1
|
Hu Y, Li Z, Zhu Y, Xing M, Xie X, Zhao P, Cheng X, Xiao C, Xia Y, Wu J, Luo Y, Ko H, Tang Y, Ye X, Lin WJ. Microglial repopulation reverses radiation-induced cognitive dysfunction by restoring medial prefrontal cortex activity and modulating leukotriene-C4 synthesis. Acta Neuropathol Commun 2025; 13:105. [PMID: 40390112 PMCID: PMC12087111 DOI: 10.1186/s40478-025-02026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Cranial radiotherapy and environmental radiation exposure are associated with increased risk of cognitive dysfunction, including memory deficits and mood disorders, yet the underlying mechanisms remain poorly understood. In this study, we demonstrate that cranial irradiation induces hypoactivity in the medial prefrontal cortex (mPFC) of mice, leading to anxiety-like behaviors and memory impairments, which can be prevented by optogenetic activation of mPFC excitatory neurons. Radiaiton exposure also causes a significant reduction in microglial density within the mPFC, accompanied by morphological and transcriptional alterations in the remaining microglia. Notably, microglial repopulation, achieved through CSF1R antagonist-mediated depletion prior to irradiation and subsequent repopulation, restores mPFC neuronal acitivity and reverses cognitive and behavioral deficits. Integrated bulk RNA sequencing and microglial proteomic analysis of the mPFC reveal that microglial repopulation specifically modulates the leukotriene-C4 biosynthesis pathway, without significant changes in canonical pro-inflammatory cytokines or chemokines. Importantly, pharmacological inhibition of leukotriene-C4 synthase ameliorates radiation-induced anxiety and memory impairments. These findings identify leukotriene-C4 signaling as a critical mechanism underlying radiation-induced cognitive dysfunction and suggest that microglial repopulation and targted inhibition of leukotriene-C4 represent potential therapeutic strategies for mitigating radiation-associated cognitive disorders.
Collapse
Affiliation(s)
- Yubo Hu
- Medical College of Jiaying University, Meizhou, Guangdong, 514031, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhe Li
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yafeng Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Mengdan Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Xiaoru Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Panwu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Chuan Xiao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jingru Wu
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuan Luo
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Gerald Choa Neuroscience Center, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
| | - Yamei Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Wei-Jye Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
2
|
Cao L, Ba Y, Chen F, Li D, Zhang S, Zhang H. The prognostic significance of epoxide hydrolases in colorectal cancer. Biochem Biophys Rep 2025; 41:101912. [PMID: 39850362 PMCID: PMC11754166 DOI: 10.1016/j.bbrep.2024.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Colorectal cancer (CRC) is a common malignant cancer. Epoxide hydrolases (EHs) are involved in the development of cancer by regulating epoxides, but their relationship with CRC is unclear. We used multiple datasets to confirm the expression of different EPHX family members in CRC tissues, and to explore their association with different clinicopathologic characteristics. The Kaplan-Meier method, correlation analysis and random forest algorithm were used to evaluate the prognostic value of EPHX family members for CRC. Finally, the cell experiment verified function of EPHX4 in CRC. The expressions of EPHX1 and EPHX2 were significantly decreased, while those of EPHX3 and EPHX4 were significantly increased in CRC. The expressions of EPHX family members were correlated with some clinicopathologic features and overall survival. The expressions of the EPHX family were positively associated with CD274, CTLA4, HAVCR2, and TIGIT. EPHX2 and EPHX4 were diagnostic and predictive biomarkers for CRC. EPHX4 promoted the malignant phenotype of CRC cells. Our study firstly elucidated the prognostic significance of EPHX family members in CRC and identified novel diagnostic and prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Lichao Cao
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Ying Ba
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Fang Chen
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Dandan Li
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Shenrui Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| | - Hezi Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, 518071, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, 201401, China
- Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, 518110, China
| |
Collapse
|
3
|
Wu M, Sun T, Xing L. Circ_0004913 Inhibits Cell Growth, Metastasis, and Glycolysis by Absorbing miR-184 to Regulate HAMP in Hepatocellular Carcinoma. Cancer Biother Radiopharm 2023; 38:708-719. [PMID: 33021399 DOI: 10.1089/cbr.2020.3779] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Circular RNA (circRNA) can regulate the progression of hepatocellular carcinoma (HCC). However, the role and potential mechanism of circ_0004913 in HCC are not explored. Methods: Circ_0004913 was identified from two GSE datasets (GSE94508 and GSE97322) as a differentially expressed circRNA between HCC and normal tissues. Levels of circ_0004913, microRNA-184 (miR-184), and hepcidin (HAMP) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, migration, and invasion were estimated by methyl thiazolyl tetrazolium, colony formation, and Transwell assays, respectively. Levels of all proteins were examined by Western blot. Glucose consumption and lactate and ATP production were analyzed by the glucose, lactate, and ATP assay kits. Dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pull-down assays were performed to verify the interactions among miR-184 and circ_0004913 or HAMP. The mice xenograft models were established to assess the effect of circ_0004913 on tumor growth in vivo. Results: Circ_0004913 was downregulated in HCC, and its expression impeded cell proliferation, migration, and invasion, EMT, and glycolysis in HCC cells. miR-184 was identified as a target miRNA of circ_0004913, and their expression levels were negatively correlated. miR-184 overexpression could reverse the inhibitory effect of circ_0004913 on HCC cell progression. Moreover, as a target gene of miR-184, HAMP expression was positively correlated with circ_0004913 expression in HCC tissues, and repression of miR-184 could inhibit the progression of HCC cells by increasing HAMP expression. Circ_0004913 could inhibit JAK2/STAT3/AKT signaling pathway and tumor growth in vivo by regulating the miR-184/HAMP axis. Conclusion: Circ_0004913 inhibited the tumorigenesis of HCC by sponging miR-184 to regulate HAMP expression in vitro and in vivo.
