1
|
Zhou J, Liu S, Zhang J, Zeng Q, Lin Z, Fu R, Lin Y, Hu Z. Discovery and validation of Hsa-microRNA-3665 promoter methylation as a potential biomarker for the prognosis of esophageal squaous cell carcinoma. Int J Clin Oncol 2025; 30:309-319. [PMID: 39630213 PMCID: PMC11785691 DOI: 10.1007/s10147-024-02656-3] [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] [Received: 03/05/2024] [Accepted: 11/03/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Methylation of microRNA (miRNA) promoters associated with diseases is a common epigenetic mechanism in the development of various human cancers. However, its relationship with prognosis in esophageal squamous cell carcinoma (ESCC) remains unclear. This study aims to explore the association between the methylation level of has-miR-3665 promoter and prognosis in ESCC. METHODS Human miRNA data were downloaded from miRbase, and we identified CpG islands of these human miRNAs by genomics browser analysis. MiRNA methylation levels were detected by methylation-specific high-resolution melting. Gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to explore the molecular mechanism of hsa-miR-3665. Cox regression analysis was used to investigate prognostic factors. The overall survival rate was predicted by a nomogram. RESULTS We found that 88 human miRNAs had promoter methylatio, of which 15 miRNAs were found to be epigenetically regulated in ESCC cells compared with their normal counterparts, including hsa-miR-3665. Meanwhile, hsa-miR-3665 expression was significantly lower in ESCC tumour tissue than in adjacent tissue (P = 0.03). GO and KEGG analyses demonstrated that the target genes are involved in protein transport, transcription regulator activity, MAPK and RAS signaling pathway. High hsa-miR-3665 promoter methylation levels were associated with a poor prognosis (HR = 3.89, 95% CI 1.11 ~ 13.55). Moreover, a nomogram incorporating the hsa-miR-3665 methylation level and clinical factors presented a good performance for predicting survival in the training and validation tests, with C-indices of 0.748 and 0.751, respectively. CONCLUSIONS High hsa-miR-3665 promoter methylation levels may be a potential biomarker for the progression of ESCC.
Collapse
Affiliation(s)
- Jinsong Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Shuang Liu
- Sun Yat-Sen University Cancer Center/Cancer Hospital, Guangzhou, 510060, China
| | - Juwei Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Qiaoyan Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Rong Fu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Yulan Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350108, China.
| |
Collapse
|
2
|
Yin X, Li J, Zhao J, Zheng W, Zhang A, Ma J. Epigenetic modifications involving ncRNAs in digestive system cancers: focus on histone modification. Clin Epigenetics 2024; 16:162. [PMID: 39563475 PMCID: PMC11577885 DOI: 10.1186/s13148-024-01773-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
In recent years, epigenetic modifications have been strongly linked to tumor development, with histone modifications representing a key epigenetic mechanism. In addition, non-coding RNAs (ncRNAs) play a critical role in regulating cancer-related pathways. The abnormal interaction between histone modifications and ncRNAs, both pivotal epigenetic regulators, has been widely observed across various cancer types. Here, we systematically explore the molecular mechanisms through which histone modifications and ncRNAs contribute in the pathogenesis of digestive system cancers, and aberrant ncRNA-mediated histone modifications manipulate various biological behaviors of tumor cells including proliferation, migration, angiogenesis, etc. In addition, we provide new insights into diagnostic, prognostic markers, therapeutic targets and chemoradiation resistance for digestive system cancers from the epigenetic perspective.
Collapse
Affiliation(s)
- Xiaodi Yin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Jingyi Li
- Intensive Care Medicine, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jiahui Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Weihan Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Aohua Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Jun Ma
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China.
| |
Collapse
|
3
|
Zhang L, Wang Y, Gao J, Zhou X, Huang M, Wang X, He Z. Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review). Oncol Lett 2024; 27:255. [PMID: 38646493 PMCID: PMC11027111 DOI: 10.3892/ol.2024.14388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Esophageal cancer (EC) is a common form of malignant tumor in the digestive system that is classified into two types: Esophageal squamous cell carcinomas (ESCC) and esophageal adenocarcinoma. ESCC is known for its early onset of symptoms, which can be difficult to identify, as well as its rapid progression and tendency to develop drug resistance to chemotherapy and radiotherapy. These factors contribute to the high incidence of disease and low cure rate. Therefore, a diagnostic biomarker and therapeutic target need to be identified for ESCC. Non-coding RNAs (ncRNAs) are a class of molecules that are transcribed from DNA but do not encode proteins. Initially, ncRNAs were considered to be non-functional segments generated during transcription. However, with advancements in high-throughput sequencing technologies in recent years, ncRNAs have been associated with poor prognosis, drug resistance and progression of ESCC. The present study provides a comprehensive overview of the biogenesis, characteristics and functions of ncRNAs, particularly focusing on microRNA, long ncRNAs and circular RNAs. Furthermore, the ncRNAs that could potentially be used as diagnostic biomarkers and therapeutic targets for ESCC are summarized to highlight their application value and prospects in ESCC.
