51
|
Wang S, Ling Y, Yao Y, Zheng G, Chen W. Luteolin inhibits respiratory syncytial virus replication by regulating the MiR-155/SOCS1/STAT1 signaling pathway. Virol J 2020; 17:187. [PMID: 33239033 PMCID: PMC7688008 DOI: 10.1186/s12985-020-01451-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract infection in infants, children, immunocompromised adults, and elderly individuals. Currently, there are few therapeutic options available to prevent RSV infection. The present study aimed to investigate the effects of luteolin on RSV replication and the related mechanisms. Material and methods We pretreated cells and mice with luteolin before infection with RSV, the virus titer, expressions of RSV-F, interferon (IFN)-stimulated genes (ISGs), and production of IFN-α and IFN-β were determined by plaque assay, RT-qPCR, and ELISA, respectively. The activation of Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) signaling pathway was detected by Western blotting and luciferase assay. Proteins which negatively regulate STAT1 were determined by Western blotting. Then cells were transfected with suppressor of cytokine signaling 1 (SOCS1) plasmid and virus replication and ISGs expression were determined. Luciferase reporter assay and Western blotting were performed to detect the relationship between SOCS1 and miR-155. Results Luteolin inhibited RSV replication, as shown by the decreased viral titer and RSV-F mRNA expression both in vitro and in vivo. The antiviral activity of luteolin was attributed to the enhanced phosphorylation of STAT1, resulting in the increased production of ISGs. Further study showed that SOCS1 was downregulated by luteolin and SOCS1 is a direct target of microRNA-155 (miR-155). Inhibition of miR-155 rescued luteolin-mediated SOCS1 downregulation, whereas upregulation of miR-155 enhanced the inhibitory effect of luteolin. Conclusion Luteolin inhibits RSV replication by regulating the miR-155/SOCS1/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Saisai Wang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Yiting Ling
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Yuanyuan Yao
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Gang Zheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
52
|
Liang S, Hu J, Zhang A, Li F, Li X. miR-155 induces endothelial cell apoptosis and inflammatory response in atherosclerosis by regulating Bmal1. Exp Ther Med 2020; 20:128. [PMID: 33082860 PMCID: PMC7557345 DOI: 10.3892/etm.2020.9259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death from vascular diseases worldwide, and endothelial cell (EC) dysfunction is the key cause of atherosclerosis. miR-155 was found to induce endothelial injury and to trigger atherosclerosis. In addition, brain and muscle ARNT-like protein-1 (Bmal1) has been found to be closely related to EC function. Therefore, the present study aimed to explore the mechanism underlying the regulation of Bmal1 by miR-155 in the induction of EC apoptosis and inflammatory response in atherosclerosis. The atherosclerosis model in apolipoprotein E (ApoE)- / - mice was established. miR-155 and Bmal1 expression was quantified by RT-qPCR and western blot analysis, respectively. The role of miR-155 and Bmal1 in atherosclerosis was evaluated through changes in cardiac function, plaque area, cardiomyocyte apoptosis, and inflammatory factor levels in mice. Moreover, the regulatory relationship between them was identified by dual-luciferase reporter gene assay to explore the mechanism of action of miR-155. After the modeling, the expression of miR-155 was upregulated and Bmal1 was downregulated in aorta, and there was a significant linear correlation between them. Upregulation of miR-155 increased the atherosclerotic plaque area, cell apoptosis, total cholesterol (TC) and triglyceride (TG), as well as weakened aortic diastolic function. However, opposite changes occurred after downregulation of miR-155 or an increase in Bmal1. In addition, the microRNA.org website predicted that there were targeted binding sites between miR-155 and Bmal1, which was verified with a dual-luciferase reporter gene assay. miR-155 was able to inhibit the expression by targeting Bmal1. Moreover, a rescue experiment showed that Bmal1 hindered the promotion of miR-155 in regards to atherosclerosis. In conclusion, miR-155 induces EC apoptosis and inflammatory response, weakens aortic diastolic function, and promotes the progression of atherosclerosis through targeted inhibition of Bmal1.