Collapse
Affiliation(s)
- Mingyuan Wu
- Department of Gastroenterology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tanlezi Sun
- Basic Medical College, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lianjun Xing
- Department of Gastroenterology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Lin X, Ma Q, Chen L, Guo W, Huang Z, Huang T, Cai YD. Identifying genes associated with resistance to KRAS G12C inhibitors via machine learning methods. Biochim Biophys Acta Gen Subj 2023; 1867:130484. [PMID: 37805078 DOI: 10.1016/j.bbagen.2023.130484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Targeted therapy has revolutionized cancer treatment, greatly improving patient outcomes and quality of life. Lung cancer, specifically non-small cell lung cancer, is frequently driven by the G12C mutation at the KRAS locus. The development of KRAS inhibitors has been a breakthrough in the field of cancer research, given the crucial role of KRAS mutations in driving tumor growth and progression. However, over half of patients with cancer bypass inhibition show limited response to treatment. The mechanisms underlying tumor cell resistance to this treatment remain poorly understood. METHODS To address above gap in knowledge, we conducted a study aimed to elucidate the differences between tumor cells that respond positively to KRAS (G12C) inhibitor therapy and those that do not. Specifically, we analyzed single-cell gene expression profiles from KRAS G12C-mutant tumor cell models (H358, H2122, and SW1573) treated with KRAS G12C (ARS-1620) inhibitor, which contained 4297 cells that continued to proliferate under treatment and 3315 cells that became quiescent. Each cell was represented by the expression levels on 8687 genes. We then designed an innovative machine learning based framework, incorporating seven feature ranking algorithms and four classification algorithms to identify essential genes and establish quantitative rules. RESULTS Our analysis identified some top-ranked genes, including H2AFZ, CKS1B, TUBA1B, RRM2, and BIRC5, that are known to be associated with the progression of multiple cancers. CONCLUSION Above genes were relevant to tumor cell resistance to targeted therapy. This study provides important insights into the molecular mechanisms underlying tumor cell resistance to KRAS inhibitor treatment.
Collapse
Affiliation(s)
- Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou 350014, China.
| | - QingLan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China
| | - Zhiyi Huang
- College of Chemistry, Fuzhou University, Fuzhou 350000, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
5
|
El-Aziz MKA, Dawoud A, Kiriacos CJ, Fahmy SA, Hamdy NM, Youness RA. Decoding hepatocarcinogenesis from a noncoding RNAs perspective. J Cell Physiol 2023; 238:1982-2009. [PMID: 37450612 DOI: 10.1002/jcp.31076] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Being a leading lethal malignancy worldwide, the pathophysiology of hepatocellular carcinoma (HCC) has gained a lot of interest. Yet, underlying mechanistic basis of the liver tumorigenesis is poorly understood. The role of some coding genes and their respective translated proteins, then later on, some noncoding RNAs (ncRNAs) such as microRNAs have been extensively studied in context of HCC pathophysiology; however, the implication of long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) in HCC is indeed less investigated. As a subclass of the ncRNAs which has been elusive for long time ago, lncRNAs was found to be involved in plentiful cellular functions such as DNA, RNA, and proteins regulation. Hence, it is undisputed that lncRNAs dysregulation profoundly contributes to HCC via diverse etiologies. Accordingly, lncRNAs represent a hot research topic that requires prime focus in HCC. In this review, the authors discuss breakthrough discoveries involving lncRNAs and circRNAs dysregulation that have contributed to the contemporary concepts of HCC pathophysiology and how these concepts could be leveraged as potential novel diagnostic and prognostic HCC biomarkers. Further, this review article sheds light on future trends, thereby discussing the pathological roles of lncRNAs and circRNAs in HCC proliferation, migration, and epithelial-to-mesenchymal transition. Along this line of reasoning, future recommendations of how these targets could be exploited to achieve effective HCC-related drug development is highlighted.
Collapse
Affiliation(s)
- Mostafa K Abd El-Aziz
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| | - Alyaa Dawoud
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Caroline J Kiriacos
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Sherif Ashraf Fahmy
- Chemistry Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rana A Youness
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
6
|
Jesenko T, Brezar SK, Cemazar M, Biasin A, Tierno D, Scaggiante B, Grassi M, Grassi C, Dapas B, Truong NH, Abrami M, Zanconati F, Bonazza D, Rizzolio F, Parisi S, Pastorin G, Grassi G. Targeting Non-Coding RNAs for the Development of Novel Hepatocellular Carcinoma Therapeutic Approaches. Pharmaceutics 2023; 15:1249. [PMID: 37111734 PMCID: PMC10145575 DOI: 10.3390/pharmaceutics15041249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a global health challenge, representing the third leading cause of cancer deaths worldwide. Although therapeutic advances have been made in the few last years, the prognosis remains poor. Thus, there is a dire need to develop novel therapeutic strategies. In this regard, two approaches can be considered: (1) the identification of tumor-targeted delivery systems and (2) the targeting of molecule(s) whose aberrant expression is confined to tumor cells. In this work, we focused on the second approach. Among the different kinds of possible target molecules, we discuss the potential therapeutic value of targeting non-coding RNAs (ncRNAs), which include micro interfering RNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). These molecules represent the most significant RNA transcripts in cells and can regulate many HCC features, including proliferation, apoptosis, invasion and metastasis. In the first part of the review, the main characteristics of HCC and ncRNAs are described. The involvement of ncRNAs in HCC is then presented over five sections: (a) miRNAs, (b) lncRNAs, (c) circRNAs, (d) ncRNAs and drug resistance and (e) ncRNAs and liver fibrosis. Overall, this work provides the reader with the most recent state-of-the-art approaches in this field, highlighting key trends and opportunities for more advanced and efficacious HCC treatments.
