1
|
Chen Y, Zhang J, Xu H. Recent developments in cuproptosis of glioblastoma. Pathol Res Pract 2025; 269:155939. [PMID: 40164043 DOI: 10.1016/j.prp.2025.155939] [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: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Glioblastoma (GBM) is the most malignant tumor within the central nervous system, attributed to its high-grade malignancy, propensity for recurrence, refractoriness to conventional therapeutic modalities, and the suboptimal efficacy of current targeted therapies. Hence, there is an urgent need to identify more efficacious molecular targets for the therapeutic intervention of GBM. The regulated cell death (RCD) has specific signaling factors and signaling pathways. Hence, targeting RCD is considered to be one of the effective targeted therapies for GBM. At present, cuproptosis is a novel form of RCD, characterized by a distinct molecular mechanism that differentiates it from apoptosis, pyroptosis, necroptosis, and ferroptosis. It is characterized by its principal mechanisms, which include copper dependency, the accumulation of acylated proteins, and the reduction of Fe-S cluster-containing proteins. These processes collectively induce proteotoxic stress, culminating in cell death. In previous studies, copper-ionized formulations have demonstrated cytotoxic effects on gliomas. Thus, the key factors of cuproptosis may be able to serve as a new target for GBM treatment. This review delves into several pivotal aspects, including the discovery of cuproptosis, the impact of copper homeostasis on tumorigenesis, the role of cuproptosis in GBM, and its potential as a therapeutic target in molecular targeted therapy for GBM. Hence, this article could reveal novel strategies for GBM treatment.
Collapse
Affiliation(s)
- Yajia Chen
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Jingxian Zhang
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Hongwu Xu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Wandao Rd, 78, Wanjiang Subdistrict, Dongguan, Guangdong Province 523059, China; Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China.
| |
Collapse
|
2
|
SINGH RAHULKUMAR, MANDAL SUROJIT, MOHANTA ADRIJA, YADAV RITU, KUMAR RAJIVRANJAN, KHATKAR RINKU, UTTAM VIVEK, SHARMA UTTAM, RANA MANJITKAUR, JAIN MANJU, TULI HARDEEPSINGH, JAIN AKLANK. The regulatory role of ZFAS1/miRNAs/mRNAs axis in cancer: a systematic review. Oncol Res 2025; 33:591-604. [PMID: 40109869 PMCID: PMC11915068 DOI: 10.32604/or.2024.050548] [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: 02/09/2024] [Accepted: 05/16/2024] [Indexed: 03/22/2025] Open
Abstract
Objectives Recently, we and others have demonstrated the involvement of Zinc Finger Antisense 1 (ZFAS1) in cancer development. However, the intricate interplay of ZFAS1 with miRNAs and mRNAs remains to be fully understood. Materials and methods We followed PRISMA guidelines to retrieve and assess the available literature on the topic "ZFAS1/miRNA/mRNA axis" and "Cancer" from databases such as PubMed, Google Scholar, and ScienceDirect. We also used bioinformatic webtools for analyzing the potential miRNA targets of ZFAS1 and its role in survival of cancer patients along with their role in various biological functions and pathways. Results Our literature search and bioinformatic analysis reveals that ZFAS1 serves as a sponge for numerous miRNAs. Among the various targeted miRNAs, miR-150-5p stands out as significantly correlated with ZFAS1 across multiple databases (p-value = 3.27e-16, R-value = -0.346). Additionally, our Kaplan-Meier survival analysis indicates a noteworthy association between ZFAS1 expression levels and overall poor prognosis and survival rates in ovarian, sarcoma, and pancreatic cancers. We also underscore the involvement of various signaling pathways, including Signal Transducer and Activator of Transcription 3 (STAT3), Spindle and Kinetochore-associated Protein 1 (SKA1), Lysophosphatidic acid receptor 1 (LPAR1), and Wnt β-catenin, in cancer development through the ZFAS1/miRNAs/mRNAs axis. Furthermore, we identify ZFAS1's pivotal roles in diverse molecular processes, such as RNA binding and ribonucleoprotein formation. Conclusion In conclusion, this review comprehensively summarizes the latest advancements in understanding the regulatory relationships among ZFAS1, miRNAs, and mRNAs, emphasizing their collective role in cancer development to propose innovative avenues for cancer treatment. We believe that the intricate relationship among the ZFAS1-miRNA-mRNA axis may yield potential therapeutic targets for effective cancer management.
Collapse
Affiliation(s)
- RAHUL KUMAR SINGH
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - SUROJIT MANDAL
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - ADRIJA MOHANTA
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - RITU YADAV
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - RAJIV RANJAN KUMAR
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - RINKU KHATKAR
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - VIVEK UTTAM
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - UTTAM SHARMA
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - MANJIT KAUR RANA
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences, Bathinda, 151001, India
| | - MANJU JAIN
- Department of Biochemistry, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - HARDEEP SINGH TULI
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - AKLANK JAIN
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| |
Collapse
|
3
|
Goleij P, Pourali G, Raisi A, Ravaei F, Golestan S, Abed A, Razavi ZS, Zarepour F, Taghavi SP, Ahmadi Asouri S, Rafiei M, Mousavi SM, Hamblin MR, Talei S, Sheida A, Mirzaei H. Role of Non-coding RNAs in the Response of Glioblastoma to Temozolomide. Mol Neurobiol 2025; 62:1726-1755. [PMID: 39023794 DOI: 10.1007/s12035-024-04316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/16/2024] [Indexed: 07/20/2024]
Abstract
Chemotherapy and radiotherapy are widely used in clinical practice across the globe as cancer treatments. Intrinsic or acquired chemoresistance poses a significant problem for medical practitioners and researchers, causing tumor recurrence and metastasis. The most dangerous kind of malignant brain tumor is called glioblastoma multiforme (GBM) that often recurs following surgery. The most often used medication for treating GBM is temozolomide chemotherapy; however, most patients eventually become resistant. Researchers are studying preclinical models that accurately reflect human disease and can be used to speed up drug development to overcome chemoresistance in GBM. Non-coding RNAs (ncRNAs) have been shown to be substantial in regulating tumor development and facilitating treatment resistance in several cancers, such as GBM. In this work, we mentioned the mechanisms of how different ncRNAs (microRNAs, long non-coding RNAs, circular RNAs) can regulate temozolomide chemosensitivity in GBM. We also address the role of these ncRNAs encapsulated inside secreted exosomes.