Collapse
Affiliation(s)
- Shuangchao Liang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Jiqiong Hu
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Andong Zhang
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Fangkuan Li
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
53
|
Teng C, Lin C, Huang F, Xing X, Chen S, Ye L, Azevedo HS, Xu C, Wu Z, Chen Z, He W. Intracellular codelivery of anti-inflammatory drug and anti-miR 155 to treat inflammatory disease. Acta Pharm Sin B 2020; 10:1521-1533. [PMID: 32963947 PMCID: PMC7488359 DOI: 10.1016/j.apsb.2020.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/18/2020] [Accepted: 05/25/2020] [Indexed: 12/27/2022] Open
Abstract
Atherosclerosis (AS) is a lipid-driven chronic inflammatory disease occurring at the arterial subendothelial space. Macrophages play a critical role in the initiation and development of AS. Herein, targeted codelivery of anti-miR 155 and anti-inflammatory baicalein is exploited to polarize macrophages toward M2 phenotype, inhibit inflammation and treat AS. The codelivery system consists of a carrier-free strategy (drug-delivering-drug, DDD), fabricated by loading anti-miR155 on baicalein nanocrystals, named as baicalein nanorods (BNRs), followed by sialic acid coating to target macrophages. The codelivery system, with a diameter of 150 nm, enables efficient intracellular delivery of anti-miR155 and polarizes M1 to M2, while markedly lowers the level of inflammatory factors in vitro and in vivo. In particular, intracellular fate assay reveals that the codelivery system allows for sustained drug release over time after internalization. Moreover, due to prolonged blood circulation and improved accumulation at the AS plaque, the codelivery system significantly alleviates AS in animal model by increasing the artery lumen diameter, reducing blood pressure, promoting M2 polarization, inhibiting secretion of inflammatory factors and decreasing blood lipids. Taken together, the codelivery could potentially be used to treat vascular inflammation.
Collapse
Affiliation(s)
- Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chenshi Lin
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Feifei Huang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuyang Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shenyu Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, University of London, London E1 4NS, UK
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Zhengfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
54
|
MicroRNAs as sentinels and protagonists of carotid artery thromboembolism. Clin Sci (Lond) 2020; 134:169-192. [PMID: 31971230 DOI: 10.1042/cs20190651] [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: 10/18/2019] [Revised: 12/12/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Stroke is the leading cause of serious disability in the world and a large number of ischemic strokes are due to thromboembolism from unstable carotid artery atherosclerotic plaque. As it is difficult to predict plaque rupture and surgical treatment of asymptomatic disease carries a risk of stroke, carotid disease continues to present major challenges with regard to clinical decision-making and revascularization. There is therefore an imminent need to better understand the molecular mechanisms governing plaque instability and rupture, as this would allow for the development of biomarkers to identify at-risk asymptomatic carotid plaque prior to disease progression and stroke. Further, it would aid in creation of therapeutics to stabilize carotid plaque. MicroRNAs (miRNAs) have been implicated as key protagonists in various stages of atherosclerotic plaque initiation, development and rupture. Notably, they appear to play a crucial role in carotid artery thromboembolism. As the molecular pathways governing the role of miRNAs are being uncovered, we are learning that their involvement is complex, tissue- and stage-specific, and highly selective. Notably, miRNAs can be packaged and secreted in extracellular vesicles (EVs), where they participate in cell-cell communication. The measurement of EV-encapsulated miRNAs in the circulation may inform disease mechanisms occurring in the plaque itself, and therefore may serve as sentinels of unstable plaque as well as therapeutic targets.