Collapse
Affiliation(s)
- Tanja Jesenko
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.J.); (S.K.B.); (M.C.)
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.J.); (S.K.B.); (M.C.)
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.J.); (S.K.B.); (M.C.)
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| | - Alice Biasin
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy; (A.B.); (M.G.); (M.A.)
| | - Domenico Tierno
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (B.S.); (B.D.)
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (B.S.); (B.D.)
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy; (A.B.); (M.G.); (M.A.)
| | - Chiara Grassi
- Degree Course in Medicine, University of Trieste, I-34149 Trieste, Italy;
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (B.S.); (B.D.)
| | - Nhung Hai Truong
- Faculty of Biology and Biotechnology, VNUHCM-University of Science, Ho Chi Minh City 70000, Vietnam;
| | - Michela Abrami
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy; (A.B.); (M.G.); (M.A.)
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I-34149 Trieste, Italy; (F.Z.)
| | - Deborah Bonazza
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, I-34149 Trieste, Italy; (F.Z.)
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, I-33081 Aviano, Italy;
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, I-30172 Venezia, Italy;
| | - Salvatore Parisi
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, I-30172 Venezia, Italy;
- Doctoral School in Molecular Biomedicine, University of Trieste, I-34149 Trieste, Italy
| | - Giorgia Pastorin
- Pharmacy Department, National University of Singapore, Block S9, Level 15, 4 Science Drive 2, Singapore 117544, Singapore;
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (B.S.); (B.D.)
| |
Collapse
|
7
|
Wang H, Xu W, Chen X, Mei X, Guo Z, Zhang J. LncRNA LINC00205 stimulates osteoporosis and contributes to spinal fracture through the regulation of the miR-26b-5p/KMT2C axis. BMC Musculoskelet Disord 2023; 24:262. [PMID: 37016415 PMCID: PMC10071705 DOI: 10.1186/s12891-023-06136-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/05/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Osteoporosis (OP) is a common bone disease marked by decreased bone strength. Increasing evidence suggests that long non-coding RNA (lncRNAs) play important roles in the occurrence and progression of OP. This study aimed to investigate the role and mechanism of LINC00205 in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) and OP. METHODS Bone tissue samples were obtained from healthy controls and patients with osteoporosis with a spinal fracture (OP-Frx) or without a spinal fracture (OP-no-Frx). HMSCs were cultured and induced to undergo osteogenic differentiation. The expression of LINC00205, lysine (K)-specific methyltransferase 2C (KMT2C), and miR-26b-5p in bone tissues and cells was evaluated using western blotting and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). The effects of LINC00205, miR-26b-5p, and KMT2C on calcium deposition, alkaline phosphatase (ALP) activity, and mRNA levels of the osteogenic differentiation marker genes [ALP, osteocalcin (OCN), and runt-related transcription factor 2 (RUNX2)] were investigated using alizarin red S staining, an ALP activity assay, and qRT-PCR, respectively. Dual-luciferase reporter assay was performed to ascertain the binding relationship between miR-26b-5p and LINC00205/KMT2C. RESULTS LINC00205 and KMT2C were upregulated in patients with OP-Frx and OP-no-Frx, whereas miR-26b-5p was downregulated. Furthermore, LINC00205 and KMT2C expression decreased, whereas that of miR-26b-5p increased over time from day 7 to 21 of the osteogenic differentiation of hMSCs. The knockdown of LINC00205 and KMT2C significantly increased ALP activity, calcium deposition, and the expression of RUNX2, ALP, and OCN. In contrast, the inhibition of miR-26b-5p yielded the opposite result. These data suggest that LINC00205 inhibits the osteogenic differentiation of hMSCs by modulating the miR-26b-5p/KMT2C signaling axis. CONCLUSION LINC00205 promotes OP and is involved in spinal fractures. LINC00205 is also a potential negative regulator of the osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Hongtao Wang
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Weilin Xu
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Xiaoqing Chen
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Xiongfeng Mei
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Zhonghua Guo
- Department of Orthopaedics, People's Hospital of Dongxihu District, Wuhan, 430040, Hubei, China
| | - Juan Zhang
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China.