Collapse
Affiliation(s)
- Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahin Golestan
- Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atena Abed
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Sadat Razavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Sahand Talei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Mirzaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Albulescu A, Botezatu A, Fudulu A, Hotnog CM, Bostan M, Mihăilă M, Iancu IV, Plesa A, Brasoveanu L. Combined Effect of Conventional Chemotherapy with Epigenetic Modulators on Glioblastoma. Genes (Basel) 2025; 16:138. [PMID: 40004468 PMCID: PMC11855767 DOI: 10.3390/genes16020138] [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: 12/31/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Glioblastoma is the most common malignant primary brain tumor, characterized by necrosis, uncontrolled proliferation, infiltration, angiogenesis, apoptosis resistance, and genomic instability. Epigenetic modifiers hold promise as adjuvant therapies for gliomas, with synergistic combinations being explored to enhance efficacy and reduce toxicity. This study aimed to evaluate the effects of single or combined treatments with various anticancer drugs (Carboplatin, Paclitaxel, Avastin), natural compounds (Quercetin), and epigenetic modulators (suberoylanilide hydroxamic acid and 5-Azacytidine) on the expression of some long noncoding RNAs and methylation drivers or some functional features in the U87-MG cell line. METHODS Treated and untreated U87-MG cells were used for the evaluation of drug-induced cytotoxicity, apoptotic events, and distribution in cell cycle phases, detection of cytokine release, and assessment of gene expression and global methylation. RESULTS Cytotoxicity assays led to the selection of drug concentrations to be used in further experiments. Expression analysis revealed distinct downregulation of nearly all investigated genes and long noncoding RNAs following treatments. All treatments resulted in a higher percentage of global methylation compared to untreated controls. All treatments effectively increased levels of apoptosis, while the epigenetic modulators exhibited a lower proliferation profile, with combined treatments showing elevated values of cell lysis. CONCLUSIONS The results indicate a link between Carboplatin and Avastin treatments and DNA methylation mechanisms involving EZH2, DNMT3A, and DNMT3B, with Avastin's direct impact on these enzymes warranting further study. This research underscores the promise of platinum-based therapies combined with epigenetic drugs to reactivate silenced tumor suppressor genes and optimize methylation profiles.
Collapse
Affiliation(s)
- Adrian Albulescu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
- Pharmacology Department, National Institute for Chemical Pharmaceutical Research and Development, 031299 Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Alina Fudulu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Camelia Mia Hotnog
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Mirela Mihăilă
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Iulia Virginia Iancu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Adriana Plesa
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Lorelei Brasoveanu
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| |
Collapse
|
5
|
Liu X, Ma Z, Zhang X, Li S, An J, Luo Z. Research Progress of Long Non-coding RNA-ZFAS1 in Malignant Tumors. Cell Biochem Biophys 2024; 82:3145-3156. [PMID: 39060915 DOI: 10.1007/s12013-024-01441-3] [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] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Long non-coding RNAs (lncRNAs), although incapable of encoding proteins, play crucial roles in multiple layers of gene expression regulation, epigenetic modifications, and post-transcriptional regulation. Zinc finger antisense 1 (ZFAS1), a lncRNA located in the 20q13 region of the human genome, exhibits dual functions as an oncogene or tumor suppressor in various human malignancies. ZFAS1 plays a crucial role in cancer progression, metastasis, invasion, apoptosis, cell cycle regulation, and drug resistance through complex molecular mechanisms. Additionally, ZFAS1 has a long half-life of over 16 h, demonstrating exceptional stability, and making it a potential biomarker. This review integrates recent studies on the role and molecular mechanisms of ZFAS1 in malignancies and summarizes its clinical significance. By summarizing the role of ZFAS1 in cancer, we aim to highlight its potential as an anti-cancer biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Zhong Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Xianxu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Shicheng Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Zhiqiang Luo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
6
|
Hao S, Gao M, Li Q, Shu L, Wang P, Hao G. Machine learning predicts cuproptosis-related lncRNAs and survival in glioma patients. Sci Rep 2024; 14:22323. [PMID: 39333603 PMCID: PMC11437180 DOI: 10.1038/s41598-024-72664-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
Gliomas are the most common tumor in the central nervous system in adults, with glioblastoma (GBM) representing the most malignant form, while low-grade glioma (LGG) is a less severe. The prognosis for glioma remains poor even after various treatments, such as chemotherapy and immunotherapy. Cuproptosis is a newly defined form of programmed cell death, distinct from ferroptosis and apoptosis, primarily caused by the accumulation of the copper within cells. In this study, we compared the difference between the expression of cuproptosis-related genes in GBM and LGG, respectively, and conducted further analysis on the enrichment pathways of the exclusive expressed cuproptosis-related mRNAs in GBM and LGG. We established two prediction models for survival status using xgboost and random forest algorithms and applied the ROSE algorithm to balance the dataset to improve model performance.
Collapse
Affiliation(s)
- Shaocai Hao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Neurosurgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
| | - Maoxiang Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qin Li
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Lilu Shu
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Peter Wang
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China.
| | - Guangshan Hao
- Department of Neurosurgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
7
|
Hou X, Xu Q, Liu R. CREB3 facilitates Donafenib resistance in hepatocellular carcinoma cells via the LSD1/CoREST/p65 axis by transcriptionally activating long noncoding RNA ZFAS1. Transl Oncol 2024; 44:101684. [PMID: 38641372 PMCID: PMC11391036 DOI: 10.1016/j.tranon.2023.101684] [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: 02/09/2023] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE Drug resistance greatly limits the therapeutic effect of a drug. This study aimed to explore the role of long noncoding RNA ZFAS1 in Donafenib resistance of hepatocellular carcinoma (HCC) cells. METHODS The expression of CREB3, ZFAS1, and p65 in HCC cell lines was measured by RT-qPCR and western blotting. After transfection with sh-ZFAS1, sh-CREB3, or sh-CREB3 + oe-p65 in Donafenib-resistent (DR) HCC cell lines, the transfection efficiency was evaluated by RT-qPCR and western blotting. The proliferation and IC50 to Donafenib of HCC cell lines was examined by MTT assay. Cell proliferation and apoptosis were examined by colony formation and flow cytometry assays. Then, the correlation amongst CREB3, ZFAS1, LSD1/CoREST, and p65 was analysed by ChIP, dual-luciferase reporter gene, and RIP assays. RESULTS ZFAS1, CREB3, and p65 were upregulated in HepG2-DR and Huh7-DR cells. Silencing of ZFAS1 or CREB3 enhanced the sensitivity of HCC cells to Donafenib, inhibited cell proliferation and IC50, and increased cell apoptosis, which were reversed by p65 overexpression. Mechanistically, CREB3 bound to ZFAS1 promoter to augment ZFAS1 transcriptional expression, and ZFAS1 recruited LSD1/CoREST to the p65 promoter region to decrease H3K4 methylation and elevate p65 transcriptional expression. CONCLUSION CREB3 overexpression contributed to Donafenib resistance in HCC cells by activating the ZFAS1/p65 axis.
Collapse
Affiliation(s)
- Xunbo Hou
- Department of Interventional, Harbin Medical University Cancer Hospital, No. 150, Haping Rd, Nangang District, Harbin, Heilongjiang 150081, PR China
| | - Qiannan Xu
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Ruibao Liu
- Department of Interventional, Harbin Medical University Cancer Hospital, No. 150, Haping Rd, Nangang District, Harbin, Heilongjiang 150081, PR China.