Collapse
Affiliation(s)
- Longze Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yanyang Wang
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianmei Gao
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xue Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
4
|
Wei QY, Jin F, Wang ZY, Li BJ, Cao WB, Sun ZY, Mo SJ. MicroRNAs: A novel signature in the metastasis of esophageal squamous cell carcinoma. World J Gastroenterol 2024; 30:1497-1523. [PMID: 38617454 PMCID: PMC11008420 DOI: 10.3748/wjg.v30.i11.1497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/12/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignant epithelial tumor, characterized by squamous cell differentiation, it is the sixth leading cause of cancer-related deaths globally. The increased mortality rate of ESCC patients is predominantly due to the advanced stage of the disease when discovered, coupled with higher risk of metastasis, which is an exceedingly malignant characteristic of cancer, frequently leading to a high mortality rate. Unfortunately, there is currently no specific and effective marker to predict and treat metastasis in ESCC. MicroRNAs (miRNAs) are a class of small non-coding RNA molecules, approximately 22 nucleotides in length. miRNAs are vital in modulating gene expression and serve pivotal regulatory roles in the occurrence, progression, and prognosis of cancer. Here, we have examined the literature to highlight the intimate correlations between miRNAs and ESCC metastasis, and show that ESCC metastasis is predominantly regulated or regulated by genetic and epigenetic factors. This review proposes a potential role for miRNAs as diagnostic and therapeutic biomarkers for metastasis in ESCC metastasis, with the ultimate aim of reducing the mortality rate among patients with ESCC.
Collapse
Affiliation(s)
- Qi-Ying Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Feng Jin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Zhong-Yu Wang
- Department of Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Bing-Jie Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Wen-Bo Cao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Zhi-Yan Sun
- Division of Special Service, Department of Basic Oncology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Sai-Jun Mo
- Department of Basic Science of Oncology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
5
|
Saviana M, Le P, Micalo L, Del Valle-Morales D, Romano G, Acunzo M, Li H, Nana-Sinkam P. Crosstalk between miRNAs and DNA Methylation in Cancer. Genes (Basel) 2023; 14:1075. [PMID: 37239435 PMCID: PMC10217889 DOI: 10.3390/genes14051075] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
miRNAs are some of the most well-characterized regulators of gene expression. Integral to several physiological processes, their aberrant expression often drives the pathogenesis of both benign and malignant diseases. Similarly, DNA methylation represents an epigenetic modification influencing transcription and playing a critical role in silencing numerous genes. The silencing of tumor suppressor genes through DNA methylation has been reported in many types of cancer and is associated with tumor development and progression. A growing body of literature has described the crosstalk between DNA methylation and miRNAs as an additional layer in the regulation of gene expression. Methylation in miRNA promoter regions inhibits its transcription, while miRNAs can target transcripts and subsequently regulate the proteins responsible for DNA methylation. Such relationships between miRNA and DNA methylation serve an important regulatory role in several tumor types and highlight a novel avenue for potential therapeutic targets. In this review, we discuss the crosstalk between DNA methylation and miRNA expression in the pathogenesis of cancer and describe how miRNAs influence DNA methylation and, conversely, how methylation impacts the expression of miRNAs. Finally, we address how these epigenetic modifications may be leveraged as biomarkers in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Patrick Nana-Sinkam
- Department of Internal Medicine, Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, 1250 E. Marshall Street, Richmond, VA 23298, USA
| |
Collapse
|
6
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
7
|
Duan XK, Sun YX, Wang HY, Xu YY, Fan SZ, Tian JY, Yu Y, Zhao YY, Jiang YL. miR-124 is upregulated in diabetic mice and inhibits proliferation and promotes apoptosis of high-glucose-induced β-cells by targeting EZH2. World J Diabetes 2023; 14:209-221. [PMID: 37035229 PMCID: PMC10075033 DOI: 10.4239/wjd.v14.i3.209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/05/2023] [Accepted: 02/15/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Diabetes is a chronic metabolic disease, and a variety of miRNA are involved in the occurrence and development of diabetes. In clinical studies, miR-124 is highly expressed in the serum of patients with diabetes and in pancreatic islet β-cells. However, few reports exist concerning the role and mechanism of action of miR-124 in diabetes.
