1
|
Xuan L, Zi-Ming J, Xue-Yan T, Wen-Xuan H, Fa-Xuan W. LncRNA MRAK052509 competitively adsorbs miR-204-3p to regulate silica dust-induced EMT process. ENVIRONMENTAL TOXICOLOGY 2024; 39:3628-3640. [PMID: 38491797 DOI: 10.1002/tox.24218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Silicosis is a systemic disease caused by long-term inhalation of free SiO2 and retention in the lungs. At present, it is still the most important occupational health hazard disease in the world. Existing studies have shown that non-coding RNA can also participate in complex fibrosis regulatory networks. However, its role in regulating silicotic fibrosis is still unclear. In this study, we constructed a NR8383/RLE-6TN co-culture system to simulate the pathogenesis of silicosis in vitro. Design of miR-204-3p mimics and inhibitors to overexpress or downregulate miR-204-3p in RLE-6TN cells. Design of short hairpin RNA (sh-RNA) to downregulate MRAK052509 in RLE-6TN cells. The regulatory mechanism of miR-204-3p and LncRNA MRAK052509 on EMT process was studied by Quantitative real-time PCR, Western blotting, Immunofluorescence and Cell scratch test. The results revealed that miR-204-3p affects the occurrence of silica dust-induced cellular EMT process mainly through regulating TGF-βRΙ, a key molecule of TGF-β signaling pathway. In contrast, Lnc MRAK052509 promotes the EMT process in epithelial cells by competitively adsorbing miR-204-3p and reducing its inhibitory effect on the target gene TGF-βRΙ, which may influence the development of silicosis fibrosis. This study perfects the targeted regulation relationship between LncRNA MRAK052509, miR-204-3p and TGF-βRΙ, and may provide a new strategy for the study of the pathogenesis and treatment of silicosis.
Collapse
Affiliation(s)
- Liu Xuan
- School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Jiao Zi-Ming
- School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Tian Xue-Yan
- School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Hu Wen-Xuan
- School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Wang Fa-Xuan
- School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| |
Collapse
|
2
|
Xiao Y, Zhu H, Lei J, Xie J, Wu K, Gu W, Ma J, wei D, Shu Z, Zhao L. MiR-182/Sestrin2 affects the function of asthmatic airway smooth muscle cells by the AMPK/mTOR pathway. J Transl Int Med 2023; 11:282-293. [PMID: 37662894 PMCID: PMC10474879 DOI: 10.2478/jtim-2023-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Background and Objectives Asthma is a chronic inflammatory airway disease and brings heavy economic and spiritual burdens to patients' families and the society. Airway smooth muscle cells (ASMCs) afect the development of asthma by secreting cytokines, growth factors, and prostates. The stress-inducing protein, Sestrin2, plays a vital role in antioxidant defense. The aim of this study is to investigate the role of Sestrin2 in asthma and its corresponding molecular mechanism. Materials and Methods Airway remodeling was induced by construction of asthma rat model. Primary ASMCs were isolated through combining tissue block adherence and enzymatic digestion and identified by immunofluorescence staining. Gene expression was measured by quantitative real-time PCR (qPCR) and western blot (WB) experiments. Cell viability, proliferation, migration, and calcium flow of ASMCs were measured by Cell Counting Kit-8 (CCK-8), 5-ethynyl-deoxyuridine (EdU), Transwell, and Fluo-3AM, respectively. The binding of miR-182 and Sestrin2 3'-untranslated region (3'-UTR) was measured by luciferase reporter system and RNA-binding protein immunoprecipitation (RIP) analysis. Results Sestrin2 expression was upregulated in asthma rat model and cell model. Overexpression of Sestrin2 enhanced the growth, migration, and calcium flow, and inversely, repression of Sestrin2 was reduced in ASMCs from the asthma group. MiR-182, one of the microRNAs (miRNAs) that possesses the potential to regulate Sestrin2, was downregulated in ASMCs from the asthma group. Further experiments revealed that Sestrin2 was inhibited by miR-182 and that overexpression of Sestrin2 reversed the miR-182-induced inhibition of the cellular progression of ASMCs from the asthma group. This study further investigated the downstream signaling pathway of Sestrin2 and found that increased expression of Sestrin2 activated 5'-adenosine monophosphate-activated protein kinase (AMPK), leading to the inactivation of mammalian target of rapamycin (mTOR) and thus promoting the growth, migration, and calcium flow of ASMCs from the asthma group. Conclusion This study investigated the role of Sestrin2 for the first time and further dissected the regulatory factor of Sestrin2, ultimately elucidating the downstream signaling pathway of Sestrin2 in asthma, providing a novel pathway, and improving the understanding of the development and progression of asthma.
Collapse
Affiliation(s)
- Yali Xiao
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - He Zhu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Jiahui Lei
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Jing Xie
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Ke Wu
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Wenbo Gu
- Department of Respiratory and Critical Care Medicine, Henan University of Traditional Chinese Medicine, Henan Provincial People’s Hospital, Zhengzhou450046, Henan Province, China
| | - Jinxin Ma
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Dongxue wei
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou450003, Henan Province, China
| | - Zhenhui Shu
- Department of Respiratory and Critical Care Medicine, Henan University of Traditional Chinese Medicine, Henan Provincial People’s Hospital, Zhengzhou450046, Henan Province, China
| | - Limin Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou450003, Henan Province, China
| |
Collapse
|
3
|
Xiao B, Li L, Yao D, Mo B. Noncoding RNAs in asthmatic airway smooth muscle cells. Eur Respir Rev 2023; 32:32/168/220184. [PMID: 37076176 PMCID: PMC10113956 DOI: 10.1183/16000617.0184-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/07/2023] [Indexed: 04/21/2023] Open
Abstract
Asthma is a complex and heterogeneous airway disease caused by genetic, environmental and epigenetic factors treated with hormones and biologics. Irreversible pathological changes to airway smooth muscle cells (ASMCs) such as hyperplasia and hypertrophy can occur in asthmatic patients. Determining the mechanisms responsible is vital for preventing such changes. In recent years, noncoding RNAs (ncRNAs), especially microRNAs, long noncoding RNAs and circular RNAs, have been found to be associated with abnormalities of the ASMCs. This review highlights recent ncRNA research into ASMC pathologies. We present a schematic that illustrates the role of ncRNAs in pathophysiological changes to ASMCs that may be useful in future research in diagnostic and treatment strategies for patients with asthma.