| |
Collapse
|
8
|
Xie F, Qu J, Lin D, Feng K, Tan M, Liao H, Zeng L, Xiong Q, Huang J, Chen W. Reduced Proteolipid Protein 2 promotes endoplasmic reticulum stress-related apoptosis and increases drug sensitivity in acute myeloid leukemia. Mol Biol Rep 2023; 51:10. [PMID: 38085372 DOI: 10.1007/s11033-023-08994-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND The Proteolipid Protein 2 (PLP2), a protein in the Endoplasmic Reticulum (ER) membrane, has been reported to be highly expressed in various tumors. Previous studies have demonstrated that the reduced PLP2 can induce apoptosis and autophagy through ER stress-related pathways, leading to a decreased proliferation and aggressiveness. However, there is no research literature on the role of PLP2 in Acute Myeloid Leukemia (AML). METHODS PLP2 expression, clinical data, genetic mutations, and karyotype changes from GEO, TCGA, and timer2.0 databases were analyzed through the R packages. The possible functions and pathways of cells were explored through GO, KEGG, and GSEA enrichment analysis using the clusterProfiler R package. Immuno-infiltration analysis was conducted using the Cibersort algorithm and the Xcell R package. RT-PCR and western blot techniques were employed to identify the PLP2 expression, examine the knockdown effects in THP-1 cells, and assess the expression of genes associated with endoplasmic reticulum stress and apoptosis. Flow cytometry was utilized to determine the apoptosis and survival rates of different groups. RESULTS PLP2 expression was observed in different subsets of AML and other cancers. Enrichment analyses revealed that PLP2 was involved in various tumor-related biological processes, primarily apoptosis and lysosomal functions. Additionally, PLP2 expression showed a strong association with immune cell infiltration, particularly monocytes. In vitro, the knockdown of PLP2 enhanced endoplasmic reticulum stress-related apoptosis and increased drug sensitivity in THP-1 cells. CONCLUSIONS PLP2 could be a novel therapeutic target in AML, in addition, PLP2 is a potential endoplasmic reticulum stress regulatory gene in AML.
Collapse
Affiliation(s)
- Fahui Xie
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jia Qu
- Department of Hematology, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dainan Lin
- Department of Hematology, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Kexin Feng
- Department of Hematology, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingzhu Tan
- Department of Hematology, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haixiu Liao
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Longhui Zeng
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingquan Xiong
- Department of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Jun Huang
- Department of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.
| | - Weiwen Chen
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Rezaee A, Tehrany PM, Tirabadi FJ, Sanadgol N, Karimi AS, Ajdari A, Eydivandi S, Etemad S, Rajabi R, Rahmanian P, Khorrami R, Nabavi N, Aref AR, Fan X, Zou R, Rashidi M, Zandieh MA, Hushmandi K. Epigenetic regulation of temozolomide resistance in human cancers with an emphasis on brain tumors: Function of non-coding RNAs. Biomed Pharmacother 2023; 165:115187. [PMID: 37499452 DOI: 10.1016/j.biopha.2023.115187] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
Brain tumors, which are highly malignant, pose a significant threat to health and often result in substantial rates of mortality and morbidity worldwide. The brain cancer therapy has been challenging due to obstacles such as the BBB, which hinders effective delivery of therapeutic agents. Additionally, the emergence of drug resistance further complicates the management of brain tumors. TMZ is utilized in brain cancer removal, but resistance is a drawback. ncRNAs are implicated in various diseases, and their involvement in the cancer is particularly noteworthy. The focus of the current manuscript is to explore the involvement of ncRNAs in controlling drug resistance, specifically in the context of resistance to the chemotherapy drug TMZ. The review emphasizes the function of ncRNAs, particularly miRNAs, in modulating the growth and invasion of brain tumors, which significantly influences their response to TMZ treatment. Through their interactions with various molecular pathways, miRNAs are modulators of TMZ response. Similarly, lncRNAs also associate with molecular pathways and miRNAs, affecting the efficacy of TMZ chemotherapy. Given their functional properties, lncRNAs can either induce or suppress TMZ resistance in brain tumors. Furthermore, circRNAs, which are cancer controllers, regulate miRNAs by acting as sponges, thereby impacting the response to TMZ chemotherapy. The review explores the correlation between ncRNAs and TMZ chemotherapy, shedding light on the underlying molecular pathways involved in this process.
Collapse
Affiliation(s)
- Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Farimah Jafari Tirabadi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Negin Sanadgol
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Asal Sadat Karimi
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Atra Ajdari
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Eydivandi
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sara Etemad
- Faculty of Veterinary Medicine, Islamic Azad University, Garmsar Branch, Semnan, Iran.
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada.
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA.
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
10
|
Recent progress in nanocarrier-based drug delivery systems for antitumour metastasis. Eur J Med Chem 2023; 252:115259. [PMID: 36934485 DOI: 10.1016/j.ejmech.2023.115259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Tumour metastasis is one of the major factors leading to poor prognosis as well as lower survival among cancer patients. A number of studies investigating the inhibition of tumour metastasis have been conducted. It is difficult to achieve satisfactory results with surgery alone for distant metastatic tumours, and chemotherapy can boost the healing rate and prognosis of patients. However, the poor therapeutic efficacy of chemotherapy drugs due to their low solubility, lack of tumour targeting, instability in vivo, high toxicity and multidrug resistance hinder their application. Immunotherapy is beneficial to the treatment of metastatic cancers, but it also has disadvantages such as adverse reactions and acquired resistance. Fortunately, delivery of chemotherapeutic drugs with nanocarriers can reduce systemic reactions caused by chemotherapeutic agents and inhibit metastasis. This review discusses the underlying mechanisms of metastasis, therapeutic approaches for antitumour metastasis, the advantages of nanodrug delivery systems and their application in reducing metastasis.
Collapse
|
11
|
Zhu P, Pei Y, Yu J, Ding W, Yang Y, Liu F, Liu L, Huang J, Yuan S, Wang Z, Gu F, Pan Z, Chen J, Qiu J, Liu H. High-throughput sequencing approach for the identification of lncRNA biomarkers in hepatocellular carcinoma and revealing the effect of ZFAS1/miR-150-5p on hepatocellular carcinoma progression. PeerJ 2023; 11:e14891. [PMID: 36855431 PMCID: PMC9968462 DOI: 10.7717/peerj.14891] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/23/2023] [Indexed: 02/25/2023] Open
Abstract