Collapse
|
55
|
MicroRNAs as regulators of brain function and targets for treatment of epilepsy. Nat Rev Neurol 2020; 16:506-519. [DOI: 10.1038/s41582-020-0369-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
|
56
|
Liu M, Luo C, Dong J, Guo J, Luo Q, Ye C, Guo Z. CircRNA_103809 Suppresses the Proliferation and Metastasis of Breast Cancer Cells by Sponging MicroRNA-532-3p (miR-532-3p). Front Genet 2020; 11:485. [PMID: 32499818 PMCID: PMC7243809 DOI: 10.3389/fgene.2020.00485] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
Breast cancer has become one of the most serious disease threatening mankind health in the world. Accumulating studies indicated that circRNAs played an important role in the occurrence and progression of breast cancer, however, the roles of circRNA_103809 in breast cancer progression remain unclear. Therefore, in this study, we aimed to clarify the potential role and regulatory mechanism of circRNA_103809 in the development of breast cancer. Firstly, the expression level of circRNA_103809 and microRNA-532-3p (miR-532-3p) in breast cancer tissues and normal tissues were detected with the quantitative real-time polymerase chain reaction (RT-qPCR). In addition, the cell proliferation ability, metastasis ability and related pathways were identified by Cell Counting Kit-8 (CCK-8), flow cytometry, and western blot, respectively. Furthermore, the connection between circRNA_103809 and miR-532-3p was detected by dual-luciferase reporter assay. Then, our data showed that circRNA_103809 was down-regulated in breast cancer tissues in contrast to adjacent non-tumor tissues, and the relative expression level of circRNA_103809 was closely associated with distant metastasis size, TNM stage, HER-2 status and overall survival time. In addition, our in vitro assays showed that the overexpression of circRNA_103809 could significantly inhibit epithelial-mesenchymal transition (EMT) pathway, then suppress breast cancer cell proliferation and metastasis ability. Moreover, we also found that the antitumor effect induced by circRNA_103809 could be reversed with the addition of miR-532-3p mimics. Taken together, this study showed that circRNA_103809 could inhibit cell proliferation and metastasis in breast cancer by sponging miR-532-3p, and circRNA_103809 might be a prospective target of breast cancer therapy.
Collapse
Affiliation(s)
- Minfeng Liu
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Can Luo
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianyu Dong
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyun Guo
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Luo
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changsheng Ye
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaoze Guo
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, China.,The First Clinical Medical College, Southern Medical University, Guangzhou, China
| |
Collapse
|
57
|
Cheng M, Wang B, Yang M, Ma J, Ye Z, Xie L, Zhou M, Chen W. microRNAs expression in relation to particulate matter exposure: A systematic review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 260:113961. [PMID: 32006883 DOI: 10.1016/j.envpol.2020.113961] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/27/2019] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
MicroRNAs (miRNAs) are a class of small, non-coding RNAs with a post-transcriptional regulatory function on gene expression and cell processes, including proliferation, apoptosis and differentiation. In recent decades, miRNAs have attracted increasing interest to explore the role of epigenetics in response to air pollution. Air pollution, which always contains kinds of particulate matters, are able to reach respiratory tract and blood circulation and then causing epigenetics changes. In addition, extensive studies have illustrated that miRNAs serve as a bridge between particulate matter exposure and health-related effects, like inflammatory cytokines, blood pressure, vascular condition and lung function. The purpose of this review is to summarize the present knowledge about the expression of miRNAs in response to particulate matter exposure. Epidemiological and experimental studies were reviewed in two parts according to the size and source of particles. In this review, we also discussed various functions of the altered miRNAs and predicted potential biological mechanism participated in particulate matter-induced health effects. More rigorous studies are worth conducting to understand contribution of particulate matter on miRNAs alteration and the etiology between environmental exposure and disease development.
Collapse
Affiliation(s)
- Man Cheng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Yang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jixuan Ma
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zi Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Xie
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
58
|
Hung J, Scanlon JP, Mahmoud AD, Rodor J, Ballantyne M, Fontaine MAC, Temmerman L, Kaczynski J, Connor KL, Bhushan R, Biessen EAL, Newby DE, Sluimer JC, Baker AH. Novel Plaque Enriched Long Noncoding RNA in Atherosclerotic Macrophage Regulation (PELATON). Arterioscler Thromb Vasc Biol 2019; 40:697-713. [PMID: 31826651 PMCID: PMC7043732 DOI: 10.1161/atvbaha.119.313430] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) are an emergent class of molecules with diverse functional roles, widely expressed in human physiology and disease. Although some lncRNAs have been identified in cardiovascular disease, their potential as novel targets in the prevention of atherosclerosis is unknown. We set out to discover important lncRNAs in unstable plaque and gain insight into their functional relevance. Approach and Results: Analysis of RNA sequencing previously performed on stable and unstable atherosclerotic plaque identified a panel of 47 differentially regulated lncRNAs. We focused on LINC01272, a lncRNA upregulated in unstable plaque previously detected in inflammatory bowel disease, which we termed PELATON (plaque enriched lncRNA in atherosclerotic and inflammatory bowel macrophage regulation). Here, we demonstrate that PELATON is highly monocyte- and macrophage-specific across vascular cell types, and almost entirely nuclear by cellular fractionation (90%-98%). In situ hybridization confirmed enrichment of PELATON in areas of plaque inflammation, colocalizing with macrophages around the shoulders and necrotic core of human plaque sections. Consistent with its nuclear localization, and despite containing a predicted open reading frame, PELATON did not demonstrate any protein-coding potential in vitro. Functionally, knockdown of PELATON significantly reduced phagocytosis, lipid uptake and reactive oxygen species production in high-content analysis, with a significant reduction in phagocytosis independently validated. Furthermore, CD36, a key mediator of phagocytic oxLDL (oxidized low-density lipoprotein) uptake was significantly reduced with PELATON knockdown. CONCLUSIONS PELATON is a nuclear expressed, monocyte- and macrophage-specific lncRNA, upregulated in unstable atherosclerotic plaque. Knockdown of PELATON affects cellular functions associated with plaque progression.