Collapse
Affiliation(s)
- Bo Xiao
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- These authors contributed equally to this work
| | - Liangxian Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
- These authors contributed equally to this work
| | - Dong Yao
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- These authors contributed equally to this work
| | - Biwen Mo
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Glucose and Lipid Metabolism Disorders, Guangxi Health Commission, Guilin, China
| |
Collapse
|
4
|
Yao Q, Zhang L, Liu Z, Yu L, Wang Y, Liu J, Wang Y. LncRNA MAFG-AS1-induced acute myeloid leukemia development via modulating miR-147b/HOXA9. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:19250-19258. [PMID: 36229729 DOI: 10.1007/s11356-022-23537-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/22/2021] [Indexed: 06/16/2023]
Abstract
Recent references discovered that lncRNAs acted roles in malignant cancer development. However, the role of MAFG-AS1 in acute myeloid leukemia (AML) development remains unknown. MAFG-AS1 and miR-147b were determined in AML cells and specimens using qRT-PCR assay. Cell proliferation was detected by CCK-8 analysis and flow cytometry was carried out to measure cell cycle. Luciferase reporter analysis was done to determine the mechanism of MAFG-AS1 and miR-147b. We noted that MAFG-AS1 was overexpressed in AML cells and in serum and bone narrow from AML compared with normal controls specimen. Elevated expression of MAFG-AS1 increased cell growth, cycle and EMT in AML cell HL-60 cell. MAFG-AS1 sponged miR-147b expression in HL-60 cell. Moreover, miR-147b was downregulated in AML cells and in serum and bone narrow from AML compared with normal control specimen. miR-147b was negatively correlated with MAFG-AS1 in the serum and bone narrow of AML cases. We illustrated that HOXA9 was one target of miR-147b and ectopic expression of MAFG-AS1 enhanced HOXA9 expression HL-60 cell. Forced expression of MAFG-AS1 induced cell growth, cycle, and EMT via promoting HOXA9. These data illustrated that MAFG-AS1 acted as one oncogenic gene and accelerated AML progression via modulating miR-147b/HOXA9 axis.
Collapse
Affiliation(s)
- Qiying Yao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Li Zhang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Zhengjuan Liu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Lei Yu
- Department of Infectious Disease, The Fourth Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yuchuan Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Junli Liu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Yingjie Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China.
| |
Collapse
|
5
|
Dashti F, Mirazimi SMA, Rabiei N, Fathazam R, Rabiei N, Piroozmand H, Vosough M, Rahimian N, Hamblin MR, Mirzaei H. The role of non-coding RNAs in chemotherapy for gastrointestinal cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:892-926. [PMID: 34760336 PMCID: PMC8551789 DOI: 10.1016/j.omtn.2021.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers, including colorectal, gastric, hepatic, esophageal, and pancreatic tumors, are responsible for large numbers of deaths around the world. Chemotherapy is the most common approach used to treat advanced GI cancer. However, chemoresistance has emerged as a critical challenge that prevents successful tumor elimination, leading to metastasis and recurrence. Chemoresistance mechanisms are complex, and many factors and pathways are involved. Among these factors, non-coding RNAs (ncRNAs) are critical regulators of GI tumor development and subsequently can induce resistance to chemotherapy. This occurs because ncRNAs can target multiple signaling pathways, affect downstream genes, and modulate proliferation, apoptosis, tumor cell migration, and autophagy. ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could possibly act as new targets in chemotherapy combinations to treat GI cancer and to predict treatment response.
Collapse
Affiliation(s)
- Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Fathazam
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
6
|
Wang X, Chen H, Liu J, Gai L, Yan X, Guo Z, Liu F. Emerging Advances of Non-coding RNAs and Competitive Endogenous RNA Regulatory Networks in Asthma. Bioengineered 2021; 12:7820-7836. [PMID: 34635022 PMCID: PMC8806435 DOI: 10.1080/21655979.2021.1981796] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 12/31/2022] Open
Abstract
Asthma is a chronic inflammatory disease characterized by airway remodeling and bronchial hyperresponsiveness. A variety of effector cells and cytokines jointly stimulate the occurrence of inflammatory response in asthma. Although the pathogenesis of asthma is not entirely clear, the possible roles of non-coding RNAs (ncRNAs) have been recently demonstrated. NcRNAs are non-protein-coding RNA molecules, such as circular RNAs (circRNAs), long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), which are involved in the regulation of a variety of biological processes. Mounting studies have shown that ncRNAs play pivotal roles in the occurrence and progression of asthma via competing endogenous RNA (ceRNA) regulatory networks. However, the specific mechanism and clinical application of ncRNAs and ceRNA regulatory networks in asthma have not been fully elucidated, which are worthy of further investigation. This paper comprehensively summarized the current progress on the roles of miRNAs, lncRNAs, circRNAs, and ceRNA regulatory networks in asthma, which can provide a better understanding for the disease pathogenesis and is helpful for identifying novel biomarkers for asthma.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Hui Chen
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Jingjing Liu
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Linlin Gai
- Department of Central Laboratory, The First Affiliated Hospital of Weifang Medical University/Weifang People’s Hospital, WeifangChina
| | - Xinyi Yan
- Department of Central Laboratory, The First Affiliated Hospital of Weifang Medical University/Weifang People’s Hospital, WeifangChina
| | - Zhiliang Guo
- Department of Spine Surgery, The 80th Group Army Hospital of Chinese PLA, WeifangChina
| | - Fengxia Liu
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| |
Collapse
|
7
|
Chen X, Xiao Z, Jiang Z, Jiang Y, Li W, Wang M. Schisandrin B Attenuates Airway Inflammation and Airway Remodeling in Asthma by Inhibiting NLRP3 Inflammasome Activation and Reducing Pyroptosis. Inflammation 2021; 44:2217-2231. [PMID: 34143347 DOI: 10.1007/s10753-021-01494-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 01/20/2023]
Abstract
Asthma is a chronic inflammatory disorder of the airways. Schisandrin B (SB) is the main effective component. This study investigated the effects of SB on airway inflammation and airway remodeling in asthma. The rat model of asthma was established. The rats were treated with SB to evaluate the effects of SB on airway inflammation, airway remodeling, NLRP3 inflammasome activation, and pyroptosis. Alveolar macrophages of rats were isolated, and the macrophage inflammatory model was established by lipopolysaccharide (LPS) induction. The LPS-induced macrophages were treated with SB. The binding relationship between miR-135a-5p and TPRC1 was analyzed. LPS + SB-treated macrophages were transfected with miR-135a-5p inhibitor. The expressions of key factors of the STAT3/NF-κB pathway were detected. SB reduced airway inflammation and airway remodeling in asthmatic rats. SB inhibited NLRP3 inflammasome activation and reduced pyroptosis in asthmatic rats and LPS-induced macrophages. SB reversely regulated the miR-135a-5p/TRPC1 axis. Downregulation of miR-135a-5p attenuated the inhibitory effect of SB on NLRP3 inflammasome activation. SB inhibited the STAT3/NF-κB pathway via the miR-135a-5p/TRPC1 axis. In conclusion, SB inhibited NLRP3 inflammasome activation and reduced pyroptosis via the miR-135a-5p/TRPC1/STAT3/NF-κB axis, thus alleviating airway inflammation and airway remodeling in asthma. This study may confer novel insights for the management of asthma.