Aims To screen abnormal lncRNAs and diagnostic biomarkers in the progression of hepatocellular carcinoma through high-throughput sequencing and explore the underlying mechanisms of abnormal lncRNAs in the progression of hepatocellular carcinoma. Methods The transcriptome sequencing was used to analyze the RNA expression profile and identify differentially expressed RNAs. Hub lncRNAs were screened by combining (WGCNA, ceRNA regulatory network, PPI, GO and KEGG analyses, Kaplan-Meier curve analysis, Cox analysis, risk model construction and qPCR). Thereafter, the correlation between the expression of hub lncRNAs and tumor clinicopathological parameters was analyzed, and the hub lncRNAs were analyzed by GSEA. Finally, the effects of hub RNAs on the proliferation, migration and invasion of HepG2 cells were investigated in vitro. Results Compared with the control group, a total of 610 lncRNAs, 2,593 mRNAs and 26 miRNAs were screened in patients with hepatocellular carcinoma. Through miRNA target prediction and WGCNA, a ceRNA was constructed, comprising 324 nodes and 621 edges. Enrichment analysis showed that mRNAs in ceRNA were involved mainly in cancer development progression. Then, the ZFAS1/miR-150-5p interaction pair was screened out by Kaplan Meier curve analysis, Cox analysis and qPCR analysis. Its expression was related to tumor stage, TNM stage and patient age. ROC curve analysis showed that it has a good predictive value for the risk of hepatocellular carcinoma. GSEA showed that ZFAS1 was also enriched in the regulation of immune response, cell differentiation and proliferation. Loss-of-function experiments revealed that ZFAS1 inhibition could remarkably suppress HepG2 cell proliferation, migration and invasion in vitro. Bioinformatic analysis and luciferase reporter assays revealed that ZFAS1 directly interacted with miR-150-5p. Rescue experiments showed that a miR-150-5p inhibitor reversed the cell proliferation, migration and invasion functions of ZFAS1 knockdown in vitro. Conclusion ZFAS1 is associated with the malignant status and prognosis of patients with hepatocellular carcinoma, and the ZFAS1/miR-150-5p axis is involved in hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yongyan Pei
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, Guangdong, China
| | - Jian Yu
- Department of General Surgery, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wenbin Ding
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yun Yang
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Fuchen Liu
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lei Liu
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jian Huang
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shengxian Yuan
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zongyan Wang
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Fangming Gu
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zeya Pan
- Department of Hepatic Surgery (III), The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jinzhong Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jinrong Qiu
- Department of Biotherapy, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Huiying Liu
- Department of Biotherapy, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
Zhao Y, Song X, Song X, Xie L. Identification of Diagnostic Exosomal LncRNA-miRNA-mRNA Biomarkers in Colorectal Cancer Based on the ceRNA Network. Pathol Oncol Res 2022; 28:1610493. [PMID: 36185995 PMCID: PMC9522904 DOI: 10.3389/pore.2022.1610493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/08/2022] [Indexed: 12/24/2022]
Abstract
Background: Colorectal cancer (CRC) is currently the fourth most common cancer worldwide. The roles of exosomal competing endogenous RNAs (ceRNAs) in CRC remain unclear. In this study, we constructed an exosomal ceRNA network to identify the core ceRNAs and investigate the diagnostic biomarkers in CRC.Methods and Patients: Serum exosomes were isolated from four CRC patients and two healthy donors by ultracentrifugation, and then subjected to RNA isolation, sequencing and microarray. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses were performed to identify functional enrichment implications of differentially expressed exosomal mRNAs. TargetScan and miRanda were used for identifying the miRNA-mRNA and miRNA-LncRNA interactions. The predicted lncRNAs and mRNAs were intersected with the differentially expressed genes, for which the screening criterion was fold change >1.5 in the microarray. Differentially expressed exosomal miRNAs were identified in the GSE71008 dataset, and differentially expressed mRNAs (DEmRNAs) were further summarized from The Cancer Genome Atlas (TCGA) database.Results: A total of 1186 exosomal DEmRNAs, 2088 exosomal DElncRNAs and 29 exosomal miRNAs were detected in CRC patients compared to the healthy donors. Functional enrichment analysis suggested that exosomal DEmRNAs might participate in pathways related to carcinogenesis and development of cancer. An exosomal ceRNA regulatory network of CRC was constructed based on 40 lncRNAs, two miRNAs, and five mRNAs. Exosomal miR-150-5p and miR-10b-5p expression levels were increased in healthy donors compared with CRC patients in the GSE71008 dataset, and five DEmRNAs (TOMM70A, RBM48, BEND3, RHOBTB1, and ADAMTS2) were significantly upregulated in TCGA database. Two potential exosomal regulatory axes of lncRNA G016261-miR-150-5p-RBM48 and lncRNA XLOC_011677-miR-10b-5p-BEND3 were identified from the network.Conclusion: The current study revealed potential molecular biological regulation pathways and diagnostic biomarkers through the exosomal ceRNA regulatory network.
Collapse
|
13
|
Gu J, Sun R, Tang D, Liu F, Chang X, Wang Q. Astragalus mongholicus Bunge-Curcuma aromatica Salisb. suppresses growth and metastasis of colorectal cancer cells by inhibiting M2 macrophage polarization via a Sp1/ZFAS1/miR-153-3p/CCR5 regulatory axis. Cell Biol Toxicol 2022; 38:679-697. [PMID: 35072892 DOI: 10.1007/s10565-021-09679-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is regarded as one of the commonest cancer types around the world. Due to the poor understanding on the causes of CRC formation and progression, this study sets out to investigate the physiological mechanisms by which Astragalus mongholicus Bunge-Curcuma aromatica Salisb. (ARCR) regulates CRC growth and metastasis, and the role in which M2 macrophage polarization plays in this process. An orthotopic-transplant model of CRC was established to evaluate the influence of ARCR on the polarization of M2 macrophage and the growth and metastasis of tumors. Next, the binding affinity among Sp1, ZFAS1, miR-153-5p, and CCR5 was identified using multiple assays. Finally, after co-culture of bone marrow-derived macrophages (BMDM) with CRC cell line CT26.WT, the cell proliferative, invasive, and migrated abilities were assessed in gain- or loss-of-function experiments. ARCR inhibited the infiltration of M2 macrophages into tumor microenvironment to suppress the CRC growth and metastasis in vivo. Additionally, ARCR inhibited the transcription of ZFAS1 by reducing Sp1 expression to repress M2 macrophage polarization. Moreover, ZFAS1 competitively binds to miR-153-3p to upregulate the CCR5 expression. Finally, ARCR suppressed the polarization of M2 macrophages to inhibit the tumor growth and tumor metastasis in CRC by mediating the Sp1/ZFAS1/miR-153-3p/CCR5 regulatory axis. Collectively, ARCR appears to suppress the CRC cell growth and metastasis by suppressing M2 macrophage polarization via Sp1/ZFAS1/miR-153-3p/CCR5 regulatory axis. 1. ARCR suppress the CRC cell growth and metastasis 2. ZFAS1 promotes CCR5 expression by competitively binding to miR-153-3p. 3. Sp1 promotes M2 macrophage polarization by activating ZFAS1 via miR-153-3p/CCR5. 4. The study unveiled a protective target against CRC.
Collapse
Affiliation(s)
- Junfei Gu
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138# Xianlin Road, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Ruolan Sun
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138# Xianlin Road, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Decai Tang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138# Xianlin Road, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Fuyan Liu
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138# Xianlin Road, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Xiangwei Chang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Qiaohan Wang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138# Xianlin Road, Qixia District, Nanjing, 210023, Jiangsu Province, China.
