1
|
Wang Z, Liu H. Roles of Lysine Methylation in Glucose and Lipid Metabolism: Functions, Regulatory Mechanisms, and Therapeutic Implications. Biomolecules 2024; 14:862. [PMID: 39062577 PMCID: PMC11274642 DOI: 10.3390/biom14070862] [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: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Glucose and lipid metabolism are essential energy sources for the body. Dysregulation in these metabolic pathways is a significant risk factor for numerous acute and chronic diseases, including type 2 diabetes (T2DM), Alzheimer's disease (AD), obesity, and cancer. Post-translational modifications (PTMs), which regulate protein structure, localization, function, and activity, play a crucial role in managing cellular glucose and lipid metabolism. Among these PTMs, lysine methylation stands out as a key dynamic modification vital for the epigenetic regulation of gene transcription. Emerging evidence indicates that lysine methylation significantly impacts glucose and lipid metabolism by modifying key enzymes and proteins. This review summarizes the current understanding of lysine methylation's role and regulatory mechanisms in glucose and lipid metabolism. We highlight the involvement of methyltransferases (KMTs) and demethylases (KDMs) in generating abnormal methylation signals affecting these metabolic pathways. Additionally, we discuss the chemical biology and pharmacology of KMT and KDM inhibitors and targeted protein degraders, emphasizing their clinical implications for diseases such as diabetes, obesity, neurodegenerative disorders, and cancers. This review suggests that targeting lysine methylation in glucose and lipid metabolism could be an ideal therapeutic strategy for treating these diseases.
Collapse
Affiliation(s)
| | - Huadong Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China;
| |
Collapse
|
2
|
Yang Y, Xie Q, Hu C, Xu J, Chen L, Li Y, Luo C. F-box proteins and gastric cancer: an update from functional and regulatory mechanism to therapeutic clinical prospects. Int J Med Sci 2024; 21:1575-1588. [PMID: 38903918 PMCID: PMC11186432 DOI: 10.7150/ijms.91584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy characterized by significant morbidity and mortality, yet its underlying pathogenesis remains elusive. The etiology of GC is multifaceted, involving the activation of oncogenes and the inactivation of antioncogenes. The ubiquitin-proteasome system (UPS), responsible for protein degradation and the regulation of physiological and pathological processes, emerges as a pivotal player in GC development. Specifically, the F-box protein (FBP), an integral component of the SKP1-Cullin1-F-box protein (SCF) E3 ligase complex within the UPS, has garnered attention for its prominent role in carcinogenesis, tumor progression, and drug resistance. Dysregulation of several FBPs has recently been observed in GC, underscoring their significance in disease progression. This comprehensive review aims to elucidate the distinctive characteristics of FBPs involved in GC, encompassing their impact on cell proliferation, apoptosis, invasive metastasis, and chemoresistance. Furthermore, we delve into the emerging role of FBPs as downstream target proteins of non-coding RNAs(ncRNAs) in the regulation of gastric carcinogenesis, outlining the potential utility of FBPs as direct therapeutic targets or advanced therapies for GC.
Collapse
Affiliation(s)
- Yanzhen Yang
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Qu Xie
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Jingli Xu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Lei Chen
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Yuan Li
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| | - Cong Luo
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310005, China
| |
Collapse
|
3
|
Zhang C, Pan G, Qin JJ. Role of F-box proteins in human upper gastrointestinal tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189035. [PMID: 38049014 DOI: 10.1016/j.bbcan.2023.189035] [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: 08/23/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Protein ubiquitination and degradation is an essential physiological process in almost all organisms. As the key participants in this process, the E3 ubiquitin ligases have been widely studied and recognized. F-box proteins, a crucial component of E3 ubiquitin ligases that regulates diverse biological functions, including cell differentiation, proliferation, migration, and apoptosis by facilitating the degradation of substrate proteins. Currently, there is an increasing focus on studying the role of F-box proteins in cancer. In this review, we present a comprehensive overview of the significant contributions of F-box proteins to the development of upper gastrointestinal tumors, highlighting their dual roles as both carcinogens and tumor suppressors. We delve into the molecular mechanisms underlying the involvement of F-box proteins in upper gastrointestinal tumors, exploring their interactions with specific substrates and their cross-talks with other key signaling pathways. Furthermore, we discuss the implications of F-box proteins in radiotherapy resistance in the upper gastrointestinal tract, emphasizing their potential as clinical therapeutic and prognostic targets. Overall, this review provides an up-to-date understanding of the intricate involvement of F-box proteins in human upper gastrointestinal tumors, offering valuable insights for the identification of prognostic markers and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Che Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guangzhao Pan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jiang-Jiang Qin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
4
|
Chen X, Wei C, Huang L, Syrigos K, Li Y, Li P. Non-coding RNAs regulate mitochondrial dynamics in the development of gastric cancer. Front Mol Biosci 2023; 10:1107651. [PMID: 36714260 PMCID: PMC9877238 DOI: 10.3389/fmolb.2023.1107651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer (GC) is a malignant cancer that reduces life expectancy worldwide. Although treatment strategies have improved, patients with GC still have poor prognoses. Hence, it is necessary to understand the molecular mechanisms of GC and to find new therapeutic targets. Mitochondrial dynamics and mitochondrial dysfunction are associated with cancer cell growth and progression. Numerous studies have reported that non-coding RNAs (ncRNAs) can participate in the occurrence and development of GC by regulating mitochondrial dynamics. Elucidating the crosstalk between ncRNAs and mitochondria would be helpful in preventing and treating GC. Herein, we review and summarize the functions of oncogenes and tumor suppressors in suppressing ncRNAs and regulating mitochondrial dynamics in GC tumor growth, proliferation, invasion and metastasis. This review provides new insights into the pathogenesis of and intervention for GC.