Collapse
Affiliation(s)
- Xiufeng Chen
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| | - Zhen Xiao
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China.
| | - Zhiyan Jiang
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China.
| | - Yonghong Jiang
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| | - Wen Li
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| | - Mingjing Wang
- Department of Pediatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| |
Collapse
|
8
|
Soni DK, Biswas R. Role of Non-Coding RNAs in Post-Transcriptional Regulation of Lung Diseases. Front Genet 2021; 12:767348. [PMID: 34819948 PMCID: PMC8606426 DOI: 10.3389/fgene.2021.767348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Non-coding RNAs (ncRNAs), notably microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have recently gained increasing consideration because of their versatile role as key regulators of gene expression. They adopt diverse mechanisms to regulate transcription and translation, and thereby, the function of the protein, which is associated with several major biological processes. For example, proliferation, differentiation, apoptosis, and metabolic pathways demand fine-tuning for the precise development of a specific tissue or organ. The deregulation of ncRNA expression is concomitant with multiple diseases, including lung diseases. This review highlights recent advances in the post-transcriptional regulation of miRNAs and lncRNAs in lung diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, and idiopathic pulmonary fibrosis. Further, we also discuss the emerging role of ncRNAs as biomarkers as well as therapeutic targets for lung diseases. However, more investigations are required to explore miRNAs and lncRNAs interaction, and their function in the regulation of mRNA expression. Understanding these mechanisms might lead to early diagnosis and the development of novel therapeutics for lung diseases.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
9
|
Subramanian H, Hashem T, Bahal D, Kammala AK, Thaxton K, Das R. Ruxolitinib Ameliorates Airway Hyperresponsiveness and Lung Inflammation in a Corticosteroid-Resistant Murine Model of Severe Asthma. Front Immunol 2021; 12:786238. [PMID: 34777398 PMCID: PMC8586657 DOI: 10.3389/fimmu.2021.786238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022] Open
Abstract
Asthma prevalence has increased considerably over the decades and it is now considered as one of the most common chronic disorders in the world. While the current anti-asthmatic therapies are effective for most asthma patients, there are 5-10% subjects whose disease is not controlled by such agents and they account for about 50% of the asthma-associated healthcare costs. Such patients develop severe asthma (SA), a condition characterized by a dominant Th1/Th17 cytokine response that is accompanied by Type 2 (T2)-low endotype. As JAK (Janus Kinase) signaling is very important for the activation of several cytokine pathways, we examined whether inhibition of JAKs might lessen the clinical and laboratory manifestations of SA. To that end, we employed a recently described murine model that recapitulates the complex immune response identified in the airways of human SA patients. To induce SA, mice were sensitized with house dust mite extract (HDME) and cyclic (c)-di-GMP and then subsequently challenged with HDME and a lower dose of c-di-GMP. In this model, treatment with the JAK inhibitor, Ruxolitinib, significantly ameliorated all the features of SA, including airway hyperresponsiveness and lung inflammation as well as total IgE antibody titers. Thus, these studies highlight JAKs as critical targets for mitigating the hyper-inflammation that occurs in SA and provide the framework for their incorporation into future clinical trials for patients that have severe or difficult-to manage asthma.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Tanwir Hashem
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Devika Bahal
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Ananth K Kammala
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Kanedra Thaxton
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Rupali Das
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
10
|
Zhou H, Long C, Liu P, Chen Y, Luo L, Xiao Z. Long non-coding RNA TUG1 accelerates abnormal growth of airway smooth muscle cells in asthma by targeting the miR-138-5p/E2F3 axis. Exp Ther Med 2021; 22:1229. [PMID: 34539825 DOI: 10.3892/etm.2021.10663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease. The present study aimed to explore the effect of the long non-coding RNA taurine-upregulated gene 1 (TUG1) on the viability and migration of airway smooth muscle cells (ASMCs) in asthma. Rat asthma models were constructed with ovalbumin sensitization and challenge and the level of serum immunoglobulin E (IgE) and the rates of inspiratory and expiratory resistance were measured. Reverse transcription-quantitative PCR was also performed to determine the expression levels of TUG1. Platelet-derived growth factor-BB (PDGF-BB)-treated ASMCs were then used as a cell model of asthma. The viability and migratory abilities of ASMCs were analysed with the MTT and Transwell assays. Additionally, a dual-luciferase reporter assay was used to confirm the relationship between TUG1 and microRNA (miR)-138-5p and between transcription factor E2F3 and miR-138-5p. The expression of TUG1, level of serum IgE, inspiratory resistance and expiratory resistance were clearly increased in the rat asthma model in comparison with controls. Knockdown of TUG1 the viability and migration of PDGF-BB-induced ASMCs and reduced the inspiratory and expiratory resistances. In addition, TUG1 functioned as a bait of miR-138-5p, and miR-138-5p modulated E2F3 expression. Knockdown of E2F3 hindered the abnormal growth of ASMCs. Moreover, miR-138-5p inhibition or E2F3 overexpression reversed the inhibitory effects of TUG1 knockdown on viability and migration of PDGF-BB-induced ASMCs. The TUG1/miR-138-5p/E2F3 regulatory axis appeared to play a critical role in accelerating the viability and migration of ASMCs and may therefore have a role in asthma.