| |
Collapse
|
8
|
Huang PS, Wang LY, Wang YW, Tsai MM, Lin TK, Liao CJ, Yeh CT, Lin KH. Evaluation and Application of Drug Resistance by Biomarkers in the Clinical Treatment of Liver Cancer. Cells 2023; 12:869. [PMID: 36980210 PMCID: PMC10047572 DOI: 10.3390/cells12060869] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
Liver cancer is one of the most lethal cancers in the world, mainly owing to the lack of effective means for early monitoring and treatment. Accordingly, there is considerable research interest in various clinically applicable methods for addressing these unmet needs. At present, the most commonly used biomarker for the early diagnosis of liver cancer is alpha-fetoprotein (AFP), but AFP is sensitive to interference from other factors and cannot really be used as the basis for determining liver cancer. Treatment options in addition to liver surgery (resection, transplantation) include radiation therapy, chemotherapy, and targeted therapy. However, even more expensive targeted drug therapies have a limited impact on the clinical outcome of liver cancer. One of the big reasons is the rapid emergence of drug resistance. Therefore, in addition to finding effective biomarkers for early diagnosis, an important focus of current discussions is on how to effectively adjust and select drug strategies and guidelines for the treatment of liver cancer patients. In this review, we bring this thought process to the drug resistance problem faced by different treatment strategies, approaching it from the perspective of gene expression and molecular biology and the possibility of finding effective solutions.
Collapse
Affiliation(s)
- Po-Shuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
| | - Ling-Yu Wang
- Department of Biochemistry and Molecular Biology, Chang Gung University, Taoyuan 333, Taiwan;
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan
| | - Yi-Wen Wang
- School of Nursing, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Ming-Ming Tsai
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan;
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of General Surgery, New Taipei Municipal Tu Cheng Hospital, New Taipei 236, Taiwan
| | - Tzu-Kang Lin
- Neurosurgery, School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- Neurosurgery, Department of Surgery, Fu Jen Catholic University Hospital, New Taipei City 24352, Taiwan
| | - Chia-Jung Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| | - Kwang-Huei Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| |
Collapse
|
9
|
Construction and Validation of Pyroptosis-Related lncRNA Prediction Model for Colon Adenocarcinoma and Immune Infiltration Analysis. DISEASE MARKERS 2022; 2022:4492608. [PMID: 36168326 PMCID: PMC9509522 DOI: 10.1155/2022/4492608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022]
Abstract
Objective Colon adenocarcinoma (COAD) is one of the most prevalent cancers worldwide. However, the pyroptosis-related lncRNAs of COAD have not been deeply examined and validated. Here, we constructed and validated a risk model on pyroptosis-related lncRNAs in COAD. Methods The RNA sequencing transcriptome and clinical data of COAD patients were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed pyroptosis-related mRNAs and mRNA-lncRNA coexpression network were identified. After univariate and multifactorial cox analyses of prognosis-related lncRNAs, a risk model was constructed. Next, we analyzed the differences in immune infiltration, immune checkpoint blockade-, immune checkpoint-, and N6-methyladenosine-related gene expressions between the high- and low-risk groups. RT-qPCR was used to validate the expression of lncRNAs. Result A risk model was constructed based on 9 pyroptosis-related lncRNAs and separated COAD patients into the high- and low-risk groups. Immune infiltration analysis and immune checkpoint blockade-, immune checkpoint-, and N6-methyladenosine-related genes showed significant differences between the two subgroups. RT-qPCR showed that the 9 pyroptosis-related lncRNAs could be used as prognostic indicators. Conclusion A novel risk model based on pyroptosis-related lncRNAs was constructed and demonstrated that these lncRNAs might be used as independent prognostic biomarkers. This will also assist shed light on the COAD prognosis and therapy.
Collapse
|
10
|
Wang F, Lin H, Su Q, Li C. Cuproptosis-related lncRNA predict prognosis and immune response of lung adenocarcinoma. World J Surg Oncol 2022; 20:275. [PMID: 36050740 PMCID: PMC9434888 DOI: 10.1186/s12957-022-02727-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) accounts for 50% of lung cancers, with high mortality and poor prognosis. Long non-coding RNA (lncRNA) plays a vital role in the progression of tumors. Cuproptosis is a newly discovered form of cell death that is highly investigated. Therefore, in the present study, we aimed to investigate the role of cuproptosis-related lncRNA signature in clinical prognosis prediction and immunotherapy and the relationship with drug sensitivity. MATERIAL AND METHODS Genomic and clinical data were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and cuproptosis-related genes were obtained from cuproptosis-related studies. The prognostic signature was constructed by co-expression analysis and Cox regression analysis. Patients were divided into high and low risk groups, and then, a further series of model validations were carried out to assess the prognostic value of the signature. Subsequently, lncRNAs were analyzed for gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes Enrichment (KEGG), immune-related functions, and tumor mutation burden (TMB). Finally, we used tumor immune dysfunction and exclusion (TIDE) algorithms on immune escape and immunotherapy of cuproptosis-related lncRNAs, thereby identifying its sensitivity toward potential drugs for LUAD. RESULTS A total of 16 cuproptosis-related lncRNAs were obtained, and a prognostic signature was developed. We found that high-risk patients had worse overall survival (OS) and progression-free survival (PFS) and higher mortality. Independent prognostic analyses, ROC, C-index, and nomogram showed that the cuproptosis-related lncRNAs can accurately predict the prognosis of patients. The nomogram and heatmap showed a distinct distribution of the high- and low-risk cuproptosis-related lncRNAs. Enrichment analysis showed that the biological functions of lncRNAs are associated with tumor development. We also found that immune-related functions, such as antiviral activity, were suppressed in high-risk patients who had mutations in oncogenes. OS was poorer in patients with high TMB. TIDE algorithms showed that high-risk patients have a greater potential for immune escape and less effective immunotherapy. CONCLUSION To conclude, the 16 cuproptosis-related lncRNAs can accurately predict the prognosis of patients with LUAD and may provide new insights into clinical applications and immunotherapy.