AIM To investigate the expression of miR-124 in diabetic mice and the potential mechanism of action in islet β-cells.
METHODS The expression levels of miR-124 and enhancer of zeste homolog 2 (EZH2) in pancreatic tissues of diabetic mice were detected. The targeted relationship between miR-124 and EZH2 was predicted by Targetscan software and verified by a double luciferase reporter assay. Mouse islet β-cells Min6 were grown in a high glucose (HG) medium to mimic a diabetes model. The insulin secretion, proliferation, cell cycle and apoptosis of HG-induced Min6 cells were detected after interference of miR-124a and/or EZH2.
RESULTS The expression of miR-124 was upregulated and EZH2 was downregulated in the pancreatic tissue of diabetic mice compared with control mice, and the expression of miR-124 was negatively correlated with that of EZH2. miR-124 was highly expressed in HG-induced Min6 cells. Inhibition of miR-124 promoted insulin secretion and cell proliferation, induced the transition from the G0/G1 phase to the S phase of the cell cycle, and inhibited cell apoptosis in HG-induced Min6 cells. EZH2 was one of the targets of miR-124. Co-transfection of miR-124 inhibitor and siRNA-EZH2 could reverse the effects of the miR-124 inhibitor in HG-induced Min6 cells.
CONCLUSION miR-124 is highly expressed in diabetic mice and HG-induced Min6 cells and regulates insulin secretion, proliferation and apoptosis of islet β-cells by targeting EZH2.
Collapse
Affiliation(s)
- Xiao-Kai Duan
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Yong-Xiang Sun
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Hong-Yun Wang
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Yan-Yan Xu
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Shi-Zhen Fan
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Jin-Ya Tian
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Yong Yu
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Yan-Yun Zhao
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| | - Yan-Li Jiang
- Department of General Practice, Zhengzhou First People’s Hospital, Zhengzhou 450000, Henan Province, China
| |
Collapse
|
8
|
Thomaidou AC, Batsaki P, Adamaki M, Goulielmaki M, Baxevanis CN, Zoumpourlis V, Fortis SP. Promising Biomarkers in Head and Neck Cancer: The Most Clinically Important miRNAs. Int J Mol Sci 2022; 23:ijms23158257. [PMID: 35897831 PMCID: PMC9367895 DOI: 10.3390/ijms23158257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Head and neck cancers (HNCs) comprise a heterogeneous group of tumors that extend from the oral cavity to the upper gastrointestinal tract. The principal etiologic factors for oral tumors include tobacco smoking and alcohol consumption, while human papillomavirus (HPV) infections have been accused of a high incidence of pharyngeal tumors. Accordingly, HPV detection has been extensively used to categorize carcinomas of the head and neck. The diverse nature of HNC highlights the necessity for novel, sensitive, and precise biomarkers for the prompt diagnosis of the disease, its successful monitoring, and the timely prognosis of patient clinical outcomes. In this context, the identification of certain microRNAs (miRNAs) and/or the detection of alterations in their expression patterns, in a variety of somatic fluids and tissues, could serve as valuable biomarkers for precision oncology. In the present review, we summarize some of the most frequently studied miRNAs (including miR-21, -375, -99, -34a, -200, -31, -125a/b, -196a/b, -9, -181a, -155, -146a, -23a, -16, -29, and let-7), their role as biomarkers, and their implication in HNC pathogenesis. Moreover, we designate the potential of given miRNAs and miRNA signatures as novel diagnostic and prognostic tools for successful patient stratification. Finally, we discuss the currently ongoing clinical trials that aim to identify the diagnostic, prognostic, or therapeutic utility of miRNAs in HNC.
Collapse
Affiliation(s)
- Arsinoe C. Thomaidou
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (A.C.T.); (M.A.)
| | - Panagiota Batsaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (P.B.); (M.G.); (C.N.B.)
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (A.C.T.); (M.A.)
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (P.B.); (M.G.); (C.N.B.)
| | - Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (P.B.); (M.G.); (C.N.B.)
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (A.C.T.); (M.A.)