| |
Collapse
|
14
|
Zeng Z, Chen Y, Geng X, Zhang Y, Wen X, Yan Q, Wang T, Ling C, Xu Y, Duan J, Zheng K, Sun Z. NcRNAs: Multi‑angle participation in the regulation of glioma chemotherapy resistance (Review). Int J Oncol 2022; 60:76. [PMID: 35506469 PMCID: PMC9083885 DOI: 10.3892/ijo.2022.5366] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022] Open
Abstract
As the most common primary tumour of the central nervous system, gliomas have a high recurrence rate after surgical resection and are resistant to chemotherapy, particularly high‑grade gliomas dominated by glioblastoma multiforme (GBM). The prognosis of GBM remains poor despite improvements in treatment modalities, posing a serious threat to human health. At present, although drugs such as temozolomide, cisplatin and bevacizumab, are effective in improving the overall survival of patients with GBM, most patients eventually develop drug resistance, leading to poor clinical prognosis. The development of multidrug resistance has therefore become a major obstacle to improving the effectiveness of chemotherapy for GBM. The ability to fully understand the underlying mechanisms of chemotherapy resistance and to develop novel therapeutic targets to overcome resistance is critical to improving the prognosis of patients with GBM. Of note, growing evidence indicates that a large number of abnormally expressed noncoding RNAs (ncRNAs) have a central role in glioma chemoresistance and may target various mechanisms to modulate chemosensitivity. In the present review, the roles and molecular mechanisms of ncRNAs in glioma drug resistance were systematically summarized, the potential of ncRNAs as drug resistance markers and novel therapeutic targets of glioma were discussed and prospects for glioma treatment were outlined. ncRNAs are a research direction for tumor drug resistance mechanisms and targeted therapies, which not only provides novel perspectives for reversing glioma drug resistance but may also promote the development of precision medicine for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Zhaomu Zeng
- Department of Surgery, School of Clinical Medicine, Hebei University, Baoding, Hebei 071000, P.R. China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Xiuchao Geng
- Department of Nursing, School of Medicine, Taizhou University, Jiaojiang, Zhejiang 318000, P.R. China
| | - Yuhao Zhang
- Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated to Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Xichao Wen
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Qingyu Yan
- Office of Academic Research, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Tingting Wang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Chen Ling
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yan Xu
- Clinical Laboratory, Affiliated Hospital of Jinggangshan University, Ji'an, Jiangxi 343100, P.R. China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Kebin Zheng
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Zhiwei Sun
- Department of Surgery, School of Clinical Medicine, Hebei University, Baoding, Hebei 071000, P.R. China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
15
|
Ghafouri-Fard S, Kamali MJ, Abak A, Shoorei H, Taheri M. LncRNA ZFAS1: Role in tumorigenesis and other diseases. Biomed Pharmacother 2021; 142:111999. [PMID: 34385106 DOI: 10.1016/j.biopha.2021.111999] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/23/2021] [Accepted: 08/01/2021] [Indexed: 12/28/2022] Open
Abstract
Residing on chromosome 20q13.13, Zinc Finger NFX1-Type Containing 1 (ZNFX1) antisense RNA 1 (ZFAS1) is a transcript which has been primarily recognized as a modulator of differentiation of alveolar and epithelial cell in the mammary gland. This long non-coding RNA (lncRNA) partakes in the molecular cascades leading to several non-neoplastic conditions such as osteoarthritis, epilepsy, rheumatoid arthritis, atherosclerosis, pulmonary fibrosis, myocardial infarction, and cardiac dysfunction. More importantly, ZFAS1 is considered as an oncogene in almost all types of cancers. Using expression amounts of ZFAS1, it is possible to forecast the clinical outcome of patients with different neoplasms such as colorectal cancer, gastric cancer, cholangiocarcinoma, hepatoblastoma, and other types of cancer. We describe the role of ZFAS1 in the development of neoplastic and non-neoplastic disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Atefe Abak
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Wang H, Hu H, Ma J, Jiang Y, Cheng R. lncRNA ZFAS1 promotes the ox-LDL induced proliferation, invasion and migration of vascular smooth muscle cells. Exp Ther Med 2021; 22:835. [PMID: 34149881 PMCID: PMC8200810 DOI: 10.3892/etm.2021.10267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory vascular disease. The dysfunction of vascular smooth muscle cells (VSMCs) induced by oxidized low-density lipoprotein (ox-LDL) contributes to the formation of atherosclerotic lesions. Additionally, upregulation of the long non-coding RNA zinc finger antisense 1 (ZFAS1) was observed in the plaques of patients with atherosclerosis. The aim of the present study was to explore the functional role of ZFAS1 in atherosclerosis progression. Reverse transcription-quantitative PCR was performed to analyze ZFAS1 mRNA expression, and western blotting was performed to determine the protein expression levels of Ki67, proliferating cell nuclear antigen (PCNA), matrix metallopeptidase (MMP)2 and MMP9. The Cell Counting Kit-8 assay was used to test cell viability. Finally, wound healing and Transwell chamber assays were performed to evaluate cell migration and invasion, respectively. The current findings demonstrated that ZFAS1 expression was upregulated by ox-LDL stimulation in VSMCs. Moreover, ZFAS1 overexpression promoted the ox-LDL-induced proliferation, migration and invasion of VSMCs, and upregulated the expression levels of proteins associated with cellular proliferation (Ki67 and PCNA), migration and invasion (MMP2 and 9). By contrast, ZFAS1-knockdown inhibited the proliferation, migration and invasion of VSMCs, and suppressed cell proliferation-, migration- and invasion-associated protein expression. In conclusion, ZFAS1 promoted the ox-LDL-induced proliferation, invasion and migration of VSMCs. Thus, ZFAS1 may represent a novel biomarker for dysfunction of VSMCs in the pathological condition of atherosclerosis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Huajie Hu
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Junjie Ma
- Department of Clinical Laboratory, Linhai Traditional Chinese Medicine Hospital Medical Community, Linhai, Zhejiang 317000, P.R. China
| | - Yafeng Jiang
- Department of Medical Genetics and Prenatal Diagnosis, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu 222000, P.R. China
| | - Ruifei Cheng
- Department of Clinical Laboratory, Lianyungang Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu 222004, P.R. China
| |
Collapse
|
17
|
Kim SH, Lim KH, Yang S, Joo JY. Long non-coding RNAs in brain tumors: roles and potential as therapeutic targets. J Hematol Oncol 2021; 14:77. [PMID: 33980320 PMCID: PMC8114507 DOI: 10.1186/s13045-021-01088-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022] Open
Abstract
Brain tumors are associated with adverse outcomes despite improvements in radiation therapy, chemotherapy, and photodynamic therapy. However, treatment approaches are evolving, and new biological phenomena are being explored to identify the appropriate treatment of brain tumors. Long non-coding RNAs (lncRNAs), a type of non-coding RNA longer than 200 nucleotides, regulate gene expression at the transcriptional, post-transcriptional, and epigenetic levels and are involved in a variety of biological functions. Recent studies on lncRNAs have revealed their aberrant expression in various cancers, with distinct expression patterns associated with their instrumental roles in cancer. Abnormal expression of lncRNAs has also been identified in brain tumors. Here, we review the potential roles of lncRNAs and their biological functions in the context of brain tumors. We also summarize the current understanding of the molecular mechanisms and signaling pathways related to lncRNAs that may guide clinical trials for brain tumor therapy.
Collapse
Affiliation(s)
- Sung-Hyun Kim
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Key-Hwan Lim
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Sumin Yang
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jae-Yeol Joo
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| |
Collapse
|
18
|
Liu L, Li X, Shi Y, Chen H. Long noncoding RNA DLGAP1-AS1 promotes the progression of glioma by regulating the miR-1297/EZH2 axis. Aging (Albany NY) 2021; 13:12129-12142. [PMID: 33901010 PMCID: PMC8109124 DOI: 10.18632/aging.202923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Dysregulated lncRNAs have been implicated in a plethora of tumors, including glioma. One such oncogenic lncRNAs that has been reported in several cancers is the lncRNA DLGAP1 antisense RNA 1 (DLGAP1-AS1). This study seeks to characterize the expression of DLGAP1-AS1 in glioma tissues, which we found to be raised in both glioma samples and cell lines. Functional experiments revealed that DLGAP1-AS1 promoted in vitro glioma cell invasion, migration and proliferation. DLGAP1-AS1 was found to function as a miR-1297 sponge, based on information from luciferase reporter assays, RNA pull-down assays and publicly available online databases. miR-1297 was in turn found to functionally target EZH2. DLGAP1-AS1 modulated EZH2 expressions through miR-1297 sponging. Glioma progression appears to be supported DLGAP1-AS1 -promoted activation of the miR-1297/EZH2 axis. The components of this axis may function as therapeutic targets for glioma.