Collapse
Affiliation(s)
- Fangwei Wang
- grid.412594.f0000 0004 1757 2961Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nan’ning, China
| | - Hongsheng Lin
- grid.256607.00000 0004 1798 2653Department of Microbiology, School of Basic Medical Sciences, Guangxi Medical University, Nan’ning, China
| | - Qisheng Su
- grid.412594.f0000 0004 1757 2961Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nan’ning, China
| | - Chaoqian Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nan'ning, China.
| |
Collapse
|
11
|
Long non-coding RNA DARS-AS1 promotes tumor progression by directly suppressing PACT-mediated cellular stress. Commun Biol 2022; 5:822. [PMID: 35970927 PMCID: PMC9378715 DOI: 10.1038/s42003-022-03778-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/29/2022] [Indexed: 11/08/2022] Open
Abstract
Cancer cells evolve various mechanisms to overcome cellular stresses and maintain progression. Protein kinase R (PKR) and its protein activator (PACT) are the initial responders in monitoring diverse stress signals and lead to inhibition of cell proliferation and cell apoptosis in consequence. However, the regulation of PACT-PKR pathway in cancer cells remains largely unknown. Herein, we identify that the long non-coding RNA (lncRNA) aspartyl-tRNA synthetase antisense RNA 1 (DARS-AS1) is directly involved in the inhibition of the PACT-PKR pathway and promotes the proliferation of cancer cells. Using large-scale CRISPRi functional screening of 971 cancer-associated lncRNAs, we find that DARS-AS1 is associated with significantly enhanced proliferation of cancer cells. Accordingly, knocking down DARS-AS1 inhibits cell proliferation of multiple cancer cell lines and promotes cancer cell apoptosis in vitro and significantly reduces tumor growth in vivo. Mechanistically, DARS-AS1 directly binds to the activator domain of PACT and prevents PACT-PKR interaction, thereby decreasing PKR activation, eIF2α phosphorylation and inhibiting apoptotic cell death. Clinically, DARS-AS1 is broadly expressed across multiple cancers and the increased expression of this lncRNA indicates poor prognosis. This study elucidates the lncRNA DARS-AS1 directed cancer-specific modulation of the PACT-PKR pathway and provides another target for cancer prognosis and therapeutic treatment. A loss-of-function screen reveals a role for lncRNA DARS-AS1 in promoting cancer cell proliferation and further experiments shows DARS-AS1 regulates the PACT-PKR pathway, overall suggesting it as a potential target for cancer therapy and prognosis.
Collapse
|
12
|
Kang L, Chen J, Wang J, Zhao T, Wei Y, Wu Y, Han L, Zheng X, Shen L, Long C, Wei G, Wu S. Multiple transcriptomic profiling: potential novel metabolism-related genes predict prepubertal testis damage caused by DEHP exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:13478-13490. [PMID: 34595713 DOI: 10.1007/s11356-021-16701-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
The toxic effect of di(2-ethylhexyl) phthalate (DEHP) on prepubertal testes was examined in this study. We treated 3-week-old male mice with 4.8 mg/kg/day (milligram/kilogram/day) (no observed adverse effect level), 30 mg/kg/day (high exposure dose relative to humans), 100 mg/kg/day (level causing a reproductive system disorder), and 500 mg/kg/day (dose causing a multigenerational reproductive system disorder) of DEHP via gavage. Obvious abnormalities in the testicular organ coefficient, spermatogenic epithelium, and testosterone levels occurred in the 500 mg/kg DEHP group. Ribonucleic acid sequencing (RNA-seq) showed that differentially expressed genes (DEGs) in each group could enrich reproduction and reproductive process terms according to the gene ontology (GO) results, and coenrichment of metabolism pathway was observed by the Reactome pathway analysis. Through the analysis of common genes in the metabolism pathway, we discovered that DEHP exposure at 4.8 to 500 mg/kg or 100 mg/kg caused the same damages to the prepubertal testis. In general, we identified two key transcriptional biomarkers (fatty acid binding protein 3 (Fabp3) and carboxylesterase (Ces) 1d), which provided new insight into the gene regulatory mechanism associated with DEHP exposure and will contribute to the prediction and diagnosis of prepuberty testis injury caused by DEHP.
Collapse
Affiliation(s)
- Lian Kang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Jiadong Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Junke Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Tianxin Zhao
- Department of Pediatric Urology, Guangzhou Woman and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Yuhao Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Lindong Han
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Xiangqin Zheng
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Lianju Shen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Chunlan Long
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.
- National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China.
- Chongqing Key Laboratory of Pediatrics Chongqing, Room 806, Kejiao Building (NO.6), No.136, Zhongshan 2nd Road, Yuzhong District, Chongqing, People's Republic of China.