Collapse
Affiliation(s)
- Haiyin Zhou
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Caixia Long
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Pingping Liu
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Yanying Chen
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Lan Luo
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Zhenghui Xiao
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
11
|
Chen L, Shi Q, Fan B, Cai Y. Role of lncRNA BCYRN1 in trophoblast cell physiology and pathogenesis of preeclampsia. Exp Ther Med 2021; 22:1137. [PMID: 34466147 PMCID: PMC8383326 DOI: 10.3892/etm.2021.10571] [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: 09/21/2020] [Accepted: 04/12/2021] [Indexed: 11/20/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) may play a key role in the pathogenesis of preeclampsia (PE). The present study investigated the role of the lncRNA brain cytoplasmic RNA 1 (BCYRN1) in PE. A total of 30 patients with severe PE (SPE) and 30 patients with mild PE (MPE) were recruited, whilst 30 healthy pregnant individuals were enrolled as controls. Placental tissues of enrolled subjects were collected after delivery. The clinical data of pregnant women and newborns were recorded before the correlation between BCYRN1 expression and clinical characteristics was analyzed. Furthermore, HTR-8/SVneo cells were transfected with BCYRN1 overexpression plasmids and BCYRN1 small interfering (si)RNA. Cell Counting Kit-8, Transwell, flow cytometry and tube formation assays were used to detect the function of BCYRN1 in HTR-8/SVneo cells. Reverse transcription-quantitative PCR was used to detect BCYRN1 expression in placental tissues and HTR-8/SVneo cells. Western blotting was used to detect the protein expression levels of Wnt1 and β-catenin. BCYRN1 expression was lower in placenta with mild PE compared with in normal placenta, and was in turn lower in placenta with severe PE. BCYRN1 was negatively correlated with systolic blood pressure and 24-h urinary protein in patients with PE. BCYRN1 siRNA inhibited cell viability, migration, invasion and tube forming abilities whilst increasing apoptosis. By contrast, BCYRN1 overexpression conferred opposite effects. The levels of Wnt1 and β-catenin expression in the cells and placental tissues were next measured. Cells overexpressing BCYRN1 were further treated with the Wnt pathway inhibitor XAV939. Wnt1 and β-catenin expression were elevated when BCYRN1 was overexpressed, but were decreased after BCYRN1 knockdown. XAV939 attenuated the effect of BCYRN1 overexpression on HTR-8/SVneo cells. Overall, the resulted indicated that upregulation of BCYRN1 increased trophoblast viability and prevented apoptosis by activating the Wnt/β-catenin pathway to delay PE onset.
Collapse
Affiliation(s)
- Liping Chen
- Antenatal Diagnosis Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China.,School of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qi Shi
- Department of Gynecology and Obstetrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Bo Fan
- Department of Gynecology and Obstetrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yan Cai
- Antenatal Diagnosis Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
12
|
Duan XJ, Zhang X, Ding N, Zhang JY, Chen YP. LncRNA NEAT1 regulates MMP-16 by targeting miR-200a/b to aggravate inflammation in asthma. Autoimmunity 2021; 54:439-449. [PMID: 34448644 DOI: 10.1080/08916934.2021.1966769] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Asthma is a common respiratory disease which is characterized by persistent airway inflammation. Abnormal expression of long non-coding RNAs (lncRNAs) is observed in asthma. However, whether lncRNA nuclear-enriched abundant transcript 1 (NEAT1) regulates asthmatic inflammation and its mechanism still needs to be further investigated. The expression levels of inflammatory factors (tumour necrosis factor (TNF)-α, interleukin (IL)-4, IL-13, and IL-10) were detected using reverse transcription quantitative real-time PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). MTT and flow cytometry assays were employed to determine cell proliferation and apoptosis, respectively. Dual luciferase reporter assay was performed to verify the relationship between miR-200a/b and MMP-16 or NEAT1. NEAT1 silencing markedly reduced TNF-α, IL-4, and IL-13 levels, while elevated IL-10 expression, suppressed cell proliferation, and promoted cell apoptosis. However, NEAT1 overexpression elicited the opposite effects on cell proliferation and inflammation cytokines secretion. What is more, NEAT1 negatively regulated miR-200a/b expression, and MMP16 was a target gene of miR-200a/b. miR-200a/b overexpression suppressed inflammation, cell proliferation, and enhanced cell apoptosis through regulation of MMP16. Moreover, MMP-16 overexpression or miR-200a/b inhibition abolished the regulatory effect of sh-NEAT1 on cell inflammation and apoptosis in BEAS-2B cells. NEAT1 acted as the role of sponge for miR-200a/b to regulate MMP-16 expression, thereby promoting asthma progression, suggesting that NEAT1 might have great potential as therapeutic target for asthma.
Collapse
Affiliation(s)
- Xiao-Jun Duan
- Respiratory Department, Hunan Children's Hospital, Changsha, PR China
| | - Xi Zhang
- Respiratory Department, Hunan Children's Hospital, Changsha, PR China
| | - Niu Ding
- Respiratory Department, Hunan Children's Hospital, Changsha, PR China
| | - Ji-Yan Zhang
- Respiratory Department, Hunan Children's Hospital, Changsha, PR China
| | - Yan-Ping Chen
- Respiratory Department, Hunan Children's Hospital, Changsha, PR China
| |
Collapse
|
13
|
MiR-182/Sestrin2 affects the function of asthmatic airway smooth muscle cells by the AMPK/mTOR pathway. J Transl Int Med 2021. [DOI: 10.2478/jtim-2021-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Background and Objectives
Asthma is a chronic inflammatory airway disease and brings heavy economic and spiritual burdens to patients’ families and the society. Airway smooth muscle cells (ASMCs) affect the development of asthma by secreting cytokines, growth factors, and prostates. The stress-inducing protein, Sestrin2, plays a vital role in antioxidant defense. The aim of this study is to investigate the role of Sestrin2 in asthma and its corresponding molecular mechanism.