Collapse
Affiliation(s)
- Liang Liu
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaojian Li
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yan Shi
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Hua Chen
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
19
|
Miao C, Bai L, Yang Y, Huang J. Dysregulation of lncRNAs in Rheumatoid Arthritis: Biomarkers, Pathogenesis and Potential Therapeutic Targets. Front Pharmacol 2021; 12:652751. [PMID: 33776780 PMCID: PMC7994855 DOI: 10.3389/fphar.2021.652751] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease of unknown etiology, mainly manifested by persistent abnormal proliferation of fibroblast-like synoviocytes (FLSs), inflammation, synovial hyperplasia and cartilage erosion, accompanied by joint swelling and joint destruction. Abnormal expression or function of long noncoding RNAs (lncRNAs) are closely related to human diseases, including cancers, mental diseases, autoimmune diseases and others. The abnormal sequence and spatial structure of lncRNAs, the disorder expression and the abnormal interaction with the binding protein will lead to the change of gene expression in the way of epigenetic modification. Increasing evidence demonstrated that lncRNAs were involved in the activation of FLSs, which played a key role in the pathogenesis of RA. In this review, the research progress of lncRNAs in the pathogenesis of RA was systematically summarized, including the role of lncRNAs in the diagnosis of RA, the regulatory mechanism of lncRNAs in the pathogenesis of RA, and the intervention role of lncRNAs in the treatment of RA. Furthermore, the activated signal pathways, the role of DNA methylation and other mechanism have also been overview in this review.
Collapse
Affiliation(s)
- Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Anhui Provincial Key Laboratory of Chinese Medicine Compound, Anhui University of Chinese Medicine, Hefei, China.,Department of Pharmacy, School of Life and Health Sciences, Anhui University of Science and Technology, Fengyang, China
| | - Liangliang Bai
- Department of Biomedical Engineering, School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Yaru Yang
- Department of Pharmacy, First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Jinling Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
20
|
Zhou WJ, Zhang J, Xie F, Wu JN, Ye JF, Wang J, Wu K, Li MQ. CD45RO -CD8 + T cell-derived exosomes restrict estrogen-driven endometrial cancer development via the ERβ/miR-765/PLP2/Notch axis. Theranostics 2021; 11:5330-5345. [PMID: 33859750 PMCID: PMC8039953 DOI: 10.7150/thno.58337] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: Estrogen-dependent cancers (e.g., breast, endometrial, and ovarian cancers) are among the leading causes of morbidity and mortality in women worldwide. Recently, exosomes released by tumor-infiltrating CD8+ T cells have been under the spotlight in the field of cancer immunotherapy. Our study aims at elucidating the underlying mechanisms of the crosstalk between estrogen signaling and CD8+ T cells, and possible intervention values in uterine corpus endometrial cancer (UCEC). Methods: Micro RNA-seq was conducted to screen differentially expressed micro RNA in UCEC. Bioinformatic analysis was processed to predict the target of miR-765. RNA silencing or overexpressing and pharmacologic inhibitors were used to assess the functions of ERβ/miR-765/PLP2/Notch axis in UCEC cell proliferation and invasion in vivo and in vitro. In vivo imaging was performed to evaluate the metastasis of tumor in mice. Combined fluorescent in situ hybridization for miR-765 and immunofluorescent labeling for CD8 was carried out to prove the co-localization between miR-765 and CD8+ T cells. Exosomes derived from CD45RO-CD8+ T cells were isolated to detect the regulatory effects on UCEC. Results: miR-765 is characterized as the most downregulated miRNA in UCEC, and there is a negative correlation between miR-765 and Proteolipid protein 2 (PLP2) in UCEC lesion. Estrogen significantly down-regulates miR-765 level, and facilitates the development of UCEC by estrogen receptor (ER) β. Mechanistically, this process is mediated through the miRNAs (e.g., miR-3584-5p, miR-7-5p, miR-150-5p, and miR-124-3p) cluster-controlled regulation of the PLP2, which further regulates Ki-67 and multiple epithelial-mesenchymal transition (EMT)-related molecules (e.g, E-cadherin and Vimentin) in a Notch signaling pathway-dependent manner. Interestingly, the selective ER degrader Fulvestrant alleviates estrogen-mediated miR-765/PLP2 expression regulation and UCEC development in ERβ-dependent and -independent manners. Additionally, CD45RO-CD8+ T cell-derived exosomes release more miR-765 than that from CD45RO+CD8+ T cells. In therapeutic studies, these exosomes limit estrogen-driven disease development via regulation of the miR-765/PLP2 axis. Conclusions: This observation reveals novel molecular mechanisms underlying estrogen signaling and CD8+ T cell-released exosomes in UCEC development, and provides a potential therapeutic strategy for UCEC patients with aberrant ERβ/miR-765/PLP2/Notch signaling axis.
Collapse
Affiliation(s)
- Wen-Jie Zhou
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100, Haining Road, Shanghai 200080, People's Republic of China
| | - Feng Xie
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Jiang-Nan Wu
- Clinical Epidemiology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, People's Republic of China
| | - Jiang-Feng Ye
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, 229899, Singapore
| | - Jian Wang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
| | - Ke Wu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100, Haining Road, Shanghai 200080, People's Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| |
Collapse
|
21
|
Chaudhary R. Potential of long non-coding RNAs as a therapeutic target and molecular markers in glioblastoma pathogenesis. Heliyon 2021; 7:e06502. [PMID: 33786397 PMCID: PMC7988331 DOI: 10.1016/j.heliyon.2021.e06502] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/20/2020] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GB) is by far the most hostile type of malignant tumor that primarily affects the brain and spine, derived from star-shaped glial cells that are astrocytes and oligodendrocytes. Despite of significant efforts in recent years in glioblastoma research, the clinical efficacy of existing medical intervention is still limited and very few potential diagnostic markers are available. Long non-coding RNAs (lncRNAs) that lacks protein-coding capabilities were previously thought to be "junk sequences" in mammalian genomes are quite indispensible epigenetic regulators that can positively or negatively regulate gene expression and nuclear architecture, with significant roles in the initiation and development of tumors. Nevertheless, the precise mechanism of these distortedly expressed lncRNAs in glioblastoma pathogenesis is not yet fully understood. Since the advent of high-throughput sequencing technologies, more and more research have elucidated that lncRNAs are one of the most promising prognostic biomarkers and therapeutic targets for glioblastoma. In this paper, I briefly outlined the existing findings of lncRNAs. And also summarizes the profiles of different lncRNAs that have been broadly classified in glioblastoma research, with emphasis on both their prognostic and therapeutic values.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| |
Collapse
|
22
|
Silencing of Long Noncoding RNA Zinc Finger Antisense 1 Protects Against Hypoxia/Reoxygenation-induced Injury in HL-1 Cells Through Targeting the miR-761/Cell Death Inducing p53 Target 1 Axis. J Cardiovasc Pharmacol 2020; 76:564-573. [DOI: 10.1097/fjc.0000000000000896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Zhang B, Chen J, Cui M, Jiang Y. LncRNA ZFAS1/miR-1271-5p/HK2 Promotes Glioma Development Through Regulating Proliferation, Migration, Invasion and Apoptosis. Neurochem Res 2020; 45:2828-2839. [DOI: 10.1007/s11064-020-03131-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 01/03/2023]
|
24
|
Su D, Ji Z, Xue P, Guo S, Jia Q, Sun H. Long-Noncoding RNA FGD5-AS1 Enhances the Viability, Migration, and Invasion of Glioblastoma Cells by Regulating the miR-103a-3p/TPD52 Axis. Cancer Manag Res 2020; 12:6317-6329. [PMID: 32848452 PMCID: PMC7425657 DOI: 10.2147/cmar.s253467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose This study was designed to explore the functional role of FYVE, RhoGEF, and PH domain containing 5 antisense RNA 1 (FGD5-AS1) and the underlying regulatory mechanism in the progression of glioblastoma (GBM). Materials and Methods FGD5-AS1 expression was analyzed in The Cancer Genome Atlas (TCGA), and then detected in GBM tissues and cells by quantitative reverse-transcription polymerase chain reaction. Viability, migration, and invasion of GBM cells were assessed using the MTT, wound healing, and transwell assays, respectively. StarBase/TargetScan analysis and dual-luciferase reporter gene (DLR) assay were performed to investigate the relationship between FGD5-AS1/tumor protein D52 (TPD52) and miR-103a-3p. A xenograft tumor model was established to evaluate the role of FGD5-AS1 in GBM tumorigenesis in vivo. Results FGD5-AS1 was overexpressed in GBM tissues and cells, and silencing of FGD5-AS1 expression resulted in the inhibition of the viability, migration, and invasion of GBM cells. miR-130-3p was a target of FGD5-AS1, and its expression was negatively regulated by FGD5-AS1. Silencing miR-103a-3p expression resulted in the abrogation of the inhibitory effects of si-FGD5-AS1 on the viability, migration, and invasion of GBM cells. TPD52 was a target of miR-103a-3p and suppressed the antitumor effects of FGD5-AS1 silencing on GBM cells. In addition, FGD5-AS1 silencing inhibited the growth of xenograft tumors in vivo by modulating the miR-103a-3p/TPD52 axis. Conclusion Silencing of FGD5-AS1 inhibited the viability, migration, and invasion of GBM cells by regulating the miR-103a-3p/TPD52 axis.