Collapse
Affiliation(s)
- Xiatian Chen
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China,School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chuang Wei
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China,School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liting Huang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China,School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Yuzhen Li
- Basic Medical Department, Graduate School, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yuzhen Li, ; Peifeng Li,
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China,*Correspondence: Yuzhen Li, ; Peifeng Li,
| |
Collapse
|
5
|
Usman M, Beilerli A, Sufianov A, Kudryashov V, Ilyasova T, Balaev P, Danilov A, Lu H, Gareev I. Investigations into the impact of non-coding RNA on the sensitivity of gastric cancer to radiotherapy. Front Physiol 2023; 14:1149821. [PMID: 36909247 PMCID: PMC9998927 DOI: 10.3389/fphys.2023.1149821] [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: 01/23/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a newly discovered functional RNA different from messenger RNA, which can participate in regulating the occurrence and development of tumors. More and more research results show that ncRNAs can participate in the regulation of gastric cancer (GC) radiotherapy response, and its mechanism may be related to its effect on DNA damage repair, gastric cancer cell stemness, cell apoptosis, activation of epidermal growth factor receptor signaling pathway, etc. This article summarizes the relevant mechanisms of ncRNAs regulating the response to radiotherapy in gastric cancer, which will be directly important for the introduction of ncRNAs particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) into clinical medicine as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Muhammad Usman
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, Tyumen, Russia
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Valentin Kudryashov
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Pavel Balaev
- Department of Oncology and Radiology, Ural State Medical University, Yekaterinburg, Russia
| | - Andrei Danilov
- Department of Clinical Pharmacology, Smolensk State Medical University, Smolensk, Russia
| | - Hong Lu
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Ilgiz Gareev
- Educational and Scientific Institute of Neurosurgery, Рeoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
6
|
Yang Q, Chen Y, Guo R, Dai Y, Tang L, Zhao Y, Wu X, Li M, Du F, Shen J, Yi T, Xiao Z, Wen Q. Interaction of ncRNA and Epigenetic Modifications in Gastric Cancer: Focus on Histone Modification. Front Oncol 2022; 11:822745. [PMID: 35155211 PMCID: PMC8826423 DOI: 10.3389/fonc.2021.822745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer has developed as a very common gastrointestinal tumors, with recent effective advancements in the diagnosis and treatment of early gastric cancer. However, the prognosis for gastric cancer remains poor. As a result, there is in sore need of better understanding the mechanisms of gastric cancer development and progression to improve existing diagnostic and treatment options. In recent years, epigenetics has been recognized as an important contributor on tumor progression. Epigenetic changes in cancer include chromatin remodeling, DNA methylation and histone modifications. An increasing number of studies demonstrated that noncoding RNAs (ncRNAs) are associated with epigenetic changes in gastric cancer. Herein, we describe the molecular interactions of histone modifications and ncRNAs in epigenetics. We focus on ncRNA-mediated histone modifications of gene expression associated with tumorigenesis and progression in gastric cancer. This molecular mechanism will contribute to our deeper understanding of gastric carcinogenesis and progression, thus providing innovations in gastric cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Qingfan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yu Chen
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Rui Guo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Yalan Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Liyao Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Yueshui Zhao
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Xu Wu
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Mingxing Li
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Fukuan Du
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Jing Shen
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Tao Yi
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Zhangang Xiao
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| |
Collapse
|
7
|
Epi-miRNAs: Regulators of the Histone Modification Machinery in Human Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4889807. [PMID: 35087589 PMCID: PMC8789461 DOI: 10.1155/2022/4889807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of death and disability worldwide. Epigenetic deregulation is one of the most critical mechanisms in carcinogenesis and can be classified into effects on DNA methylation and histone modification. MicroRNAs are small noncoding RNAs involved in fine-tuning their target genes after transcription. Various microRNAs control the expression of histone modifiers and are involved in a variety of cancers. Therefore, overexpression or downregulation of microRNAs can alter cell fate and cause malignancies. In this review, we discuss the role of microRNAs in regulating the histone modification machinery in various cancers, with a focus on the histone-modifying enzymes such as acetylases, deacetylases, methyltransferases, demethylases, kinases, phosphatases, desumoylases, ubiquitinases, and deubiquitinases. Understanding of microRNA-related aberrations underlying histone modifiers in pathogenesis of different cancers can help identify novel therapeutic targets or early detection approaches that allow better management of patients or monitoring of treatment response.
Collapse
|
8
|
Gulay KCM, Aoshima K, Kim S, Kitaguchi R, Kobayashi A, Kimura T. The expression of histone lysine demethylase 2B in canine hemangiosarcoma is associated with disease progression. Vet Comp Oncol 2021; 20:529-534. [DOI: 10.1111/vco.12796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Kevin Christian M. Gulay
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Sangho Kim
- Laboratory of Veterinary Surgery, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Ryusei Kitaguchi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| |
Collapse
|
9
|
Gulay KCM, Aoshima K, Shibata Y, Yasui H, Yan Q, Kobayashi A, Kimura T. KDM2B promotes cell viability by enhancing DNA damage response in canine hemangiosarcoma. J Genet Genomics 2021; 48:618-630. [PMID: 34023294 DOI: 10.1016/j.jgg.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/16/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022]
Abstract
Epigenetic regulators have been implicated in tumorigenesis of many types of cancer; however, their roles in endothelial cell cancers such as canine hemangiosarcoma (HSA) have not been studied. In this study, we find that lysine-specific demethylase 2b (KDM2B) is highly expressed in HSA cell lines compared with normal canine endothelial cells. Silencing of KDM2B in HSA cells results in increased cell death in vitro compared with the scramble control by inducing apoptosis through the inactivation of the DNA repair pathways and accumulation of DNA damage. Similarly, doxycycline-induced KDM2B silencing in tumor xenografts results in decreased tumor sizes compared with the control. Furthermore, KDM2B is also highly expressed in clinical cases of HSA. We hypothesize that pharmacological KDM2B inhibition can also induce HSA cell death and can be used as an alternative treatment for HSA. We treat HSA cells with GSK-J4, a histone demethylase inhibitor, and find that GSK-J4 treatment also induces apoptosis and cell death. In addition, GSK-J4 treatment decreases tumor size. Therefore, we demonstrate that KDM2B acts as an oncogene in HSA by enhancing the DNA damage response. Moreover, we show that histone demethylase inhibitor GSK-J4 can be used as a therapeutic alternative to doxorubicin for HSA treatment.
Collapse
Affiliation(s)
- Kevin Christian Montecillo Gulay
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan.
| | - Yuki Shibata
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818 Japan
| |
Collapse
|
10
|
Sui Y, Liu J, Zhang J, Zheng Z, Wang Z, Jia Z, Meng Z. Expression and Gene Regulation Network of Adenosine Receptor A2B in Lung Adenocarcinoma: A Potential Diagnostic and Prognostic Biomarker. Front Mol Biosci 2021; 8:663011. [PMID: 34350210 PMCID: PMC8326519 DOI: 10.3389/fmolb.2021.663011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenosinereceptor A2B (ADORA2B) encodes a protein belonging to the G protein–coupled receptor superfamily. Abnormal expression of ADORA2B may play a pathophysiological role in some human cancers. We investigated whether ADORA2B is a potential diagnostic and prognostic biomarker for lung adenocarcinoma (LUAD). The expression, various mutations, copy number variations, mRNA expression levels, and related network signaling pathways of ADORA2B were analyzed using bioinformatics-related websites, including Oncomine, UALCAN, cBioPortal, GeneMANIA, LinkedOmics, KM Plotter, and TIMER. We found that ADORA2B was overexpressed and amplified in LUAD, and a high ADORA2B expression predicted a poor prognosis for LUAD patients. Pathway analyses of ADORA2B in LUAD revealed ADORA2B-correlated signaling pathways, and the expression level of ADORA2B was associated with immune cell infiltration. Furthermore, ADORA2B mRNA and protein levels were significantly higher in human LUAD cell lines (A549 cells and NCl-H1299 cells) than in normal human bronchial epithelial (HBE) cells, and the transcript levels of genes positively or negatively correlated with ADORA2B were consistent and statistically significant. siRNA transfection experiments and functional experiments further confirmed these results. In vitro results were also consistent with those of bioinformatics analysis. Our findings provide a foundation for studying the role of ADORA2B in tumorigenesis and support the development of new drug targets for LUAD.
Collapse
Affiliation(s)
- Yutong Sui
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jiayin Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jing Zhang
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zena Zheng
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Ziwei Wang
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhenghu Jia
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Yang Y, Li S, Li B, Li Y, Xia K, Aman S, Yang Y, Ahmad B, Zhao B, Wu H. FBXL10 promotes ERRα protein stability and proliferation of breast cancer cells by enhancing the mono-ubiquitylation of ERRα. Cancer Lett 2021; 502:108-119. [PMID: 33450359 DOI: 10.1016/j.canlet.2021.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 01/25/2023]
Abstract
The underlying mechanism of orphan nuclear receptor estrogen-related receptor α (ERRα) in breast cancer was investigated by identifying its interaction partners using mass spectrometry. F-box and leucine-rich repeat protein 10 (FBXL10), which modulates various physiological processes, may interact with ERRα in breast cancer. Here, we investigated the interaction between FBXL10 and ERRα, and their protein expression and correlation in breast cancer. Mechanical studies revealed that FBXL10 stabilized ERRα protein levels by reducing its poly-ubiquitylation and promoting its mono-ubiquitylation. The reporter gene assay and examination of ERRα target genes validated the increased transcriptional activity of ERRα due to its increased protein levels by FBXL10. FBXL10 also increased ERRα enrichment at the promoter region of its target genes. Functionally, FBXL10 facilitated the ERRα/peroxisome proliferator-activated receptor gamma coactivator 1 β (PGC1β)-mediated proliferation and tumorigenesis of breast cancer cells in vitro and in vivo. Our results uncovered a molecular mechanism linking the mono-ubiquitylation and protein stability of ERRα to functional interaction with FBXL10. Moreover, a novel regulatory axis of FBXL10 and ERRα regulating the proliferation and tumorigenesis of breast cancer cells was established.