Materials and Methods
Airway remodeling was induced by construction of asthma rat model. Primary ASMCs were isolated through combining tissue block adherence and enzymatic digestion and identified by immunofluorescence staining. Gene expression was measured by quantitative real-time PCR (qPCR) and western blot (WB) experiments. Cell viability, proliferation, migration, and calcium flow of ASMCs were measured by Cell Counting Kit-8 (CCK-8), 5-ethynyl-deoxyuridine (EdU), Transwell, and Fluo-3AM, respectively. The binding of miR-182 and Sestrin2 3′-untranslated region (3′-UTR) was measured by luciferase reporter system and RNA-binding protein immunoprecipitation (RIP) analysis.
Results
Sestrin2 expression was upregulated in asthma rat model and cell model. Overexpression of Sestrin2 enhanced the growth, migration, and calcium flow, and inversely, repression of Sestrin2 was reduced in ASMCs from the asthma group. MiR-182, one of the microRNAs (miRNAs) that possesses the potential to regulate Sestrin2, was downregulated in ASMCs from the asthma group. Further experiments revealed that Sestrin2 was inhibited by miR-182 and that overexpression of Sestrin2 reversed the miR-182–induced inhibition of the cellular progression of ASMCs from the asthma group. This study further investigated the downstream signaling pathway of Sestrin2 and found that increased expression of Sestrin2 activated 5′-adenosine monophosphate-activated protein kinase (AMPK), leading to the inactivation of mammalian target of rapamycin (mTOR) and thus promoting the growth, migration, and calcium flow of ASMCs from the asthma group.
Conclusion
This study investigated the role of Sestrin2 for the first time and further dissected the regulatory factor of Sestrin2, ultimately elucidating the downstream signaling pathway of Sestrin2 in asthma, providing a novel pathway, and improving the understanding of the development and progression of asthma.
Collapse
|
14
|
Wang WL, Luo XM, Zhang Q, Zhu HQ, Chen GQ, Zhou Q. The lncRNA PVT1/miR-590-5p/FSTL1 axis modulates the proliferation and migration of airway smooth muscle cells in asthma. Autoimmunity 2021; 54:138-147. [PMID: 33825599 DOI: 10.1080/08916934.2021.1897977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Asthma is a prevalent chronic inflammatory airway disease that is characterised by airway remodelling and airway hyperresponsiveness. Abnormal proliferation and migration of airway smooth muscle cells (ASMCs) contribute to airway remodelling in asthma. However, the molecular mechanism underlying an increased ASMC mass in asthma remains elusive. Herein, we aimed at investigating the regulation of lncRNA PVT1 on ASMCs and focussing on the mechanism in the proliferation and migration. METHODS Expression levels of lncRNA PVT1 and miR-590-5p in the serum collected from 24 children with asthma and 10 control children were determined by qRT-PCR. ASMCs proliferation and migration prior to and post platelet-derived growth factor subunit B (PDGF-BB) stimulation were examined by CCK-8 test and transwell assay. Dual-luciferase reporter assay was performed to determine miR-590-5p interaction with lncRNA PVT1 and follistatin-like 1 (FSTL1). Expression of lncRNA PVT1, miR-590-5p, FSTL1, C-Myc, cyclin D1, and cyclin-dependent kinase 1 (CDK1) was tested by quantitative real-time PCR (qRT-PCR) and immunoblotting analysis. RESULTS The expression level of lncRNA PVT1 was higher but the expression level of miR-590-5p was lower in the serum of children with asthma than in control children. The expression level of lncRNA PVT1 was negatively correlated with the expression level of miR-590-5p in asthma. LncRNA PVT1 was upregulated upon PDGF-BB stimulation. LncRNA PVT1 knockdown by its specific shRNA repressed PDGF-BB-induced promotion of proliferation and migration in ASMCs and triggered an elevated miR-590-5p along with declined C-Myc, cyclin D1, and CDK1. The effects of lncRNA PVT1 knockdown on PDGF-BB-induced ASMCs were lost upon miR-590-5p inhibition. MiR-590-5p targeted FSTL1 gene and declined its expression, thus suppressing ASMC proliferation and migration following PDGF-BB stimulation and downregulating C-Myc, cyclin D1, and CDK1 expressions. The effects of miR-590-5p on PDGF-BB-induced ASMCs were lost upon FSTL1 overexpression. CONCLUSION These results support the notion that the lncRNA PVT1/miR-590-5p/FSTL1 axis modulates ASMCs proliferation and migration following PDGF-BB stimulation, providing a potential therapeutic target to attenuate airway remodelling in asthma.
Collapse
Affiliation(s)
- Wen-Lan Wang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Xiao-Ming Luo
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Hai-Qiao Zhu
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Guo-Qing Chen
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhou
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| |
Collapse
|
15
|
Liu Y, Li L, Wu X, Qi H, Gao Y, Li Y, Chen D. MSC-AS1 induced cell growth and inflammatory mediators secretion through sponging miR-142-5p/DDX5 in gastric carcinoma. Aging (Albany NY) 2021; 13:10387-10395. [PMID: 33819916 PMCID: PMC8064188 DOI: 10.18632/aging.202800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022]
Abstract
Emerging studies have noted that dysregulated lncRNAs are implicated in cancer progression and tumorigenesis. We first showed that MSC-AS1 was overexpressed in gastric cancer (GC) cells (HGC-27, MKN-45, SGC-7901 and MGC-803 cells) compared with GES cells. We observed that MSC-AS1 was upregulated in GC specimens compared with paired normal specimens. MSC-AS1 increased cell growth and cycle progression. Moreover, the overexpression of MSC-AS1 enhanced the secretion of the inflammatory mediators IL-1β, IL-6 and TNF-α. We found that the overexpression of MSC-AS1 inhibited the expression of miR-142-5p in HGC-27 cells. We noted that DDK5 was a target gene of miR-142-5p. The overexpression of miR-142-5p suppressed the luciferase activity of wild-type DDX5, but the luciferase activity of the mutant DDX5 was not changed. We showed that miR-142-5p was downregulated in GC specimens compared with paired normal specimens. MSC-AS1 expression was inversely correlated with miR-142-5p expression in GC specimens. MSC-AS1 induced cell growth, cell cycle progression and inflammatory mediator secretion by modulating DDX5. These results showed that MSC-AS1 functions as a key oncogene in the development of GC.