Collapse
Affiliation(s)
- Daoqing Su
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Zhengang Ji
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Pengfei Xue
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Shengfu Guo
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| | - Hanyu Sun
- Department of Neurosurgery, Liaocheng People's Hospital and Liaocheng Brain Hospital, Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng City, Shandong Province, People's Republic of China
| |
Collapse
|
25
|
Shi W, Gao Z, Song J, Wang W. Silence of FOXD2-AS1 inhibited the proliferation and invasion of esophagus cells by regulating miR-145-5p/CDK6 axis. Histol Histopathol 2020; 35:1013-1021. [PMID: 32524576 DOI: 10.14670/hh-18-232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study aimed to investigate the function of long non-coding RNA FOXD2 adjacent opposite strand RNA 1 (lncRNA FOXD2-AS1) during the progression of esophagus cancer (EC) and explore its underlying molecular mechanisms. The level of FOXD2-AS1 in EC tissues and paracancerous tissues was detected by using RT-qPCR; ROC curve was used to evaluate the diagnostic value of FOXD2-AS1 for EC. In addition, CCK8 assay and immunofluorescence staining assay were used to detect the proliferation of Eca-109 and TE-1 cells. To investigate the function of FOXD2-AS1 on cell apoptosis and cell cycle, flow cytometry was performed. To detect the invasion ability of EC cells, transwell invasion assay was performed. Starbase3.0 and Targetscan were used to predict the target genes of FOXD2-AS1 and miR-145-5p, and protein expressions were detected with western blot. We found FOXD2-AS1 was significantly upregulated in EC tissues compared with adjacent normal tissues, which was positively correlated with clinicopathological parameters of patients with EC. Downregulation of FOXD2-AS1 inhibited the proliferation and invasion by inducing apoptosis of EC cells. Moreover, FOXD2-AS1 may regulate the expression of CDK6 by targeting miR-145-3p. Meanwhile, silencing of FOXD2-AS1 caused G1 phase arrest of EC cells by reducing the expression of CDK6. In conclusion, silening FOXD2-AS1 significantly inhibited the proliferation and invasion of EC cells by regulating the miR-145-5p/CDK6 axis. Therefore, FOXD2-AS1 might be used as diagnostic biomarker and therapeutic target for EC.
Collapse
Affiliation(s)
- Woda Shi
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Zhengya Gao
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Jianxiang Song
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Wencai Wang
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu, China.
| |
Collapse
|
26
|
Wu H, Qin W, Lu S, Wang X, Zhang J, Sun T, Hu X, Li Y, Chen Q, Wang Y, Zhao H, Piao H, Zhang R, Wei M. Long noncoding RNA ZFAS1 promoting small nucleolar RNA-mediated 2'-O-methylation via NOP58 recruitment in colorectal cancer. Mol Cancer 2020; 19:95. [PMID: 32443980 PMCID: PMC7243338 DOI: 10.1186/s12943-020-01201-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/15/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Increasing evidence supports the role of small nucleolar RNAs (snoRNAs) and long non-coding RNAs (lncRNAs) as master gene regulators at the epigenetic modification level. However, the underlying mechanism of these functional ncRNAs in colorectal cancer (CRC) has not been well investigated. METHODS The dysregulated expression profiling of lncRNAs-snoRNAs-mRNAs and their correlations and co-expression enrichment were assessed by GeneChip microarray analysis. The candidate lncRNAs, snoRNAs, and target genes were detected by in situ hybridization (ISH), RT-PCR, qPCR and immunofluorescence (IF) assays. The biological functions of these factors were investigated using in vitro and in vivo studies that included CCK8, trans-well, cell apoptosis, IF assay, western blot method, and the xenograft mice models. rRNA 2'-O-methylation (Me) activities were determined by the RTL-P assay and a novel double-stranded primer based on the single-stranded toehold (DPBST) assay. The underlying molecular mechanisms were explored by bioinformatics and RNA stability, RNA fluorescence ISH, RNA pull-down and translation inhibition assays. RESULTS To demonstrate the involvement of lncRNA and snoRNAs in 2'-O-Me modification during tumorigenesis, we uncovered a previously unreported mechanism linking the snoRNPs NOP58 regulated by ZFAS1 in control of SNORD12C, SNORD78 mediated rRNA 2'-O-Me activities in CRC initiation and development. Specifically, ZFAS1 exerts its oncogenic functions and significantly up-regulated accompanied by elevated NOP58, SNORD12C/78 expression in CRC cells and tissues. ZFAS1 knockdown suppressed CRC cell proliferation, migration, and increased cell apoptosis, and this inhibitory effect could be reversed by NOP58 overexpression in vitro and in vivo. Mechanistically, the NOP58 protein could be recognized by the specific motif (AAGA or CAGA) of ZFAS1. This event accelerates the assembly of SNORD12C/78 to allow for further guiding of 2'-O-Me at the corresponding Gm3878 and Gm4593 sites. Importantly, silencing SNORD12C or 78 reduced the rRNAs 2'-O-Me activities, which could be rescued by overexpression ZFAS1, and this subsequently inhibits the RNA stability and translation activity of their downstream targets (e.g., EIF4A3 and LAMC2). CONCLUSION The novel ZFAS1-NOP58-SNORD12C/78-EIF4A3/LAMC2 signaling axis that functions in CRC tumorigenesis provides a better understanding regarding the role of lncRNA-snoRNP-mediated rRNAs 2'-O-Me activities for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Wenyan Qin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Senxu Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiufang Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Tong Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaoyun Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yalun Li
- Department of Anorectal Surgery, First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Qiuchen Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yuanhe Wang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Shenyang, 110042, People's Republic of China
| | - Haishan Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China
| | - Haiyan Piao
- Department of Medical Oncology, Cancer Hospital of China Medical University, Shenyang, 110042, People's Republic of China
| | - Rui Zhang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Shenyang, 110042, People's Republic of China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
27
|