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, People's Republic of China.
| |
Collapse
|
13
|
Yu Y, Li H, Wu C, Li J. Circ_0021087 acts as a miR-184 sponge and represses gastric cancer progression by adsorbing miR-184 and elevating FOSB expression. Eur J Clin Invest 2021; 51:e13605. [PMID: 34076278 DOI: 10.1111/eci.13605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Gastric cancer (GC) ranks third among the causes of cancer-related deaths in the world. Circular RNA hsa_circ_0021087 (circ_0021087) plays a repressive role in GC. Nevertheless, the mechanism by which circ_0021087 constrains GC advancement is unclear. MATERIALS AND METHODS Expression patterns of circ_0021087, microRNA (miR)-184 and FBJ murine osteosarcoma viral oncogene homolog B (FOSB) mRNA were assessed by quantitative real-time polymerase chain reaction (RT-qPCR). Gain-of-function experiments were conducted to verify the biological function of circ_0021087 in vitro and in vivo, including cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell and xenograft assays. Protein levels were analysed by Western blotting and immunohistochemistry (IHC). The regulatory mechanism of circ_0021087 was analysed by bioinformatics analysis, dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. RESULTS AND CONCLUSION Circ_0021087 and FOSB were lowly expressed in GC, whereas miR-184 had an opposite result. Circ_0021087 overexpression repressed GC cell proliferation and epithelial-mesenchymal transition (EMT) in xenograft models in vivo and induced GC cell apoptosis, repressed GC cell proliferation, EMT, migration and invasion in vitro. Circ_0021087 could elevate FOSB expression by adsorbing miR-184. MiR-184 mimic reversed the inhibitory influence of circ_0021087 overexpression on GC cell malignancy. Also, FOSB knockdown offset the suppressive impact of miR-184 silencing on GC cell malignancy. In conclusion, circ_0021087 played a repressive influence on GC progression by elevating FOSB expression by adsorbing miR-184, offering a new mechanism for circ_0021087 to inhibit the progression of GC.
Collapse
Affiliation(s)
- Yin Yu
- School of Basic Medicine, Zhengzhou University, Zhengzhou City, China
| | - Hong Li
- Department of Radiology, Zhumadian Central Hospital Affiliated to Huanghuai University, Zhumadian City, China
| | - Chunhua Wu
- Department of Oncology, Zhumadian Central Hospital Affiliated to Huanghuai University, Zhumadian City, China
| | - Jinfeng Li
- Department of Obstetrics and Gynecology, Zhumadian Central Hospital Affiliated to Huanghuai University, Zhumadian City, China
| |
Collapse
|
14
|
Li Y, Hu Y, Wu Y, Zhang D, Huang D. LINC00205 Promotes Tumor Malignancy of Lung Adenocarcinoma Through Sponging miR-185-5p. Lab Med 2021; 53:39-46. [PMID: 34270733 DOI: 10.1093/labmed/lmab041] [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] [Indexed: 01/12/2023] Open
Abstract
The emerging role of long noncoding RNAs (lncRNAs) in cancer, especially in lung adenocarcinoma (LUAD), is attracting increasingly more attention as a potential therapeutic target. However, whether lncRNA LINC00205 regulates the malignancy of LUAD has not been characterized. In this study, we discovered that LINC00205 was markedly upregulated in LUAD tissues and cell lines and correlated with poor prognosis of patients with LUAD. Our data showed that LINC00205 promoted the migration and proliferation of LUAD cells in vitro and tumor growth in vivo. Notably, the tumor suppressor miR-185-5p was found to be a direct target of LINC00205. In addition, miR-185-5p diminished the promotion of cell proliferation and migration mediated by LINC00205, whereas miR-185-5p inhibition had the opposite effect. In summary, our results show that LINC00205 contributes to LUAD malignancy by sponging miR-185-5p, which provides new insight into LUAD progression.
Collapse
Affiliation(s)
- Yongqiang Li
- Department of Respiratory Medicine, PLA General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Yahui Hu
- Department of Geriatric Respiratory Medicine, PLA General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Yuting Wu
- Department of Respiratory Medicine, PLA General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Deming Zhang
- Department of Geriatric Respiratory Medicine, PLA General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Dongwei Huang
- Department of Geriatric Respiratory Medicine, PLA General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Luo Y, Li H, Huang H, Xue L, Li H, Liu L, Fu H. Integrated analysis of ceRNA network in hepatocellular carcinoma using bioinformatics analysis. Medicine (Baltimore) 2021; 100:e26194. [PMID: 34087888 PMCID: PMC8183720 DOI: 10.1097/md.0000000000026194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 05/13/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) can work as microRNA (miRNA) sponges through a competitive endogenous RNA (ceRNA) mechanism. LncRNAs and miRNAs are important components of competitive endogenous binding, and their expression imbalance in hepatocellular carcinoma (HCC) is closely related to tumor development, diagnosis, and prognosis. This study explored the potential impact of the ceRNA regulatory network in HCC on the prognosis of HCC patients. METHODS We thoroughly researched the differential expression profiles of lncRNAs, miRNAs, and mRNAs from 2 HCC Gene Expression Omnibus datasets (GSE98269 and GSE60502). Then, a dysregulated ceRNA network was constructed by bioinformatics. In addition, hub genes in the ceRNA network were screened by Cytoscape, these hub genes functional analysis was performed by gene set enrichment analysis, and the expression of these hub genes in tumors and their correlation with patient prognosis were verified with Gene Expression Profiling Interactive Analysis. RESULTS A ceRNA network was successfully constructed in this study including 4 differentially expressed (DE) lncRNAs, 7 DEmiRNAs, and 166 DEmRNAs. Importantly, 4 core genes (CCNA2, CHEK1, FOXM1, and MCM2) that were significantly associated with HCC prognosis were identified. CONCLUSIONS Our study provides comprehensive and meaningful insights into HCC tumorigenesis and the underlying molecular mechanisms of ceRNA. Furthermore, the specific ceRNAs can be further used as potential therapeutic targets and prognostic biomarkers for HCC.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Liu
- Department of Liver Disease, The 3rd People's Hospital of Kunming, Kunming, Yunnan, China
| | | |
Collapse
|
16
|