Collapse
Affiliation(s)
- Yangyang Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Bowen Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Yanan Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Kangkai Xia
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Sattout Aman
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Yuxi Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Bashir Ahmad
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Binggong Zhao
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China.
| |
Collapse
|
12
|
Xia R, Tang H, Shen J, Xu S, Liang Y, Zhang Y, Gong X, Min Y, Zhang D, Tao C, Wang S, Zhang Y, Yang J, Wang C. Prognostic value of a novel glycolysis-related gene expression signature for gastrointestinal cancer in the Asian population. Cancer Cell Int 2021; 21:154. [PMID: 33663535 PMCID: PMC7934443 DOI: 10.1186/s12935-021-01857-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Globally, gastrointestinal (GI) cancer is one of the most prevalent malignant tumors. However, studies have not established glycolysis-related gene signatures that can be used to construct accurate prognostic models for GI cancers in the Asian population. Herein, we aimed at establishing a novel glycolysis-related gene expression signature to predict the prognosis of GI cancers. METHODS First, we evaluated the mRNA expression profiles and the corresponding clinical data of 296 Asian GI cancer patients in The Cancer Genome Atlas (TCGA) database (TCGA-LIHC, TCGA-STAD, TCGA-ESCA, TCGA-PAAD, TCGA-COAD, TCGA-CHOL and TCGA-READ). Differentially expressed mRNAs between GI tumors and normal tissues were investigated. Gene Set Enrichment Analysis (GSEA) was performed to identify glycolysis-related genes. Then, univariate, LASSO regression and multivariate Cox regression analyses were performed to establish a key prognostic glycolysis-related gene expression signature. The Kaplan-Meier and receiver operating characteristic (ROC) curves were used to evaluate the efficiency and accuracy of survival prediction. Finally, a risk score to predict the prognosis of GI cancers was calculated and validated using the TCGA data sets. Furthermore, this risk score was verified in two Gene Expression Omnibus (GEO) data sets (GSE116174 and GSE84433) and in 28 pairs of tissue samples. RESULTS Prognosis-related genes (NUP85, HAX1, GNPDA1, HDLBP and GPD1) among the differentially expressed glycolysis-related genes were screened and identified. The five-gene expression signature was used to assign patients into high- and low-risk groups (p < 0.05) and it showed a satisfactory prognostic value for overall survival (OS, p = 6.383 × 10-6). The ROC curve analysis revealed that this model has a high sensitivity and specificity (0.757 at 5 years). Besides, stratification analysis showed that the prognostic value of the five-gene signature was independent of other clinical characteristics, and it could markedly discriminate between GI tumor tissues and normal tissues. Finally, the expression levels of the five prognosis-related genes in the clinical tissue samples were consistent with the results from the TCGA data sets. CONCLUSIONS Based on the five glycolysis-related genes (NUP85, HAX1, GNPDA1, HDLBP and GPD1), and in combination with clinical characteristics, this model can independently predict the OS of GI cancers in Asian patients.
Collapse
Affiliation(s)
- Rong Xia
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Hua Tang
- Department of General Surgery, Tongling People's Hospital, 468 Bijiashan Road, Tongling, Anhui Province, 244000, People's Republic of China
| | - Jiemiao Shen
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Shuyu Xu
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yinyin Liang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yuxin Zhang
- The First Clinical Medical College of Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Xing Gong
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yue Min
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Di Zhang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Chenzhe Tao
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Shoulin Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yi Zhang
- Department of Colorectal Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, People's Republic of China.
| | - Jinyou Yang
- Department of Clinical Medicine and Rehabilitation, Jiangsu College of Nursing, 9 Keji Road, Huai'an, 223005, People's Republic of China.
| | - Chao Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China. .,State Key Lab of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
13
|
DLX6 Antisense RNA 1 Modulates Glucose Metabolism and Cell Growth in Gastric Cancer by Targeting microRNA-4290. Dig Dis Sci 2021; 66:460-473. [PMID: 32239379 DOI: 10.1007/s10620-020-06223-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/18/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most commonly diagnosed malignancy worldwide. DLX6 antisense RNA 1 (DLX6-AS1) is a long noncoding RNA (lncRNA) that exhibits oncogenic effects on multiple human carcinomas. AIMS This study aimed to investigate the regulatory effect of DLX6-AS1 in GC progression. METHODS The expression of DLX6-AS1 in GC tissues and cell lines was examined. The cell viability, number of clones, and apoptosis, aerobic glycolysis, and mitochondrial respiration was assessed. The effect of DLX6-AS1 on tumor growth in nude mice was also evaluated. RESULTS DLX6-AS1 was overexpressed in GC tissues and cell lines. DLX6-AS1 knockdown by short hairpin RNA (shRNA) significantly inhibited cell viability and colony formation, and induced apoptosis. DLX6-AS1 silencing impaired aerobic glycolysis but stimulated mitochondrial respiration in GC cells. miR-4290 was confirmed as a downstream target of DLX6-AS1, and their expression levels were inversely correlated. GC cells expressing sh-DLX6-AS1 showed significantly lower level of 3-phosphoinositide-dependent protein kinase 1 (PDK1), a target of miR-4290, compared to cells expressing control shRNA. In addition, the suppressed GC cell malignancy upon DLX6-AS1 knockdown could be prominently reversed by PDK1 overexpression. Meanwhile, PDK1 overexpression enhanced aerobic glycolysis but repressed mitochondrial respiration under sh-DLX6-AS1 treatment. Furthermore, DLX6-AS1 knockdown significantly delayed the tumor growth in a mouse xenograft model inoculated with GC cells. CONCLUSIONS LncRNA DLX6-AS1 regulated tumor growth and aerobic glycolysis in GC by targeting miR-4290 and PDK1, suggesting DLX6-AS1 might serve as a novel potential therapeutic target for GC treatment from bench to clinic.