- Correspondence: (V.Z.); (S.P.F.); Tel.: +30-210-727-3730 (V.Z.); +30-210-640-9462 (S.P.F.)
| | - Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (P.B.); (M.G.); (C.N.B.)
- Correspondence: (V.Z.); (S.P.F.); Tel.: +30-210-727-3730 (V.Z.); +30-210-640-9462 (S.P.F.)
| |
Collapse
|
9
|
Histone Modifications and Non-Coding RNAs: Mutual Epigenetic Regulation and Role in Pathogenesis. Int J Mol Sci 2022; 23:ijms23105801. [PMID: 35628612 PMCID: PMC9146199 DOI: 10.3390/ijms23105801] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 12/07/2022] Open
Abstract
In the last few years, more and more scientists have suggested and confirmed that epigenetic regulators are tightly connected and form a comprehensive network of regulatory pathways and feedback loops. This is particularly interesting for a better understanding of processes that occur in the development and progression of various diseases. Appearing on the preclinical stages of diseases, epigenetic aberrations may be prominent biomarkers. Being dynamic and reversible, epigenetic modifications could become targets for a novel option for therapy. Therefore, in this review, we are focusing on histone modifications and ncRNAs, their mutual regulation, role in cellular processes and potential clinical application.
Collapse
|
10
|
Yao D, Lin S, Chen S, Wang Z. circHIPK3 regulates cell proliferation and migration by sponging microRNA-124 and regulating serine/threonine kinase 3 expression in esophageal squamous cell carcinoma. Bioengineered 2022; 13:9767-9780. [PMID: 35443871 PMCID: PMC9161938 DOI: 10.1080/21655979.2022.2060776] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Circular RNAs (circRNAs) are a type of important non-coding RNAs that widely involve in the physiological and pathophysiological process. Recent research has established a link between circHIPK3 and the malignant activity of cancer cells. However, circHIPK3’ role in esophageal squamous cell carcinoma (ESCC) still needs more focus. To determine the prognostic value of circHIPK3 in patients with ESCC, the expression of circHIPK3 was quantified in 32 pairs of ESCC using real-time polymerase chain reaction (RT-qPCR). Then, the correlation between circHIPK3 expression and clinical characteristics of patients was also analyzed. The function of circHIPK3 in the development of ESCC was investigated using cell biology studies and bioinformatics. The results showed that the expression of circHIPK3 was considerably higher in tumor tissues from ESCC patients than that of adjacent tissues, which was associated with a poor prognosis. Additionally, silencing of circHIPK3 expression retarded esophageal cancer cell proliferation, migration and epithelial-mesenchymal transition (EMT) in vitro, as well as the growth in vivo. Mechanistically, we discovered that circHIPK3 behaved like a sponge, absorbing microRNA-124 (miR-124) and promoting serine/threonine kinase 3 (AKT3) expression. Our findings indicate that circHIPK3 acts as an oncogene in ESCC and that the circHIPK3-AKT3 axis may be a therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Da Yao
- Department of Thoracic Surgery, Shenzhen Second People's Hospital. The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Shengcheng Lin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and PeKing Union Medical College, Shenzhen, Guangdong, China
| | | | - Zhe Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and PeKing Union Medical College, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Long noncoding RNA TTTY15 promotes growth and metastasis of esophageal squamous cell carcinoma by sponging microRNA-337-3p to upregulate the expression of JAK2. Anticancer Drugs 2021; 31:1038-1045. [PMID: 32868648 DOI: 10.1097/cad.0000000000000960] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Long noncoding RNA (lncRNA) testis-specific transcript, Y-linked 15 (TTTY15) plays an important regulatory role in prostate cancer, but its role in esophageal squamous cell carcinoma (ESCC) remains unclear. This study aimed to explore the expression pattern, biological function and underlying mechanism of TTTY15 in ESCC. Quantitative real-time PCR (qRT-PCR) was used to detect the expression of TTTY15 and microRNA (miR)-337-3p in ESCC tissues and cell lines. Cell counting kit-8 method was used to detect the proliferation of ESCC cells. Transwell method was used to determine the migration and invasion of ESCC cells. Luciferase reporter assay was used to verify the interaction between TTTY15 and miR-337-3p. Western blot was used to analyze the effects of TTTY15 and miR-337-3p on Janus kinase 2 (JAK2) expression. In the present study, we demonstrated that the expression level of TTTY15 was significantly upregulated in ESCC tissues, while the expression of miR-337-3p was downregulated. In ESCC samples, the expression levels of TTTY15 and miR-337-3p were negatively correlated. TTTY15 knockdown could significantly reduce the proliferation, migration and invasion of ESCC cells, and miR-337-3p mimics had similar effects. In addition, overexpression of TTTY15 inhibited miR-337-3p by binding with it. TTTY15 could indirectly modulate JAK2, and overexpression of TTTY15 could reverse the inhibitory effects of miR-337-3p on malignant phenotypes of ESCC cells. In conclusion, TTTY15 plays an oncogenic role in ESCC by targeting miR-337-3p/JAK2 axis.