Wu Y, Deng J, Lai S, You Y, Wu J. A risk score model with five long non-coding RNAs for predicting prognosis in gastric cancer: an integrated analysis combining TCGA and GEO datasets. PeerJ 2021; 9:e10556. [PMID: 33614260 PMCID: PMC7879943 DOI: 10.7717/peerj.10556] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/22/2020] [Indexed: 12/12/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common carcinomas of the digestive tract, and the prognosis for these patients may be poor. There is evidence that some long non-coding RNAs(lncRNAs) can predict the prognosis of patients with GC. However, few lncRNA signatures have been used to predict prognosis. Herein, we aimed to construct a risk score model based on the expression of five lncRNAs to predict the prognosis of patients with GC and provide new potential therapeutic targets. Methods We performed differentially expressed and survival analyses to identify differentially expressed survival-ralated lncRNAs by using GC patient expression profile data from The Cancer Genome Atlas (TCGA) database. We then established a formula including five lncRNAs to predict the prognosis of patients with GC. In addition, to verify the prognostic value of this risk score model, two independent Gene Expression Omnibus (GEO) datasets, GSE62254 (N = 300) and GSE15459 (N = 200), were employed as validation groups. Results Based on the characteristics of five lncRNAs, patients with GC were divided into high or low risk subgroups. The prognostic value of the risk score model with five lncRNAs was confirmed in both TCGA and the two independent GEO datasets. Furthermore, stratification analysis results showed that this model had an independent prognostic value in patients with stage II-IV GC. We constructed a nomogram model combining clinical factors and the five lncRNAs to increase the accuracy of prognostic prediction. Enrichment analysis based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) suggested that the five lncRNAs are associated with multiple cancer occurrence and progression-related pathways. Conclusion The risk score model including five lncRNAs can predict the prognosis of patients with GC, especially those with stage II-IV, and may provide potential therapeutic targets in future.
Collapse
Affiliation(s)
- Yiguo Wu
- Department of Medicine, Nanchang University, Nan Chang, China
| | - Junping Deng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Shuhui Lai
- Department of Medicine, Nanchang University, Nan Chang, China
| | - Yujuan You
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nan Chang, China
| | - Jing Wu
- Shenzhen Prevention and Treatment Center for Occupational Diseases, Shen Zhen, China
| |
Collapse
|
17
|
Shi Q, Xue C, Yuan X, He Y, Yu Z. Gene signatures and prognostic values of m1A-related regulatory genes in hepatocellular carcinoma. Sci Rep 2020; 10:15083. [PMID: 32934298 PMCID: PMC7492257 DOI: 10.1038/s41598-020-72178-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks fourth in cancer-related mortality worldwide. N1-methyladenosine (m1A), a methylation modification on RNA, is gaining attention for its role across diverse biological processes. However, m1A-related regulatory genes expression, its relationship with clinical prognosis, and its role in HCC remain unclear. In this study, we utilized The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database to investigate alterations within 10 m1A-related regulatory genes and observed a high mutation frequency (23/363). Cox regression analysis and least absolute shrinkage and selection operator were used to explore the association between m1A-related regulatory genes expression and HCC patient survival and identified four regulators that were remarkably associated with HCC patient prognosis. Additionally, an independent cohort from International Cancer Genome Consortium was studied to validate our discoveries and found to be consistent with those in the TCGA dataset. In terms of mechanism, gene set enrichment analysis linked these four genes with various physiological roles in cell division, the MYC pathway, protein metabolism, and mitosis. Kyoto Encyclopedia of Genes and Genomes analysis revealed that PI3K/Akt signaling pathway had potential relevance to m1A-related regulatory genes in HCC. These findings indicate that m1A-related regulatory genes may play crucial roles in regulating HCC progression and be exploited for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Qingmiao Shi
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Chen Xue
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xin Yuan
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yuting He
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China. .,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zujiang Yu
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China. .,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
18
|
Filippov-Levy N, Reich R, Davidson B. The Biological and Clinical Role of the Long Non-Coding RNA LOC642852 in Ovarian Carcinoma. Int J Mol Sci 2020; 21:E5237. [PMID: 32718068 PMCID: PMC7432776 DOI: 10.3390/ijms21155237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022] Open
Abstract
The objective of the present study was to analyze the biological and clinical role of the long non-coding RNA LOC642852 in ovarian carcinoma (OC). LOC642852 expression was analyzed in seven OC cell lines (OVCAR-3, OVCAR-8, OVCA 433, OVCA 429, OC 238, DOV13, ES-2) and 139 high-grade serous carcinoma (HGSC) specimens (85 effusions, 54 surgical specimens). Following LOC642852 knockout (KO) using the CRISPR/Cas9 system, OVCAR-8 HGSC cells were analyzed for spheroid formation, migration, invasion, proliferation, matrix metalloproteinase (MMP) activity, and expression of cell signaling proteins. OVCAR-8 cells with LOC642852 KO were significantly less motile and less invasive compared to controls, with no differences in spheroid formation, proliferation, or matrix metalloproteinase (MMP) activity. Total Akt and Erk levels were comparable in controls and KO cells, but their phosphorylation was significantly increased in the latter. In clinical specimens, LOC642852 was overexpressed in ovarian tumors and omental/peritoneal metastases compared to effusion specimens (p = 0.013). A non-significant trend for shorter overall (p = 0.109) and progression-free (p = 0.056) survival was observed in patients with HGSC effusions with high LOC642852 levels. Bioinformatics analysis showed potential roles for LOC642852 as part of the TLE3/miR-221-3p ceRNA network and in relation to the FGFR3 protein. In conclusion, LOC642852 inactivation via CRISPR/Cas9 affects cell signaling, motility, and invasion in HGSC cells. LOC642852 is differentially expressed in HGSC cells at different anatomical sites. Its potential role in regulating the TLE3/miR-221-3p ceRNA network and FGFR3 merits further research.