Collapse
|
14
|
Ning MY, Cheng ZL, Zhao J. MicroRNA-448 targets SATB1 to reverse the cisplatin resistance in lung cancer via mediating Wnt/β-catenin signalling pathway. J Biochem 2021; 168:41-51. [PMID: 32525527 DOI: 10.1093/jb/mvaa024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aims to examine whether miR-448 reverses the cisplatin (DDP) resistance in lung cancer by modulating SATB1. QRT-PCR and immunohistochemistry were used to examine the miR-448 and SATB1 expressions in DDP-sensitive and -resistant lung cancer patients. A microarray was used to investigate the cytoplasmic/nucleic ratio (C/N ratios) of genes in A549 cells targeted by miR-448, followed by Dual-luciferase reporter gene assay. A549/DDP cells were transfected with miR-448 mimics/inhibitors with or without SATB1 siRNA followed by MTT assay, Edu staining, flow cytometry, qRT-PCR and western blotting. MiR-448 was lower but SATB1 was increased in DDP-resistant patients and A549/DDP cells. And the patients showed low miR-448 expression or SATB1 positive expression had poor prognosis. SATB1, as a target gene with higher C/N ratios (>1), was found negatively regulated by miR-448. Besides, miR-448 inhibitors increased resistance index of A549/DDP cells, promoted cell proliferation, increased cell distribution in S phrase, declined cell apoptosis and activated Wnt/β-catenin pathway. However, SATB1 siRNA could reverse the above effect caused by miR-448 inhibitors. MiR-448 targeting SATB1 to counteract the DDP resistance of lung cancer cells via Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Mei-Ying Ning
- Department of Pharmacy, Cangzhou Central Hospital, No.16 Xinhua West Road, Yunhe District, Cangzhou 061001, China
| | - Zhao-Lin Cheng
- Department of Pharmacy, Cangzhou People's Hospital, No.7 Qingchi Road, Xinhua District, Cangzhou 061000, China
| | - Jing Zhao
- Department of Pharmacy, Cangzhou Central Hospital, No.16 Xinhua West Road, Yunhe District, Cangzhou 061001, China
| |
Collapse
|
15
|
Weidle UH, Birzele F, Nopora A. microRNAs Promoting Growth of Gastric Cancer Xenografts and Correlation to Clinical Prognosis. Cancer Genomics Proteomics 2021; 18:1-15. [PMID: 33419892 DOI: 10.21873/cgp.20237] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The annual death toll for gastric cancer is in the range of 700,000 worldwide. Even in patients with early-stage gastric cancer recurrence within five years has been observed after surgical resection and following chemotherapy with therapy-resistant features. Therefore, the identification of new targets and treatment modalities for gastric cancer is of paramount importance. In this review we focus on the role of microRNAs with documented efficacy in preclinical xenograft models with respect to growth of human gastric cancer cells. We have identified 31 miRs (-10b, -19a, -19b, -20a, -23a/b, -25, -27a-3p, -92a, -93, -100, -106a, -130a, -135a, -135b-5p, -151-5p, -187, -199-3p, -215, -221-3p, -224, -340a, -382, -421, -425, -487a, -493, -532-3p, -575, -589, -664a-3p) covering 26 different targets which promote growth of gastric cancer cells in vitro and in vivo as xenografts. Five miRs (miRs -10b, 151-5p, -187, 532-3p and -589) additionally have an impact on metastasis. Thirteen of the identified miRs (-19b, -20a/b, -25, -92a, -106a, -135a, -187, -221-3p, -340a, -421, -493, -575 and -589) have clinical impact on worse prognosis in patients.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| |
Collapse
|
16
|
He C, Yang J, Ding J, Li S, Wu H, Zhou F, Teng L, Yang J. MiR-448 targets BLC2 and inhibits the growth of pituitary adenoma cells. Biochem Cell Biol 2020; 98:511-517. [PMID: 32648768 DOI: 10.1139/bcb-2019-0336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
There is an increasing body of evidence indicating the important roles of miRNAs in the progression of pituitary adenoma. Recent studies have shown decreased expression and tumor suppressive function of miR-448 in cancers; however, the clinical significance of miR-448 in pituitary adenoma has remained largely unknown. In our study, we found that miR-448 was down-regulated in pituitary adenoma tissues and cell lines. Overexpression of miR-448 significantly inhibited the proliferation and migration of pituitary adenoma cells. Increased cell apoptosis was also observed with overexpression of miR-448. To further understand the mechanisms behind the regulation of pituitary adenoma by miR-448 in, the targets of miR-448 were predicted using the bioinformatics tools. B cell lymphoma 2 (BCL2) was identified as a target of miR-448. MiR-448 bound the 3'-untranslated region (UTR) of BCL2 and inhibited the expression of BCL2 in pituitary adenoma cells. There was a consistent and significantly negative correlation between the level of miR-448 and BCL2 in pituitary adenoma tissues. When BCL2 was highly expressed, the inhibitory impact of miR-448 on the proliferation and apoptosis of pituitary adenoma cells was significantly inhibited. Collectively, our findings emphasize the significance of the miR-448-BCL2 axis in the development of pituitary adenoma, highlighting the potential therapeutic significance of miR-448 in pituitary adenoma.
Collapse
Affiliation(s)
- Chao He
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Jun Yang
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Jiawang Ding
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Song Li
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Hui Wu
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Fei Zhou
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Lin Teng
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| | - Jian Yang
- Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China.,Institute of Cardiology, China Three Gorges University, Yichang, Hubei 443003, P.R. China
| |
Collapse
|
17
|
Soghli N, Qujeq D, Yousefi T, Soghli N. The regulatory functions of circular RNAs in osteosarcoma. Genomics 2020; 112:2845-2856. [DOI: 10.1016/j.ygeno.2020.03.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
|
18
|
Lan F, Qin Q, Yu H, Yue X. Effect of glycolysis inhibition by miR-448 on glioma radiosensitivity. J Neurosurg 2020; 132:1456-1464. [PMID: 31003211 DOI: 10.3171/2018.12.jns181798] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/03/2018] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Although glucose metabolism reengineering is a typical feature of various tumors, including glioma, key regulators of glycolytic reprogramming are still poorly understood. The authors sought to investigate whether glycolysis inhibition by microRNA (miR)-448 increases radiosensitivity in glioma cells. METHODS The authors used glioma tissue samples from glioma patients, cells from glioblastoma (GBM) cell lines and normal human astrocyte cells, and subcutaneous tumor-bearing U87 cells in mice to examine the effects of signaling regulation by miR-448 in the response of glioma tissues and cells to radiation treatment. Techniques used for investigation included bioinformatics analyses, biochemical assays, luciferase reporter assays, and establishment of subcutaneous tumors in a mouse model. Glucose consumption, LDH activity, and cellular ATP were measured to determine the ability of glioma cells to perform glycolysis. Expression of HIF-1α was measured as a potential target gene of miR-448 in glycolysis. RESULTS miR-448 was detected and determined to be significantly downregulated in both glioma tissues from glioma patients and GBM cell lines. Furthermore, miR-448 acted as a tumor-inhibiting factor and suppressed glycolysis in glioma by negatively regulating the activity of HIF-1α signaling and then interfering with its downstream regulators relative to glycolysis, HK1, HK2, and LDHA. Interestingly, overexpression of miR-448 increased the x-radiation sensitivity of glioma cells. Finally, in in vivo experiments, subcutaneous tumor-bearing U87 cells in a mouse model verified that high expression of miR-448 also enhanced glioma radiosensitivity via inhibiting glycolytic factors. CONCLUSIONS miR-448 can promote radiosensitivity by inhibiting HIF-1α signaling and then negatively controlling the glycolysis process in glioma. A newly identified miR-448-HIF-1α axis acts as a potentially valuable therapeutic target that may be useful in overcoming radioresistance in glioma treatment.
Collapse
Affiliation(s)
- Fengming Lan
- 1Department of Radiation Oncology, National Cancer Center/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Qing Qin
- 2Neuro-oncology Chemotherapy Center, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing
| | - Huiming Yu
- 3Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Beijing University Cancer Hospital and Institute, Beijing; and
| | - Xiao Yue
- 4Department of Neurosurgery, The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, People's Republic of China
| |
Collapse
|
19
|
Anauate AC, Leal MF, Calcagno DQ, Gigek CO, Karia BTR, Wisnieski F, dos Santos LC, Chen ES, Burbano RR, Smith MAC. The Complex Network between MYC Oncogene and microRNAs in Gastric Cancer: An Overview. Int J Mol Sci 2020; 21:ijms21051782. [PMID: 32150871 PMCID: PMC7084225 DOI: 10.3390/ijms21051782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Despite the advancements in cancer treatments, gastric cancer is still one of the leading causes of death worldwide. In this context, it is of great interest to discover new and more effective ways of treating this disease. Accumulated evidences have demonstrated the amplification of 8q24.21 region in gastric tumors. Furthermore, this is the region where the widely known MYC oncogene and different microRNAs are located. MYC deregulation is key in tumorigenesis in various types of tissues, once it is associated with cell proliferation, survival, and drug resistance. microRNAs are a class of noncoding RNAs that negatively regulate the protein translation, and which deregulation is related with gastric cancer development. However, little is understood about the interactions between microRNAs and MYC. Here, we overview the MYC role and its relationship with the microRNAs network in gastric cancer aiming to identify potential targets useful to be used in clinic, not only as biomarkers, but also as molecules for development of promising therapies.