Collapse
Affiliation(s)
- Yan Liu
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Lin Li
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Xiaoxu Wu
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Haiyan Qi
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Yan Gao
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Yanqi Li
- Department of Oncology, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| | - Da Chen
- Department of General Practice, The Fourth Hospital of China Medical University, Liaoning, Shengyang 110032, China
| |
Collapse
|
16
|
Zhang K, Wang Q, Zhong B, Gong Z. LUCAT1 as an oncogene in tongue squamous cell carcinoma by targeting miR-375 expression. J Cell Mol Med 2021; 25:4543-4550. [PMID: 33787082 PMCID: PMC8107098 DOI: 10.1111/jcmm.15982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/18/2020] [Accepted: 04/27/2020] [Indexed: 01/17/2023] Open
Abstract
Emerging studies suggested that lncRNAs play a crucial molecular role in cancer development and progression. LncRNA LUCAT1 has been proved as oncogenic molecular in lung cancer, glioma, osteosarcoma, renal carcinoma and oesophageal squamous cell carcinoma. However, its roles and function mechanisms in tongue squamous cell carcinoma (TSCC) are still unknown. We showed that the expression of LUCAT1 was up‐regulated in the TSCC cells and tissues and the higher LUCAT1 expression was associated with the poor overall survival (OS). Knockdown expression of LUCAT1 suppressed TSCC cell proliferation, cycle and migration. In addition, we demonstrated that miR‐375 overexpression inhibited the luciferase activity of LUCAT1 wild‐type and knockdown LUCAT1 promoted the miR‐375 expression in TSCC cell. Furthermore, we indicated that miR‐375 expression was down‐regulated in the TSCC cell lines and tissues and the lower expression of miR‐375 was associated with poor OS. The expression of miR‐375 was inversely correlated with LUCAT1 expression in the TSCC tissues. Knockdown LUCAT1 promoted TSCC cell proliferation, cell cycle and migration partly through regulating miR‐375 expression. In summary, this study suggested the tumorigenic effect of lncRNA LUCAT1 in TSCC cells by targeting miR‐375 expression.
Collapse
Affiliation(s)
- Kai Zhang
- Center of Stomatology, China-Japan Friendship Hospital, Beijing, China
| | - Qibao Wang
- Department of Endodontics, Jinan Stomatological Hospital, Shandong, China
| | - Bo Zhong
- Center of Stomatology, China-Japan Friendship Hospital, Beijing, China
| | - Zuode Gong
- Department of Endodontics, Jinan Stomatological Hospital, Shandong, China
| |
Collapse
|
17
|
Nazimek K. The complex functions of microRNA-150 in allergy, autoimmunity and immune tolerance. AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>At present, special efforts are being made to develop the strategies allowing for activation of long-lasting antigen-specific immune tolerance in therapy of allergic and autoimmune diseases. Some of these therapeutic approaches are aimed at modulating cell functions at genetic level by using miRNA-based and miRNA-targeting treatments. Simultaneously, the crucial role of extracellular vesicles as natural miRNA conveyors is highlighted for induction of antigen-specific immune tolerance, especially that they appear to be easily manipulatable for therapeutic applications. Among other immune-related miRNAs, miR-150 is getting special attention as it is differently expressed by immune cells at various stages of their maturation and differentiation. In addition, miR-150 is involved in different signaling cascades orchestrating humoral and cell-mediated mechanisms of both innate and adaptive immune responses. Therefore, miR-150 is considered a master regulator of immunity in mammals. Currently, physiological miR-150-dependent regulatory circuits and causes of their malfunctioning that underlie the pathogenesis of allergic and autoimmune disorders are being unraveled. Thus, present review summarizes the current knowledge of the role of miR-150 in the pathogenesis and complications of these diseases. Furthermore, the involvement of miR-150 in regulation of immune responses to allergens and self-antigens and in induction of antigen-specific immune tolerance is discussed with the special emphasis on the therapeutic potential of this miRNA.</p>
</abstract>
Collapse
|
18
|
Wu X, Yan L, Liu Y, Shang L. LncRNA ROR1-AS1 accelerates osteosarcoma invasion and proliferation through modulating miR-504. Aging (Albany NY) 2020; 13:219-227. [PMID: 33401251 PMCID: PMC7835057 DOI: 10.18632/aging.103498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
Long non-coding RNAs (LncRNAs) play vital roles in the progression and development of tumors. However, the functional role of ROR1-AS1 in osteosarcoma has not been investigated. We found that ROR1-AS1 was upregulated in osteosarcoma tissues compared to non-tumor samples. Elevated expression of ROR1-AS1 promoted cyclin D1, PCNA and ki-67 expression and increased cell cycle and growth in MG-63 cell. Moreover, overexpression of ROR1-AS1 induced cell migration in MG-63 cell, promoting N-cadherin and vimentin expression and inhibiting E-cadherin expression. Dual-luciferase assay proved that ROR1-AS1 served as one sponge for miR-504 and ROR1-AS1 overexpression suppressed miR-504 expression in MG-63 cell. ROR1-AS1 expression was lower in osteosarcoma tissues compared to non-tumor samples. Pearson's correlation assay showed a negative correlation between miR-504 and ROR1-AS1 expression. MiR-504 overexpression partly abrogated ROR1-AS1-induced effects on osteosarcoma cell migration and proliferation. These data implied that ROR1-AS1 played as an oncogene and might be a new treatment target for osteosarcoma.
Collapse
Affiliation(s)
- Xiangkun Wu
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang 473000, Henan, China
| | - Lihua Yan
- Department of Medical Oncology, Nanyang Second People's Hospital, Nanyang 473000, Henan, China
| | - Yongxi Liu
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang 473000, Henan, China
| | - Lilin Shang
- Department of Orthopaedic Surgery, Nanyang Second People's Hospital, Nanyang 473000, Henan, China
| |
Collapse
|
19
|
Zhou Y, Men L, Sun Y, Wei M, Fan X. Pharmacodynamic effects and molecular mechanisms of lignans from Schisandra chinensis Turcz. (Baill.), a current review. Eur J Pharmacol 2020; 892:173796. [PMID: 33345853 DOI: 10.1016/j.ejphar.2020.173796] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Fruit of Schisandra chinensis Turcz. (Baill.) (S. chinensis) is a traditional herbal medicine widely used in China, Korea, and many other east Asian countries. At present, S. chinensis commonly forms Chinese medicinal formulae with other herbal medicines to treat liver disease and neurological disease in clinical. Modern researches indicated that lignans were the main active ingredients of S. chinensis with high content and novel dibenzocyclooctadiene skeletal structure, exhibited considerable antioxidant, anti-inflammatory, and neuroprotective properties. Additionally, some of these lignans also showed certain potentials in anti-cancer, anti-fibrosis, and other effects. In the current review, we summarize literature reported lignans from S. chinensis in the past five years, and highlight the molecular mechanisms of lignans in exerting their biological functions. Also, we point out some deficiencies of existing researches and discuss the future direction of lignans study.