Collapse
|
12
|
Miller SA, Damle M, Kim J, Kingston RE. Full methylation of H3K27 by PRC2 is dispensable for initial embryoid body formation but required to maintain differentiated cell identity. Development 2021; 148:dev196329. [PMID: 33688077 PMCID: PMC8077505 DOI: 10.1242/dev.196329] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
Polycomb repressive complex 2 (PRC2) catalyzes methylation of histone H3 on lysine 27 and is required for normal development of complex eukaryotes. The nature of that requirement is not clear. H3K27me3 is associated with repressed genes, but the modification is not sufficient to induce repression and, in some instances, is not required. We blocked full methylation of H3K27 with both a small molecule inhibitor, GSK343, and by introducing a point mutation into EZH2, the catalytic subunit of PRC2, in the mouse CJ7 cell line. Cells with substantively decreased H3K27 methylation differentiate into embryoid bodies, which contrasts with EZH2 null cells. PRC2 targets had varied requirements for H3K27me3, with a subset that maintained normal levels of repression in the absence of methylation. The primary cellular phenotype of blocked H3K27 methylation was an inability of altered cells to maintain a differentiated state when challenged. This phenotype was determined by H3K27 methylation in embryonic stem cells through the first 4 days of differentiation. Full H3K27 methylation therefore was not necessary for formation of differentiated cell states during embryoid body formation but was required to maintain a stable differentiated state.
Collapse
Affiliation(s)
- Sara A. Miller
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Manashree Damle
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jongmin Kim
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Robert E. Kingston
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Li Q, Li G, Liu C, Chen N, Deng B, Xie Y. Cell Differentiation Agent-2 (CDA-2) Inhibits the Growth and Migration of Saos-2 Cells via miR-124/MAPK1. Cancer Manag Res 2020; 12:4541-4548. [PMID: 32606947 PMCID: PMC7304673 DOI: 10.2147/cmar.s248851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND CDA-2 (cell differentiation agent 2), isolated from healthy human urine, exerts antitumor effects in multiple types of cancer cells. However, its role in osteosarcoma has not been studied. METHODS The MTT assay was used to examine the cell proliferation rate. A colony formation assay was used to examine cell growth. The Transwell assay was used to examine cell migration ability. A real-time PCR assay was used to examine the expression levels of miR-124 and MAPK1. A Western blot assay was used to examine protein expression levels. MAPK1 was selected as a possible target of miR-124, and the targeting relationship was examined by a luciferase reporter assay. RESULTS We revealed that CDA-2 decreased the growth, migration and invasion ability of the osteosarcoma cell line Saos-2. Further study revealed that CDA-2 elevated the expression level of miR-124. MAPK1 was identified as a downstream target of miR-124. Knockdown of miR-124 or overexpression of MAPK1 counteracted CDA-2's effects on cell growth and invasion. CONCLUSION Our data revealed that the miR-124/MAPK1 axis mediated CDA-2's function in Saos-2 cells. CDA-2 can be used as a new treatment strategy for osteosarcoma.