Collapse
Affiliation(s)
- Ana Carolina Anauate
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Mariana Ferreira Leal
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Danielle Queiroz Calcagno
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém PA 66075-110, Brazil; (D.Q.C.); (R.R.B.)
| | - Carolina Oliveira Gigek
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Departamento de Patologia, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Bruno Takao Real Karia
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Fernanda Wisnieski
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Disciplina de Gastroenterologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Leonardo Caires dos Santos
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Elizabeth Suchi Chen
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
| | - Rommel Rodríguez Burbano
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém PA 66075-110, Brazil; (D.Q.C.); (R.R.B.)
- Laboratório de Citogenética Humana, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém PA 66075-110, Brazil
- Laboratório de Biologia Molecular, Hospital Ophir Loyola, Belém PA 66063-240, Brazil
| | - Marília Arruda Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil; (A.C.A.); (M.F.L.); (C.O.G.); (B.T.R.K.); (F.W.); (L.C.d.S.); (E.S.C.)
- Correspondence: ; Tel.: +55-11-5576-4848
| |
Collapse
|
20
|
Koca D, Hastar N, Engür S, Kiraz Y, Ulu GT, Çekdemir D, Baran Y. Therapeutic Potentials of Inhibition of Jumonji C Domain-containing Demethylases in Acute Myeloid Leukemia. Turk J Haematol 2020; 37:5-12. [PMID: 31833715 PMCID: PMC7057756 DOI: 10.4274/tjh.galenos.2019.2019.0083] [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] [Indexed: 12/01/2022] Open
Abstract
Objective: Acute myeloid leukemia (AML) is a complex disease affected by both genetic and epigenetic factors. Histone methylation and demethylation are types of epigenetic modification in chromatin remodeling and gene expression. Abnormal expression of histone demethylases is indicated in many types of cancer including AML. Although many commercial drugs are available to treat AML, an absolute cure has not been discovered yet. However, inhibition of demethylases could be a potential cure for AML. Methylstat is a chemical agent that inhibits the Jumonji C domain-containing demethylases. Materials and Methods: The cytotoxic and apoptotic effects of methylstat and doxorubicin on HL-60 cells were detected by MTT cell viability assay, double staining of treated cells with annexin-V/propidium iodide, and caspase-3 activity assay. Mitochondrial activity was analyzed using JC-1 dye. The expression levels of the BCL2 and BCL2L1 anti-apoptotic genes in HL-60 cells were determined using real-time polymerase chain reaction (PCR). Lastly, the cytostatic effect was determined by cell cycle analysis. Results: In our research, cytotoxic, cytostatic, and apoptotic effects of methylstat on human HL-60 cells were investigated. Cytotoxic and cytostatic analyses revealed that methylstat decreased cell proliferation in a dose-dependent cytotoxic manner and arrested HL-60 cells in the G2/M and S phases. Methylstat also induced apoptosis through the loss of mitochondrial membrane potential and increases in caspase-3 enzyme activity. The expression levels of BCL2 and BCL2L1 were also decreased according to real-time PCR results. Finally, the combination of methylstat with doxorubicin resulted in synergistic cytotoxic effects on HL-60 cells. Conclusion: Taken together, these results demonstrate that methylstat may be a powerful candidate as a drug component of AML treatment protocols.
Collapse
Affiliation(s)
- Duygu Koca
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Nurcan Hastar
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Selin Engür
- Anadolu University Faculty of Pharmacy, Department of Pharmacology, Eskişehir, Turkey
| | - Yağmur Kiraz
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Gizem Tuğçe Ulu
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Demet Çekdemir
- Sakarya University Faculty of Medicine, Department of Hematology, Sakarya, Turkey
| | - Yusuf Baran
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| |
Collapse
|
21
|
Lysine demethylase 2 (KDM2B) regulates hippo pathway via MOB1 to promote pancreatic ductal adenocarcinoma (PDAC) progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:13. [PMID: 31941533 PMCID: PMC6961382 DOI: 10.1186/s13046-019-1489-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
Abstract
Background Mps1 binding protein (MOB1) is one of the core components of the mammalian Hippo pathway and plays important roles in cancer development. However, its expression, function and regulation in pancreatic ductal adenocarcinoma (PDAC) have not been revealed yet. Methods The expression of MOB1 and lysine demethylase 2B (KDM2B) in PDAC and adjacent normal pancreas tissues were measured. Also, the underlying mechanisms of altered MOB1 expression and its impact on PDAC biology were investigated. Results We revealed for the first time that MOB1 was decreased expression in PDAC and was a statistically significant independent predictor of poor survival, and restored expression of MOB1 suppressed the proliferation, migration and invasion of PDAC cells. Further studies demonstrated that KDM2B directly bound to the promoter region of MOB1, and suppressed the promoter activity of MOB1 and transcriptionally inhibited the MOB1 expression. Furthermore, KDM2B regulated Hippo pathway and promoted PDAC proliferation, migration and invasion via MOB1. Conclusion This study demonstrated the mechanism and roles of a novel KDM2B/MOB1/Hippo signaling in PDAC progression.
Collapse
|
22
|
Chu Y, Ge W, Wang X. MicroRNA-448 modulates the progression of neuropathic pain by targeting sirtuin 1. Exp Ther Med 2019; 18:4665-4672. [PMID: 31807151 PMCID: PMC6878874 DOI: 10.3892/etm.2019.8165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) play crucial roles in the pathogenesis of neuropathic pain. The present study investigated the effects of miR-448 on the progression of neuropathic pain in a rat model of chronic constriction injury (CCI) of the sciatic nerve. Reverse-transcription quantitative polymerase chain reaction was conducted to detect the gene expression. The paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were used to assess the pain threshold. The protein expression levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α (TNF-α) were detected by ELISA. The target of miR-448 was predicted by TargetScan software. The Student's t-test or one-way ANOVA were used to identify statistical differences among groups. miR-448 was persistently upregulated in CCI rats, and both mechanical allodynia and thermal hyperalgesia in CCI rats were decreased following miR-448 downregulation. The expression levels of IL-1β, IL-6 and TNF-α were significantly increased in CCI rats compared with controls, and these effects were reversed following treatment with a miR-448 inhibitor. A luciferase reporter assay revealed that sirtuin 1 (SIRT1) was a target gene of miR-448. SIRT1 was found to abrogate the effect of miR-448 on neuropathic pain development. Collectively, the results of the present study revealed that miR-448 promoted neuropathic pain in CCI rats by regulating neuroinflammation via SIRT1. Therefore, SIRT1 may be considered as a novel biomarker for neuropathic pain.
Collapse
Affiliation(s)
- Yunchao Chu
- Department of Pain Treatment, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Weipeng Ge
- Department of Pain Treatment, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Xin Wang
- Department of Anesthesiology, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| |
Collapse
|
23
|
Nagai Y, Matoba K, Kawanami D, Takeda Y, Akamine T, Ishizawa S, Kanazawa Y, Yokota T, Utsunomiya K, Nishimura R. ROCK2 regulates TGF-β-induced expression of CTGF and profibrotic genes via NF-κB and cytoskeleton dynamics in mesangial cells. Am J Physiol Renal Physiol 2019; 317:F839-F851. [PMID: 31364374 DOI: 10.1152/ajprenal.00596.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The small GTPase Rho and its effector Rho kinase (ROCK) are involved in the pathogenesis of diabetic kidney disease. Rho kinase has two isoforms: ROCK1 and ROCK2. However, it remains unclear which is mainly involved in the progression of diabetic glomerulosclerosis and the regulation of profibrotic mediators. Glomeruli isolated from type 2 diabetic db/db mice demonstrated increased gene expression of transforming growth factor (TGF)-β and its downstream profibrotic mediators. Chemical inhibition of ROCK suppressed the expression of profibrotic mediators in both isolated glomeruli and cultured mesangial cells. An investigation of mechanisms underlying this observation revealed activated ROCK functions through the phosphorylation of JNK and Erk and the nuclear translocation of NF-κB via actin dynamics. Knockdown by siRNA against ROCK1 and ROCK2 showed that ROCK2 but not ROCK1 controls this fibrotic machinery. Further in vivo experiments showed that ROCK2 activity in the renal cortex of db/db mice was elevated compared with control db/m mice. Importantly, oral administration of ROCK2 inhibitor attenuated renal ROCK2 activity, albuminuria, and glomerular fibrosis in db/db mice. These observations indicate that ROCK2 is a key player in the development of diabetic renal injury. Glomerular ROCK2 may be a potential therapeutic target for the treatment of diabetic kidney disease.