Collapse
Affiliation(s)
- John Hung
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Jessica P Scanlon
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Amira D Mahmoud
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Julie Rodor
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Margaret Ballantyne
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Margaux A C Fontaine
- Maastricht University Medical Center, Maastricht, the Netherlands (M.A.C.F., L.T., E.A.L.B., J.C.S., A.H.B.)
| | - Lieve Temmerman
- Maastricht University Medical Center, Maastricht, the Netherlands (M.A.C.F., L.T., E.A.L.B., J.C.S., A.H.B.)
| | - Jakub Kaczynski
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Katie L Connor
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Raghu Bhushan
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Erik A L Biessen
- Maastricht University Medical Center, Maastricht, the Netherlands (M.A.C.F., L.T., E.A.L.B., J.C.S., A.H.B.)
| | - David E Newby
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.)
| | - Judith C Sluimer
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.).,Maastricht University Medical Center, Maastricht, the Netherlands (M.A.C.F., L.T., E.A.L.B., J.C.S., A.H.B.)
| | - Andrew H Baker
- From the Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (J.H., J.P.S., A.D.M., J.R., M.B., J.K., K.L.C., R.B., D.E.N., J.C.S., A.H.B.).,Maastricht University Medical Center, Maastricht, the Netherlands (M.A.C.F., L.T., E.A.L.B., J.C.S., A.H.B.)
| |
Collapse
|
59
|
Pegoraro V, Missaglia S, Marozzo R, Tavian D, Angelini C. MiRNAs as biomarkers of phenotype in neutral lipid storage disease with myopathy. Muscle Nerve 2019; 61:253-257. [PMID: 31729045 PMCID: PMC7004093 DOI: 10.1002/mus.26761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neutral lipid storage disease with myopathy (NLSDM) is a rare lipid metabolism disorder. In this study, we evaluated some circulating miRNAs levels in serum samples and the MRI of three affected siblings. METHODS Three members of one NLSDM family were identified: two brothers and one sister. Muscles of lower and right upper extremities were studied by MRI. Expression profile of miRNAs, obtained from serum samples, was detected using qRT-PCR. RESULTS Two brothers presented with progressive skeletal myopathy, while the sister had severe hepatosteatosis and diabetes. NLSDM patients showed a significant increase of muscle-specific miRNAs expression compared with healthy subjects. We found a correlation between hepatic damage and elevation of miRNAs expression profile of liver origin. CONCLUSIONS The dysregulation of miRNAs might represent an indicator of skeletal and hepatic damage and it might be useful to monitor the progression of NLSDM.
