYMP-AS1, and SCAT8) signature was identified as a promising prognostic signature in GC. Results indicated that the seven-lncRNA was involved in tumorigenesis and progression pathways. LINC01614 expression was identified and found to be upregulated in GC tissues and cells. The study findings revealed that LINC01614 promoted cell proliferation, migration, invasion, and epithelial-mesenchymal transition. Knockdown of LINC01614 arrested cell cycle distribution at the G2/M phase. Further, LINC01614 also promoted tumor growth in vivo. Conclusion: We developed an independent seven-lncRNA biomarker for prognostic prediction and identified LINC01614 as an oncogenic lncRNA in GC.
Collapse
Affiliation(s)
- Huijie Wu
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jingyuan Zhou
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Songda Chen
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lingyu Zhu
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mengjie Jiang
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Aiqun Liu
- Department of Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|