Collapse
Affiliation(s)
- Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomoyo Akamine
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Sho Ishizawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Liu Y, Zhang Z, Wang J, Chen C, Tang X, Zhu J, Liu J. Metabolic reprogramming results in abnormal glycolysis in gastric cancer: a review. Onco Targets Ther 2019; 12:1195-1204. [PMID: 30863087 PMCID: PMC6389007 DOI: 10.2147/ott.s189687] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The Warburg effect in tumor cells involves the uptake of high levels of glucose, enhanced glycolysis, and the metabolism of pyruvate to lactic acid rather than oxidative phos-phorylation to generate energy under aerobic conditions. This effect is closely related to the occurrence, invasion, metastasis, drug resistance, and poor prognosis of gastric cancer (GC). Current research has further demonstrated that the Warburg effect in GC cells is not only mediated by the glycolysis pathway, but also includes roles for mitochondria, noncoding RNAs, and other proteins that do not directly regulate metabolism. As a result, changes in the glycolysis pathway not only lead to abnormal glucose metabolism, but they also affect mitochondrial functions, cellular processes such as apoptosis and cell cycle regulation, and the metabolism of lipids and amino acids. In this review, we discuss metabolic reprogramming in GC based on glycolysis, a possible link between glucose metabolism, lipid metabolism, and amino acid metabolism, and we clarify the role of mitochondria. We also examine recent studies of metabolic inhibitors in GC.
Collapse
Affiliation(s)
- Yuanda Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Ze Zhang
- Department of General Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Junyang Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Chao Chen
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Xiaohuan Tang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Jiaming Zhu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| | - Jingjing Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, China, ;
| |
Collapse
|
25
|
HIF-1α-induced miR-23a∼27a∼24 cluster promotes colorectal cancer progression via reprogramming metabolism. Cancer Lett 2018; 440-441:211-222. [PMID: 30393198 DOI: 10.1016/j.canlet.2018.10.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 12/30/2022]
Abstract
Tumor cells switch metabolic profile from oxidative phosphorylation to glycolysis in a hypoxic environment for survival and proliferation. The mechanisms governing this metabolic switch, however, remain incompletely understood. Here, we show that three miRNAs in the miR-23a∼27a∼24 cluster, miR-23a, miR-27a and miR-24, are the most upregulated miRNA cluster in colorectal cancer (CRC) under hypoxia. Gain- and loss-of-function assays, a human glucose metabolism array and gene pathway analyses confirm that HIF-1α-induced miR-23a∼27a∼24 cluster collectively regulate glucose metabolic network through regulating various metabolic pathways and targeting multiple tricarboxylic acid cycle (TCA)-related genes. In specific, miR-24/VHL/HIF-1α in CRC form a double-negative feedback loop, which in turn, promotes the cellular transition to the 'high HIF-1α/miR-24 and low VHL' state and facilitates cell survival. Our findings reveal that the miR-23a∼27a∼24 cluster is critical regulator switching CRC metabolism from oxidative phosphorylation to glycolysis, and controlling their expression can suppress colorectal cancer progression.
Collapse
|
26
|
Yan M, Yang X, Wang H, Shao Q. The critical role of histone lysine demethylase KDM2B in cancer. Am J Transl Res 2018; 10:2222-2233. [PMID: 30210666 PMCID: PMC6129528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/05/2018] [Indexed: 06/08/2023]
Abstract
The discovery of histone demethylases has revealed the dynamic nature of the regulation of histone methylation. KDM2B is an important histone lysine demethylase that removes methyl from H3K36me2 and H3K4me3. It participates in many aspects of normal cellular processes such as cell senescence, cell differentiation and stem cell self-renewal. Recent studies also showed that KDM2B was overexpressed in various types of cancers. This review focuses primarily on the current knowledge of KDM2B and its function in cancer development.
Collapse
Affiliation(s)
- Meina Yan
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Xinxin Yang
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Hui Wang
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Qixiang Shao
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
27
|
Wei Y, Liang J, Zhang R, Guo Y, Shen S, Su L, Lin X, Moran S, Helland Å, Bjaanæs MM, Karlsson A, Planck M, Esteller M, Fleischer T, Staaf J, Zhao Y, Chen F, Christiani DC. Epigenetic modifications in KDM lysine demethylases associate with survival of early-stage NSCLC. Clin Epigenetics 2018; 10:41. [PMID: 29619118 PMCID: PMC5879927 DOI: 10.1186/s13148-018-0474-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022] Open
Abstract
Background KDM lysine demethylase family members are related to lung cancer clinical outcomes and are potential biomarkers for chemotherapeutics. However, little is known about epigenetic alterations in KDM genes and their roles in lung cancer survival. Methods Tumor tissue samples of 1230 early-stage non-small cell lung cancer (NSCLC) patients were collected from the five independent cohorts. The 393 methylation sites in KDM genes were extracted from epigenome-wide datasets and analyzed by weighted random forest (Ranger) in discovery phase and validation dataset, respectively. The variable importance scores (VIS) for the sites in top 5% of both discovery and validation sets were carried forward for Cox regression to further evaluate the association with patient’s overall survival. TCGA transcriptomic data were used to evaluate the correlation with the corresponding DNA methylation. Results DNA methylation at sites cg11637544 in KDM2A and cg26662347 in KDM1A were in the top 5% of VIS in both discovery phase and validation for squamous cell carcinomas (SCC), which were also significantly associated with SCC survival (HRcg11637544 = 1.32, 95%CI, 1.16–1.50, P = 1.1 × 10−4; HRcg26662347 = 1.88, 95%CI, 1.37–2.60, P = 3.7 × 10−3), and correlated with corresponding gene expression (cg11637544 for KDM2A, P = 1.3 × 10−10; cg26662347 for KDM1A P = 1.5 × 10−5). In addition, by using flexible criteria for Ranger analysis followed by survival classification tree analysis, we identified four clusters for adenocarcinomas and five clusters for squamous cell carcinomas which showed a considerable difference of clinical outcomes with statistical significance. Conclusions These findings highlight the association between somatic DNA methylation in KDM genes and early-stage NSCLC patient survival, which may reveal potential epigenetic therapeutic targets. Electronic supplementary material The online version of this article (10.1186/s13148-018-0474-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yongyue Wei
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Junya Liang
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China
| | - Ruyang Zhang
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Yichen Guo
- 3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Sipeng Shen
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Li Su
- 2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China
| | - Xihong Lin
- 4Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Sebastian Moran
- 5Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet, Barcelona, Catalonia Spain
| | - Åslaug Helland
- 6Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Maria M Bjaanæs
- 6Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anna Karlsson
- 7Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381 Lund, Sweden
| | - Maria Planck
- 7Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381 Lund, Sweden
| | - Manel Esteller
- 5Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet, Barcelona, Catalonia Spain
| | - Thomas Fleischer
- 6Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Johan Staaf
- 7Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381 Lund, Sweden
| | - Yang Zhao
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - Feng Chen
- 1Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166 China.,2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
| | - David C Christiani
- 2China International Cooperation Center (CICC) for Environment and Human Health, Nanjing Medical University, Nanjing, 211166 China.,3Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA.,8Pulmonary and Critical Care Unit, Massachusetts General Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
28
|
Peta E, Sinigaglia A, Masi G, Di Camillo B, Grassi A, Trevisan M, Messa L, Loregian A, Manfrin E, Brunelli M, Martignoni G, Palù G, Barzon L. HPV16 E6 and E7 upregulate the histone lysine demethylase KDM2B through the c-MYC/miR-146a-5p axys. Oncogene 2018; 37:1654-1668. [PMID: 29335520 DOI: 10.1038/s41388-017-0083-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 12/13/2022]
Abstract
Persistent infection by high-risk human papillomaviruses (HPVs) is associated with the development of cervical cancer and a subset of anogenital and head and neck squamous cell carcinomas. Abnormal expression of cellular microRNAs (miRNAs) plays an important role in the development of cancer, including HPV-related tumors. In this study, we demonstrated that miR-146a-5p was down-regulated by E6 and, less efficiently, by E7 of high-risk HPV16 in keratinocytes and the presence of low levels of this miRNA in cervical carcinoma cell lines and in high-risk HPV-positive cervical specimens. Down-regulation of miR-146a-5p was mediated at least in part by the transcription repressor c-MYC, through binding sites in the miR-146a promoter. Overexpression of miR-146a-5p significantly inhibited proliferation and migration of keratinocytes and cervical cancer cells. The histone demethylase KDM2B was validated as a new direct target of miR-146a-5p and two putative binding sites for miR-146a-5p were identified in its 3'UTR sequence. Western blot analysis and immunohistochemistry showed that KDM2B was overexpressed in HPV16 E6/E7-positive keratinocytes, in cervical cancer cell lines, and in a subset of invasive cervical carcinomas and HPV-positive laryngeal squamous cell carcinomas. In these tumors, KDM2B overexpression was associated with c-MYC copy number gain. In vitro, silencing of KDM2B inhibited proliferation of cervical cancer cells. In conclusion, this study identified a novel player, the hystone demethylase KDM2B, in HPV-mediated tumorigenesis. E6 and, less efficiently, E7 of high-risk HPV16 up-regulated KDM2B expression in human keratinocytes through a pathway involving overexpression of c-MYC, which in turn downregulated miR-146a-5p.