Collapse
Affiliation(s)
- Natalie Filippov-Levy
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel;
| | - Reuven Reich
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel;
| | - Ben Davidson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway;
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310 Oslo, Norway
| |
Collapse
|
19
|
Chang W, Fa H, Xiao D, Wang J. MicroRNA-184 alleviates insulin resistance in cardiac myocytes and high fat diet-induced cardiac dysfunction in mice through the LPP3/DAG pathway. Mol Cell Endocrinol 2020; 508:110793. [PMID: 32229289 DOI: 10.1016/j.mce.2020.110793] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 01/05/2023]
Abstract
AIM Cardiovascular complication is a major cause of mortality and morbidity in patients with diabetes. Insulin sensitivity loss is a major contributor to the pathogenesis of cardiovascular diseases in diabetes. Based on our previous research, diacylglycerol (DAG) levels play an important role in high saturated fatty acid-induced insulin resistance. Phosphatidic acid phosphatase (LPP3), a key enzyme for synthesizing DAG, is indispensable for normal cardiac functions and vascular health. However, adipose knockdown of LPP3 increases insulin sensitivity, suggesting that LPP3 regulation may be complicated in hearts. The aim of this study was to investigate LPP3 roles in diabetic cardiac insulin sensitivity and to identify potential upstream targets implicated in diabetic cardiomyopathy. METHODS AND RESULTS Mice were fed a high fat diet (HF) or a low fat diet (control) for up to 24 weeks. After 24 weeks, we found that high fat diet-induced cardiac dysfunction is linked to elevated LPP3 compared to the control group (P < 0.05). In addition, knockdown of LPP3 rescued the glucose uptake that was impaired by palmitate treatment alone in cardiomyoblasts (P < 0.05). Furthermore, we identified miR-184 as an upstream regulator targeting LPP3 and further confirmed the link between DAG and insulin sensitivity. MiR-184 mimic transfection rescued the glucose uptake and glucose consumption that had been impaired by palmitate treatment alone (P < 0.05). CONCLUSION In hearts of high fat diet-fed mice, increased LPP3 contributes to insulin resistance via increased DAG levels. A small non-coding RNA, miR-184, at least partially regulates this signal pathway to alleviate insulin resistance.
Collapse
Affiliation(s)
- Wenguang Chang
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Hongge Fa
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China; School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China; School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Zhang S, Long J, Hu Y. Long noncoding RNA LINC00205 enhances the malignant characteristics of retinoblastoma by acting as a molecular sponge of microRNA-665 and consequently increasing HMGB1 expression. Biochem Biophys Res Commun 2020; 526:396-403. [PMID: 32223925 DOI: 10.1016/j.bbrc.2020.03.083] [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: 03/10/2020] [Accepted: 03/15/2020] [Indexed: 10/24/2022]
Abstract
Long intergenic non-protein-coding RNA 00205 (LINC00205) has been found to play crucial roles in hepatocellular carcinoma progression. In this study, we aimed to determine the expression pattern of LINC00205 in retinoblastoma (RB), to identify its functions in RB progression in detail, and to reveal the underlying mechanisms. Herein, we showed that LINC00205 is highly expressed in RB tissues and cell lines. The LINC00205 upregulation correlated with adverse clinicopathological parameters and shorter overall survival in patients with RB. LINC00205 depletion decreased the proliferative, migratory, and invasive abilities; promoted the apoptosis of RB cells in vitro; and impeded the tumor growth of RB cells in vivo. Mechanism investigation revealed that LINC00205 can act as a competing endogenous RNA by sponging microRNA-665 (miR-665) in RB cells, thereby upregulating miR-665's target: high-mobility group box 1 (HMGB1). Finally, rescue experiments confirmed that enhancing the miR-665-HMGB1 axis output attenuated the influence of the LINC00205 knockdown on RB cells. To sum up, the newly identified LINC00205-miR-665-HMGB1 pathway was systematically studied and may be validated as a potential target for RB diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Ophthalmology, The First People's Hospital of Jinzhou (Yangtze University Affiliated First People's Hospital), Hubei, 434000, PR China
| | - Jian Long
- Department of Oncology, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Hubei, 434020, PR China.
| | - Yin Hu
- Department of Ophthalmology, Xiangyang Central Hospital, Hubei, 441021, PR China.
| |
Collapse
|