Duan R, Li C, Wang F, Han F, Zhu L. The Long Noncoding RNA ZFAS1 Potentiates the Development of Hepatocellular Carcinoma via the microRNA-624/MDK/ERK/JNK/P38 Signaling Pathway. Onco Targets Ther 2020; 13:4431-4444. [PMID: 32547074 PMCID: PMC7250709 DOI: 10.2147/ott.s246278] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Background A long noncoding RNA (lncRNA), ZNFX1 antisense RNA 1 (ZFAS1), was increased in multiple cancers, including hepatocellular carcinoma (HCC), resulting in malignancy development and progression. However, the mechanisms involving the interaction between ZFAS1 and microRNA-624 (miRNA-624) remain largely unknown. Therefore, the goal of this study was to probe the functional role of ZFAS1 in the development of HCC and its underlying mechanism. Methods Firstly, differentially expressed lncRNAs in HCC tissues were screened out by microarray. Subsequently, the prognostic effect of ZFAS1 patients with HCC was analyzed by the Kaplan-Meier analysis and The Cancer Genome Atlas database. ZFAS1 regulation on miRNA-624 was determined after si-ZFAS1 and/or miRNA-624 inhibitor were transfected into HepG2 and SMMC7721 cell lines. Finally, the effects of ZFAS1 on the growth and metastasis of HCC were observed by in vivo tumorigenesis and metastasis tests. Results ZFAS1 was overexpressed in HCC tissues and cells and indicated worse prognosis and shorter survival in patients with HCC. Silencing of ZFAS1 inhibited the malignancy of HCC cells, but miR-624 inhibitor could partially reverse the repressive role of si-ZFAS1. Moreover, ZFAS1 induced the extracellular-regulated protein kinases/c-Jun N-terminal kinase (ERK/JNK)/P38 pathway by binding to midkine (MDK) through miR-624, thus promoting the occurrence of HCC. Conclusion Collectively, ZFAS1 depletion inhibited the occurrence of HCC by downregulating the MDK/ERK/JNK/P38 pathway through restoring miR-624 expression. Inhibition of ZFAS1 may act as an innovative target to suppress occurrence in HCC.
Collapse
Affiliation(s)
- Rui Duan
- Department of Hepatological Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Caiyan Li
- Department of Clinical Laboratory, The Second People's Hospital of Jingmen, Jingmen 448000, Hubei, People's Republic of China
| | - Fan Wang
- Department of Hepatological Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Fei Han
- Department of Oncology, Affiliated Hospital of Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Ling Zhu
- Department of Hepatological Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| |
Collapse
|
28
|
Yu Y, Lu X, Yang C, Yin F. Long Noncoding RNA LINC00173 Contributes to the Growth, Invasiveness and Chemo-Resistance of Colorectal Cancer Through Regulating miR-765/PLP2 Axis. Cancer Manag Res 2020; 12:3363-3369. [PMID: 32494200 PMCID: PMC7229794 DOI: 10.2147/cmar.s251029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Long noncoding RNA has been involved in tumorigenesis of colorectal cancer (CRC). This study aimed to illustrate the functions and mechanisms of LINC00173 in CRC progression. Methods The expression of LINC00173 in CRC tissues and cell lines were analyzed via qRT-PCR. Kaplan-Meier curve was used to determine survival rate. Luciferase reporter assay was conducted to evaluate the interactions among LINC00173, miR-765 and PLP2 (proteolipid protein 2). CCK8 assay, EdU assay, transwell assay and xenograft assay were performed to examine the effect of LINC00173/miR-765/PLP2 axis on proliferation, migration and invasion. The Ki67 expression level in tumors tissues was detected through immunofluorescence assay. Results LINC00173 expression was markedly upregulated in CRC tissues and cells. High expression level of LINC00173 in CRC patients was correlated with poor prognosis. LINC00173 knockdown inhibited proliferation, migration, invasion and chemo-resistance of CRC cells in vitro. LINC00173 downregulation delayed CRC growth in vivo. LINC00173 interacted with miR-765 to promote PLP2 expression. Conclusion Our results demonstrated that LINC00173 plays an important oncogenic role in CRC via modulating miR-765/PLP2 axis. And LINC00173 may be a potential prognostic biomarker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Yanhui Yu
- Physical Examination Center, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Xiuyun Lu
- Department of Traditional Chinese Medicine, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Chuandong Yang
- Department of Orthopedics, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Fengxiang Yin
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Harbin Chest Hospital, Harbin 150056, People's Republic of China
| |
Collapse
|
29
|
Wang J, Lv W, Lin Z, Wang X, Bu J, Su Y. Hsa_circ_0003159 inhibits gastric cancer progression by regulating miR-223-3p/NDRG1 axis. Cancer Cell Int 2020; 20:57. [PMID: 32099530 PMCID: PMC7031989 DOI: 10.1186/s12935-020-1119-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Background Abnormally expressed circular RNAs (circRNAs) are implicated in the development and treatment of gastric cancer (GC). Previous study has reported that hsa_circ_0003159 is expressed in GC. However, the role and mechanism of hsa_circ_0003159 in GC progression remain unclear. Methods GC tissues and normal tissues were harvested from 55 patients in this study. The levels of hsa_circ_0003159, microRNA (miR)-223-3p and N-myc downstream regulated gene 1 (NDRG1) were measured by quantitative real-time polymerase chain reaction or western blot. Cell proliferation, migration, invasion and apoptosis were determined by cell counting kit (CCK)-8, transwell assay, flow cytometry and western blot, respectively. The target association of miR-223-3p-hsa_circ_0003159 and miR-223-3p-NDRG1 was explored by dual-luciferase reporter assay. Xenograft model was established to assess the roles of hsa_circ_0003159 in GC in vivo. Results Hsa_circ_0003159 was lowly expressed in GC tissues and cells and mainly presented in the cytoplasm. Low expression of hsa_circ_0003159 was associated with lower overall survival and disease-free survival. Hsa_circ_0003159 overexpression inhibited proliferation, migration and invasion but induced apoptosis in GC cells. MiR-223-3p was a target of hsa_circ_0003159 and abated the effect of hsa_circ_0003159 on proliferation, migration, invasion and apoptosis in GC cells. Hsa_circ_0003159 promoted NDRG1 expression by competitively sponging miR-223-3p. Knockdown of NDRG1 reversed the suppressive effect of hsa_circ_0003159 on GC progression. Besides, hsa_circ_0003159 decreased GC cell xenograft tumor growth by regulating miR-223-3p and NDRG1. Conclusion Hsa_circ_0003159 suppressed proliferation, migration, invasion and xenograft tumor growth but promoted apoptosis by decreasing miR-223-3p and increasing NDRG1 in GC, indicating a novel target for treatment of GC.
Collapse
Affiliation(s)
- Jingyu Wang
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Weize Lv
- 2Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zhidong Lin
- 3Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xiao Wang
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Juyuan Bu
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Yonghui Su
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| |
Collapse
|