Collapse
Affiliation(s)
| | - Sara Missaglia
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | | | - Daniela Tavian
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | | |
Collapse
|
60
|
Lorente-Cebrián S, Herrera K, I. Milagro F, Sánchez J, de la Garza AL, Castro H. miRNAs and Novel Food Compounds Related to the Browning Process. Int J Mol Sci 2019; 20:E5998. [PMID: 31795191 PMCID: PMC6928892 DOI: 10.3390/ijms20235998] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 02/08/2023] Open
Abstract
Obesity prevalence is rapidly increasing worldwide. With the discovery of brown adipose tissue (BAT) in adult humans, BAT activation has emerged as a potential strategy for increasing energy expenditure. Recently, the presence of a third type of fat, referred to as beige or brite (brown in white), has been recognized to be present in certain kinds of white adipose tissue (WAT) depots. It has been suggested that WAT can undergo the process of browning in response to stimuli that induce and enhance the expression of thermogenesis: a metabolic feature typically associated with BAT. MicroRNAs (miRNAs) are small transcriptional regulators that control gene expression in a variety of tissues, including WAT and BAT. Likewise, it was shown that several food compounds could influence miRNAs associated with browning, thus, potentially contributing to the management of excessive adipose tissue accumulation (obesity) through specific nutritional and dietetic approaches. Therefore, this has created significant excitement towards the development of a promising dietary strategy to promote browning/beiging in WAT to potentially contribute to combat the growing epidemic of obesity. For this reason, we summarize the current knowledge about miRNAs and food compounds that could be applied in promoting adipose browning, as well as the cellular mechanisms involved.
Collapse
Affiliation(s)
- Silvia Lorente-Cebrián
- Department of Nutrition, Food Science and Physiology/Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (S.L.-C.)
- Navarra Institute for Health Research, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Katya Herrera
- Centro de Investigación en Nutrición y Salud Pública, Facultad de Salud Pública y Nutrición, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico; (K.H.)
- Nutrition Unit, Center for Research and Development in Health Sciences, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico
| | - Fermín I. Milagro
- Department of Nutrition, Food Science and Physiology/Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (S.L.-C.)
- Navarra Institute for Health Research, Navarra Institute for Health Research, 31008 Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, 28029 Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), University of the Balearic Islands, 07122 Palma, Spain;
- Instituto de Investigación Sanitaria Illes Balears, 07020 Palma, Spain
| | - Ana Laura de la Garza
- Centro de Investigación en Nutrición y Salud Pública, Facultad de Salud Pública y Nutrición, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico; (K.H.)
- Nutrition Unit, Center for Research and Development in Health Sciences, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico
| | - Heriberto Castro
- Centro de Investigación en Nutrición y Salud Pública, Facultad de Salud Pública y Nutrición, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico; (K.H.)
- Nutrition Unit, Center for Research and Development in Health Sciences, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Mexico
| |
Collapse
|
61
|
Cui X, Wang Z, Liu L, Liu X, Zhang D, Li J, Zhu J, Pan J, Zhang D, Cui G. The Long Non-coding RNA ZFAS1 Sponges miR-193a-3p to Modulate Hepatoblastoma Growth by Targeting RALY via HGF/c-Met Pathway. Front Cell Dev Biol 2019; 7:271. [PMID: 31781561 PMCID: PMC6856658 DOI: 10.3389/fcell.2019.00271] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022] Open
Abstract
Hepatoblastoma (HB) is the most common and aggressive malignant hepatic neoplasm in childhood and the therapeutic outcomes remain undesirable due to its recurrence and metastasis. Recently, long non-coding RNA (lncRNA) zinc finger antisense 1 (ZFAS1) has been reported to be an oncogenic gene in multiple cancers. However, the expression status and specific role of ZFAS1 involved in cancer progression of human HB remain unknown. This study aimed to identify the role of ZFAS1/miR-193a-3p/RALY axis in the development of HB. Here we showed that the expression of ZFAS1 was significantly upregulated in both HB tissues and cell lines. High ZFAS1 expression was significantly associated with aggressive tumor phenotypes and poorer overall survival in HB. In vitro and in vivo function assays indicated that silencing of ZFAS1 significantly suppressed HB cell proliferation and invasion. Furthermore, miR-193a-3p was identified to be the target of ZFAS1. Subsequently, RALY was confirmed to be regulated by miR-193a-3p/ZFAS1 axis. Mechanistically, our results indicated that the ZFAS1 participated to the progression of HB via regulating the HGF/c-Met signaling. Collectively, these data demonstrated that ZFAS1 acted as an oncogene to promote initiation and progression of HB by regulating miR-193a-3p/RALY (RALY Heterogeneous Nuclear Ribonucleoprotein) axis via HGF/c-Met Pathway, which provides an efficient marker and new therapeutic target for HB.
Collapse
Affiliation(s)
- Xichun Cui
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhifang Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwen Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dandan Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhao Li
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianming Zhu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juntao Pan
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangying Cui
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|