Collapse
Affiliation(s)
- Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Lihui Men
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yunxia Sun
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengying Wei
- Natural Medicine Institute of Zhejiang YangShengTang Co., Hangzhou, 310000, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
20
|
Yang L, Zhang Y, Bao J, Feng JF. Long non-coding RNA BCYRN1 exerts an oncogenic role in colorectal cancer by regulating the miR-204-3p/KRAS axis. Cancer Cell Int 2020; 20:453. [PMID: 32944001 PMCID: PMC7491190 DOI: 10.1186/s12935-020-01543-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/05/2020] [Indexed: 12/24/2022] Open
Abstract
Background It has been well documented that long non-coding RNAs (lncRNAs) regulate numerous characteristics of cancer, including proliferation, migration, metastasis, apoptosis, and even metabolism. LncRNA BCYRN1 (BCYRN1) is a newly identified brain cytoplasmic lncRNA with 200 nucleotides that was discovered to be highly expressed in tumour tissues, including those of hepatocellular carcinoma, gastric cancer and lung cancer. However, the roles of BCYRN1 in colorectal cancer (CRC) remain obscure. This study was designed to reveal the role of BCYRN1 in the occurrence and progression of CRC. Methods RT-PCR was used to detect the expression level of BCYRN1 in tumour tissues and CRC cell lines. BCYRN1 was knocked down in CRC cells, and cell proliferation changes were evaluated by cell counting kit-8 (CCK-8), 5-ethynyl-2′-deoxyuridine (EdU), and Ki-67 and proliferating cell nuclear antigen (PCNA) expression assays. Cell migration and invasion changes were evaluated by wound healing, Transwell and invasion-related protein expression assays. Flow cytometry analysis was used to assess whether BCYRN1 regulates the apoptosis of CRC cells. The dual luciferase reporter gene detects the competitive binding of BCYRN1 to miR-204-3p. In vivo experiments were performed to evaluate the effect of BCYRN1 on tumour development. TargetScan analysis and dual luciferase reporter gene assays were applied to detect the target gene of miR-204-3p. Rescue experiments verified that BCYRN1 affects CRC by regulating the effect of miR-204-3p on KRAS. Results We found that compared with normal tissues and human intestinal epithelial cells (HIECs), CRC tumour tissues and cell lines had significantly increased BCYRN1 levels. We further determined that knockdown of BCYRN1 inhibited the proliferation, migration, and invasion and promoted the apoptosis of CRC cells. In addition, bioinformatics analysis and dual luciferase reporter assay showed that BCYRN1 served as a competitive endogenous RNA (ceRNA) to regulate the development of CRC through competitively binding to miR-204-3p. Further studies proved that overexpression of miR-204-3p reversed the effects of BCYRN1 on CRC. Next, TargetScan analysis and dual luciferase reporter assay indicated that KRAS is a target gene of miR-204-3p and is negatively regulated by miR-204-3p. A series of rescue experiments showed that BCYRN1 affected the occurrence and development of CRC by regulating the effects of miR-204-3p on KRAS. In addition, tumorigenesis experiments in a CRC mouse model confirmed that BCYRN1 downregulation effectively inhibited tumour growth. Conclusions Our findings suggest that BCYRN1 plays a carcinogenic role in CRC by regulating the miR-204-3p/KRAS axis.
Collapse
Affiliation(s)
- Liu Yang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yinan Zhang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Jun Bao
- Department of Chemotherapy, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting, Nanjing, China
| | - Ji-Feng Feng
- Department of Chemotherapy, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting, Nanjing, China
| |
Collapse
|
21
|
Zhu X, Wei Y, Dong J. Long Noncoding RNAs in the Regulation of Asthma: Current Research and Clinical Implications. Front Pharmacol 2020; 11:532849. [PMID: 33013382 PMCID: PMC7516195 DOI: 10.3389/fphar.2020.532849] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/25/2020] [Indexed: 01/21/2023] Open
Abstract
Asthma is a chronic airway inflammatory disorder related to variable expiratory airflow limitation, leading to wheeze, shortness of breath, chest tightness, and cough. Its characteristic features include airway inflammation, airway remodeling and airway hyperresponsiveness. The pathogenesis of asthma remains extremely complicated and the detailed mechanisms are not clarified. Long noncoding RNAs (lncRNAs) have been reported to play a prominent role in asthma and function as modulators of various aspects in pathological progress of asthma. Here, we summarize recent advances of lncRNAs in asthma pathogenesis to guide future researches, clinical treatment and drug development, including their regulatory functions in the T helper (Th) 1/Th2 imbalance, Th17/T regulatory (Treg) imbalance, eosinophils dysfunction, macrophage polarization, airway smooth muscle cells proliferation, and glucocorticoid insensitivity.