Collapse
Affiliation(s)
- Elektra Peta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Giulia Masi
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Barbara Di Camillo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Angela Grassi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Marta Trevisan
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Lorenzo Messa
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Erminia Manfrin
- Department of Diagnostics and Public Health, Anatomic Pathology, AOUI Hospital Trust of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Anatomic Pathology, AOUI Hospital Trust of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Diagnostics and Public Health, Anatomic Pathology, AOUI Hospital Trust of Verona, Verona, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
29
|
Ma F, Zhang L, Ma L, Zhang Y, Zhang J, Guo B. MiR-361-5p inhibits glycolytic metabolism, proliferation and invasion of breast cancer by targeting FGFR1 and MMP-1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:158. [PMID: 29132384 PMCID: PMC5683540 DOI: 10.1186/s13046-017-0630-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/03/2017] [Indexed: 01/23/2023]
Abstract
Background MicroRNAs function as key regulators in various human cancers, including breast cancer (BC). MiR-361-5p has been proved to be a tumor suppressor in colorectal cancer and gastric cancer in our previous study. In this study, we aim to find out the function of miR-361-5p in breast cancer progression and elaborate the mechanism that miR-361-5p acts its function in breast cancer. Methods and results Here we reported that miR-361-5p was down-regulated in breast cancer tissue compared with normal breast tissue and the expression of miR-361-5p was positively associated with prognosis in BC patients. Functional studies showed that overexpression of miR-361-5p suppressed the proliferation, invasion and metastasis of breast cancer cells both in vivo and in vitro. Mechanistically, we found that miR-361-5p inhibited the proliferation of BC cells by suppressing glycolysis. FGFR1, a promoter of glycolysis-related enzyme, was identified as the target of miR-361-5p that promoted glycolysis and repressed oxidative phosphorylation. Furthermore, we demonstrated that miR-361-5p inhibited breast cancer cells invasion and metastasis by targeting MMP-1. An inverse expression pattern was also found between miR-361-5p and FGFR1 or MMP-1 in a cohort of 60 BC tissues. Conclusion Our results indicate that miR-361-5p inhibits breast cancer cells glycolysis and invasion by respectively repressing FGFR1 and MMP-1, suggesting that miR-361-5p and its targets may serve as therapeutic targets in breast cancer treatment.
Collapse
Affiliation(s)
- Fei Ma
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nangang District, Harbin, China
| | - Lei Zhang
- Department of Ultrasound, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Ma
- Computer Center, the Fifth Hospital of Harbin, Harbin, China
| | - Yiyun Zhang
- Department of Endoscopy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianguo Zhang
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nangang District, Harbin, China
| | - Baoliang Guo
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nangang District, Harbin, China.
| |
Collapse
|
30
|
Yu L, Chen X, Sun X, Wang L, Chen S. The Glycolytic Switch in Tumors: How Many Players Are Involved? J Cancer 2017; 8:3430-3440. [PMID: 29151926 PMCID: PMC5687156 DOI: 10.7150/jca.21125] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/31/2017] [Indexed: 02/07/2023] Open
Abstract
Reprogramming of cellular metabolism is a hallmark of cancers. Cancer cells more readily use glycolysis, an inefficient metabolic pathway for energy metabolism, even when sufficient oxygen is available. This reliance on aerobic glycolysis is called the Warburg effect, and promotes tumorigenesis and malignancy progression. The mechanisms of the glycolytic shift in tumors are not fully understood. Growing evidence demonstrates that many signal molecules, including oncogenes and tumor suppressors, are involved in the process, but how oncogenic signals attenuate mitochondrial function and promote the switch to glycolysis remains unclear. Here, we summarize the current information on several main mediators and discuss their possible mechanisms for triggering the Warburg effect.
Collapse
Affiliation(s)
- Li Yu
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Xun Chen
- Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, People's Republic of China
| | - Xueqi Sun
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Liantang Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Shangwu Chen
- State Key Laboratory for Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| |
Collapse
|
31
|
Zhang Y, Guan DH, Bi RX, Xie J, Yang CH, Jiang YH. Prognostic value of microRNAs in gastric cancer: a meta-analysis. Oncotarget 2017; 8:55489-55510. [PMID: 28903436 PMCID: PMC5589675 DOI: 10.18632/oncotarget.18590] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous articles have reported that expression levels of microRNAs (miRNAs) are associated with survival time of patients with gastric cancer (GC). A systematic review and meta-analysis was performed to study the outcome of it. DESIGN Meta-analysis. METHODS English studies estimating expression levels of miRNAs with any of survival curves in GC were identified up till March 19, 2017 through performing online searches in PubMed, EMBASE, Web of Science and Cochrane Database of Systematic Reviews by two authors independently. The pooled hazard ratios (HR) with 95% confidence intervals (CI) were used to estimate the correlation between miRNA expression and overall survival (OS). RESULTS Sixty-nine relevant articles about 26 miRNAs with 6148 patients were ultimately included. GC patients with high expression of miR-20b (HR=2.38, 95%CI=1.16-4.87), 21 (HR=1.77, 95%CI=1.01-3.08), 106b (HR=1.84, 95%CI=1.15-2.94), 196a (HR=2.66, 95%CI=1.94-3.63), 196b (HR=1.67, 95%CI=1.38-2.02), 214 (HR=1.84, 95%CI=1.27-2.67) or low expression of miR-125a (HR=2.06, 95%CI=1.26-3.37), 137 (HR=3.21, 95%CI=1.68-6.13), 141 (HR=2.47, 95%CI=1.34-4.56), 145 (HR=1.62, 95%CI=1.07-2.46), 146a (HR=2.60, 95%CI=1.63-4.13), 206 (HR=2.85, 95%CI=1.73-4.70), 218 (HR=2.61, 95%CI=1.74-3.92), 451 (HR=1.73, 95%CI=1.19-2.52), 486-5p (HR=2.45, 95%CI=1.65-3.65), 506 (HR=2.07, 95%CI=1.33-3.23) have significantly poor OS (P<0.05). CONCLUSIONS In summary, miR-20b, 21, 106b, 125a, 137, 141, 145, 146a, 196a, 196b, 206, 214, 218, 451, 486-5p and 506 demonstrate significantly prognostic value. Among them, miR-20b, 125a, 137, 141, 146a, 196a, 206, 218, 486-5p and 506 are strong biomarkers of prognosis in GC.