Collapse
Affiliation(s)
- Quanxiu Li
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China,Correspondence: Quanxiu Li Email
| | - Guangchun Li
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Changyi Liu
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Na Chen
- Department of Pathology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Bangyu Deng
- Department of Oncology, Affiliated Tumor Hospital of Guangzhou University, Guangzhou, Guangdong, People’s Republic of China
| | - Youke Xie
- Department of Oncology, Hospital of Ruikang Affiliated to Guangxi University of Chinese Medicine, Guangxi, People’s Republic of China
| |
Collapse
|
14
|
Li Z, Qin X, Bian W, Li Y, Shan B, Yao Z, Li S. Exosomal lncRNA ZFAS1 regulates esophageal squamous cell carcinoma cell proliferation, invasion, migration and apoptosis via microRNA-124/STAT3 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:477. [PMID: 31775815 PMCID: PMC6882153 DOI: 10.1186/s13046-019-1473-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Background In recent years, long non-coding RNAs (lncRNAs) are of great importance in development of different types of tumors, while the function of lncRNA ZFAS1 is rarely discussed in esophageal squamous cell carcinoma (ESCC). Therefore, we performed this study to explore the expression of exosomal lncRNA ZFAS1 and its molecular mechanism on ESCC progression. Methods Expression of ZFAS1 and miR-124 in ESCC tissues was detected. LncRNA ZFAS1 was silenced to detect its function in the biological functions of ESCC cells. A stable donor and recipient culture model was established. Eca109 cells transfected with overexpressed and low expressed ZFAS1 plasmid and miR-124 inhibitor labeled by Cy3 were the donor cells, and then co-cultured with recipient cells to observe the transmission of Cy3-ZFAS1 between donor cells and recipient cells. The changes of cell proliferation, apoptosis, invasion, and migration in recipient cells were detected. The in vivo experiment was conducted for verifying the in vitro results. Results LncRNA ZFAS1 was upregulated and miR-124 was down-regulated in ESCC tissues. Silencing of ZFAS1 contributed to suppressed proliferation, migration, invasion and tumor growth in vitro and induced apoptosis of ESCC cells. LncRNA ZFAS1 was considered to be a competing endogenous RNA to regulate miR-124, thereby elevating STAT3 expression. Exosomes shuttled ZFAS1 stimulated proliferation, migration and invasion of ESCC cells and restricted their apoptosis with increased STAT3 and declined miR-124. Furthermore, in vivo experiment suggested that elevated ZFAS1-exo promoted tumor growth in nude mice. Conclusion This study highlights that exosomal ZFAS1 promotes the proliferation, migration and invasion of ESCC cells and inhibits their apoptosis by upregulating STAT3 and downregulating miR-124, thereby resulting in the development of tumorigenesis of ESCC.
Collapse
Affiliation(s)
- Zhirong Li
- Clinical Laboratory Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Xuebo Qin
- Department of Thoracic Surgery, Hebei Provincial Chest Hospital, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Wei Bian
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Yishuai Li
- Department of Thoracic Surgery, Hebei Provincial Chest Hospital, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Zhimeng Yao
- Institute of Precision Medicine and Pathology, Jinan University, Guangzhou, 510000, Guangdong Province, People's Republic of China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Shijiazhuang, 050000, Hebei Province, People's Republic of China.
| |
Collapse
|
15
|
Lin T, Dai Y, Guo X, Chen W, Zhao J, Cao L, Wu Z. Silencing Of hsa_circ_0008450 Represses Hepatocellular Carcinoma Progression Through Regulation Of microRNA-214-3p/EZH2 Axis. Cancer Manag Res 2019; 11:9133-9143. [PMID: 31695501 PMCID: PMC6817349 DOI: 10.2147/cmar.s222716] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose Circular RNA (circRNA) hsa_circ_0008450 has been shown to be up-regulated in hepatocellular carcinoma (HCC). However, the functional role of hsa_circ_0008450 and its molecular mechanism are still unknown. Patients and methods We used qRT-PCR and Western blot to examine the expression levels of hsa_circ_0008450, microRNA-214-3p (miR-214-3p), and enhancer of zeste homolog 2 (EZH2) protein. CCK8 assay and wound healing assay were used to detect cell viability and cell migration capability. Cell apoptosis was assessed by flow cytometry. Luciferase reporter assay was used to explore the interaction among hsa_circ_0008450, miR-214-3p, and EZH2. Results hsa_circ_0008450 was significantly increased in HCC tissues and cells. Furthermore, knockdown of hsa_circ_0008450 in HCC cells inhibited cell proliferation, invasion, and migration. Mechanically, hsa_circ_0008450 promoted the expression of EZH2 protein through sponging miR-214-3p. Knockdown of circ_0008450 suppressed tumorigenesis of HCC cells in vivo. Conclusion Knockdown of hsa_circ_0008450 inhibits HCC progression by regulating miR-214-3p/EZH2 axis. This study suggests that hsa_circ_0008450 may serve as a novel target for the treatment of HCC.
Collapse
Affiliation(s)
- Tianyu Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| | - Yi Dai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| | - Xinli Guo
- Department of Operating Room, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| | - Wenchao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| | - Jiangang Zhao
- Department of General, Visceral and Tumor Surgery, University Hospital Cologne, Cologne 50934, Germany
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| | - Zhengrong Wu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, People's Republic of China
| |
Collapse
|