Collapse
Affiliation(s)
- Yue Zhang
- 1 First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, People's Republic of China
| | - Dong-Hui Guan
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Rong-Xiu Bi
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Jin Xie
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Chuan-Hua Yang
- 3 Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Yue-Hua Jiang
- 4 Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| |
Collapse
|
32
|
Jiang W, Wang S, Sun Y, Jiang Y, Yu T, Wang J. Overexpression of microRNA-448 inhibits osteosarcoma cell proliferation and invasion through targeting of astrocyte elevated gene-1. Mol Med Rep 2017; 16:5713-5721. [DOI: 10.3892/mmr.2017.7249] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 06/13/2017] [Indexed: 11/06/2022] Open
|
33
|
Kuang Y, Lu F, Guo J, Xu H, Wang Q, Xu C, Zeng L, Yi S. Histone demethylase KDM2B upregulates histone methyltransferase EZH2 expression and contributes to the progression of ovarian cancer in vitro and in vivo. Onco Targets Ther 2017; 10:3131-3144. [PMID: 28706445 PMCID: PMC5495092 DOI: 10.2147/ott.s134784] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aberrant histone methylation contributes to the progression and development of many tumors. Histone methylation is a dynamic process regulated by both histone demethylase and histone methyltransferase, which ultimately alters the levels of gene transcription. However, the relationship between histone demethylase and histone methyltransferase, as well as their regulatory mechanisms in ovarian cancer development, is still unclear. Lysine-specific demethylase 2B (KDM2B) is a key demethylase of H3K36me3 and H3K4me3 that regulates gene expression and plays a role in tumorigenesis via epigenetic mechanisms. To determine the expression pattern of KDM2B in ovarian neoplasms, we analyzed the mRNA and protein levels of KDM2B and the histone methyltransferase enhancer of zester homolog 2 (EZH2) in normal, benign, borderline, and malignant ovarian tissue samples. We found that KDM2B expression was gradually increased in ovarian tumors, with the highest expression found in the malignant ovarian tissues, and the differences in KDM2B expression among the different International Federation of Gynecology and Obstetrics stages and pathological grades/types were statistically significant. Moreover, KDM2B expression was positively correlated with EZH2 expression in ovarian tissues. To determine the role of KDM2B in tumorigenesis in vitro and in vivo, we silenced KDM2B expression in ovarian cancer cells using the KDM2B short hairpin RNA expression lentivirus and established a nude mouse xenograft model. Downregulation of endogenous KDM2B decreased the expression of EZH2 and reduced the proliferation and migration of ovarian cancer cells. Loss of KDM2B suppressed ovarian tumor formation in vivo. Our results suggest that KDM2B plays an important role in the tumorigenesis of ovarian cancer, with a possible mechanism of increasing the expression of the oncogene EZH2; this indicates that certain histone methyltransferase may be positively regulated by certain histone demethylase in the epigenetic regulation of ovarian tumors. KDM2B may be a novel therapeutic target for the clinical treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yan Kuang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Fangfang Lu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Jianfeng Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hong Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Qi Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Chaohuan Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Longjia Zeng
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Suyi Yi
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning
| |
Collapse
|
34
|
Wu X, Yan L, Liu Y, Xian W, Wang L, Ding X. MicroRNA-448 suppresses osteosarcoma cell proliferation and invasion through targeting EPHA7. PLoS One 2017; 12:e0175553. [PMID: 28604772 PMCID: PMC5467824 DOI: 10.1371/journal.pone.0175553] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/28/2017] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma is the most common type of malignant bone tumor, often affecting adolescents and children. MicroRNAs (miRNAs) are a group of small, non-protein coding, endogenous RNAs that play critical roles in osteosarcoma tumorigenesis. In our study, we demonstrated that miR-448 expression was downregulated in osteosarcoma tissues and cell lines. Overexpression of miR-448 suppressed osteosarcoma cell proliferation, colony formation and migration. Moreover, we found that EPHA7 was a direct target gene of miR-448 in osteosarcoma cells. We further demonstrated that the EPHA7 expression level was upregulated in osteosarcoma tissues. Interestingly, the expression level of EPHA7 was inversely correlated with the expression level of miR-448 in osteosarcoma tissues. In addition, elevated expression of miR-448 suppressed osteosarcoma cell proliferation and invasion through targeting EPHA7. Taken together, these findings suggest that miR-448 functioned as a tumor suppressor gene in the development of osteosarcoma through targeting EPHA7.
Collapse
Affiliation(s)
- Xiangkun Wu
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang, Henan, China
| | - Lihua Yan
- Department of Medical Oncology, Nanyang Second People's Hospital, Nanyang, Henan, China
- * E-mail:
| | - Yongxi Liu
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang, Henan, China
| | - Wenfeng Xian
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang, Henan, China
| | - Liuyu Wang
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang, Henan, China
| | - Xunmeng Ding
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang, Henan, China
| |
Collapse
|
35
|
Wu R, He Q, Chen H, Xu M, Zhao N, Xiao Y, Tu QQ, Zhang W, Bi X. MicroRNA-448 promotes multiple sclerosis development through induction of Th17 response through targeting protein tyrosine phosphatase non-receptor type 2 (PTPN2). Biochem Biophys Res Commun 2017; 486:759-766. [DOI: 10.1016/j.bbrc.2017.03.115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/28/2022]
|
36
|
Shan C, Fei F, Li F, Zhuang B, Zheng Y, Wan Y, Chen J. miR-448 is a novel prognostic factor of lung squamous cell carcinoma and regulates cells growth and metastasis by targeting DCLK1. Biomed Pharmacother 2017; 89:1227-1234. [DOI: 10.1016/j.biopha.2017.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 01/08/2023] Open
|
37
|
Song Y, Wang Y, Tong C, Xi H, Zhao X, Wang Y, Chen L. A unified model of the hierarchical and stochastic theories of gastric cancer. Br J Cancer 2017; 116:973-989. [PMID: 28301871 PMCID: PMC5396111 DOI: 10.1038/bjc.2017.54] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/16/2017] [Accepted: 01/26/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a life-threatening disease worldwide. Despite remarkable advances in treatments for GC, it is still fatal to many patients due to cancer progression, recurrence and metastasis. Regarding the development of novel therapeutic techniques, many studies have focused on the biological mechanisms that initiate tumours and cause treatment resistance. Tumours have traditionally been considered to result from somatic mutations, either via clonal evolution or through a stochastic model. However, emerging evidence has characterised tumours using a hierarchical organisational structure, with cancer stem cells (CSCs) at the apex. Both stochastic and hierarchical models are reasonable systems that have been hypothesised to describe tumour heterogeneity. Although each model alone inadequately explains tumour diversity, the two models can be integrated to provide a more comprehensive explanation. In this review, we discuss existing evidence supporting a unified model of gastric CSCs, including the regulatory mechanisms of this unified model in addition to the current status of stemness-related targeted therapy in GC patients.
Collapse
Affiliation(s)
- Yanjing Song
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Yao Wang
- Department of Immunology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuan Tong
- Department of Immunology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongqing Xi
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Xudong Zhao
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Yi Wang
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
38
|
Kong XZ, Hu SS, Sun Z, Zuo LH, Kang J, Zhu ZF, Tian X, Zhang XJ. Regulation of aerobic glycolysis by long non-coding RNAs in cancer. Biochem Biophys Res Commun 2016; 479:28-32. [DOI: 10.1016/j.bbrc.2016.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023]
|