Collapse
Affiliation(s)
- Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Chen X, Li Z, Xu D, Li S. LINC01121 induced intervertebral disc degeneration via modulating miR-150-5p/MMP16 axis. J Gene Med 2020; 22:e3231. [PMID: 32436632 DOI: 10.1002/jgm.3231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/02/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Growing evidence indicates that Long noncoding RNAs contribute to cell differentiation, invasion, metabolism, proliferation and metastasis. However, the potential role of LINC01121 in progression of intervertebral disc degeneration (IDD) remains unclear. METHODS LINC01121, matrix metalloprotease (MMP)-16 and miR-150-5p expression was determined by a quantitative-reverse transcriptase-polymerase chain reaction assay. Inflammatory cytokines level was measured by an enzyme-linked immunosorbent assay and cell counting kit-8 analysis was used to assess cell proliferation. MMP-16-specific binding with miR-150-5p was verified with a luciferase reporter assay. RESULTS We noted that interleukin (IL)-1β and tumor necrosis factor (TNF)-α treatment enhanced LINC01121 and MMP-16 expression in nucleus pulposus (NP) cells. LINC01121 was higher in IDD specimens compared to that in control specimens. Higher expression of LINC01121 was correlated with disc degeneration degree. Ectopic expression of LINC01121 enhanced cell proliferation and promoted ki-67, MMP-3 and ADAMTS5 expression and also suppressed collagen II expression in NP cells. We observed that overexpression of LINC01121 increased the secretion of three inflammatory cytokines, including IL-6, TNF-α and IL-1β. We found that ectopic expression of LINC01121 decreased the miR-150-5p level in NP cells. Luciferase reporter data confirmed that MMP-16 was one direct target of miR-150-5p. Overexpression of miR-150-5p inhibited MMP-16 level and elevated the expression of LINC01121 enhanced MMP-16 level. We also found that MMP-16 was up-regulated in IDD specimens compared to that in control specimens. Higher expression of MMP-16 was correlated with disc degeneration degree. Interestingly, MMP-16 expression was positively related to LINC01121 in IDD specimens. Finally, overexpression of LINC01121 regulated cell growth, extracellular matrix degradation and inflammatory cytokine secretion via modulating MMP-16. CONCLUSIONS our data suggested LINC01121 may be a new therapeutic target for IDD.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopaedic, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate school of Peking Union Medical College, Beijing, 100042, China
| | - Zheng Li
- Department of Orthopaedic, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate school of Peking Union Medical College, Beijing, 100042, China
| | - Derong Xu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shugang Li
- Department of Orthopaedic, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate school of Peking Union Medical College, Beijing, 100042, China
| |
Collapse
|
23
|
Hashem T, Kammala AK, Thaxton K, Griffin RM, Mullany K, Panettieri RA, Subramanian H, Das R. CD2 Regulates Pathogenesis of Asthma Induced by House Dust Mice Extract. Front Immunol 2020; 11:881. [PMID: 32477356 PMCID: PMC7235426 DOI: 10.3389/fimmu.2020.00881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/16/2020] [Indexed: 01/13/2023] Open
Abstract
Characteristic of allergic asthma, CD4+Th2 lymphocytes secrete Th2 cytokines, interleukin (IL)-4, IL-13, and IL-5 that mediate the inflammatory immune response. Surface expression of CD2 and its ligand, CD58, is increased on the monocytes and eosinophils of asthma patients, which correlate with elevated serum IgE levels, suggesting that CD2 may contribute to allergic airway inflammation. Using a murine model of asthma, we observed that house dust mice extract (HDME)-exposed Balb/c mice have increased airway hyperresponsiveness (AHR), lung inflammation, goblet cell hyperplasia, and elevated levels of Th2 cytokines in the lungs, as well as increased serum IgE levels as compared to the control mice. In contrast, with the exception of serum IgE levels, all the other parameters were significantly reduced in HDME-treated Cd2-/- mice. Interestingly, Il13 but not Il4 or Il5 gene expression in the lungs was dramatically decreased in HDME-exposed Cd2-/- mice. Of note, the gene expression of IL-13 downstream targets (Muc5b and Muc5ac) and high affinity IL-13Rα2 were upregulated in the lungs of HDME-exposed Balb/c mice but were significantly reduced in HDME-exposed Cd2-/- mice. Consistently, gene expression of microRNAs regulating mucin production, inflammation, airway smooth muscle cell proliferation and IL-13 transcripts were increased in the lungs of HDME-exposed Cd2-/- mice. Given the established role of IL-13 in promoting goblet cell hyperplasia, lung inflammation and AHR in allergic asthma, our studies reveal a unique role for CD2 in the regulation of Th2-associated allergic asthma.
Collapse
Affiliation(s)
- Tanwir Hashem
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Ananth K. Kammala
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Kanedra Thaxton
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Ryan M. Griffin
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Kellie Mullany
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Reynold A. Panettieri
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, United States
| | - Hariharan Subramanian
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Rupali Das
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
24
|
Ying J, Yang J, Liu Y. LncARSR promotes non-small-cell lung cancer progression via regulating PTEN/Akt. Am J Transl Res 2020; 12:857-866. [PMID: 32269718 PMCID: PMC7137040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/28/2019] [Indexed: 06/11/2023]
Abstract
LncRNAs have been suggested to be key modulators in many biological and pathological processes. LncARSR, a recently identified lncRNA, plays crucial roles in the progression of several cancers. However, the role of lncARSR in NSCLC is uninvestigated. In the present study, it was demonstrated that lncARSR expression was higher in NSCLC tissues than in noncancerous tissues. The expression of lncARSR was higher in four NSCLC cell lines than in a normal lung bronchial epithelial line. Further investigation demonstrated that increased lncARSR expression promoted NSCLC cell migration and growth and induced epithelial-mesenchymal transition in A549 cells. Moreover, ectopic expression of lncARSR suppressed PTEN expression and induced Akt phosphorylation in A549 cells. The expression level of PTEN was higher in NSCLC samples than in adjacent non-tumor specimens. PTEN expression was negatively correlated with lncARSR in NSCLC specimens. Furthermore, we demonstrated that overexpression of lncARSR induced NSCLC cell growth and migration via regulating the PTEN/Akt signaling pathway. These results suggest that lncARSR acts as an oncogene in NSCLC development and could serve as a new potential therapeutic target.
Collapse
Affiliation(s)
- Jie Ying
- Clinical Research Center, Xuyi People’s HospitalXuyi 211700, Jiangsu, China
| | - Jian Yang
- Clinical Research Center, Xuyi People’s HospitalXuyi 211700, Jiangsu, China
| | - Yingzi Liu
- Department of Occupational Disease, Daqing People’s HospitalDaqing 163316, Heilongjiang, China
| |
Collapse
|
25
|
Lang N, Wang C, Zhao J, Shi F, Wu T, Cao H. Long non‑coding RNA BCYRN1 promotes glycolysis and tumor progression by regulating the miR‑149/PKM2 axis in non‑small‑cell lung cancer. Mol Med Rep 2020; 21:1509-1516. [PMID: 32016455 PMCID: PMC7003037 DOI: 10.3892/mmr.2020.10944] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/16/2019] [Indexed: 12/23/2022] Open
Abstract
Cancer cells use aerobic glycolysis to sustain their proliferation. Long non‑coding RNA brain cytoplasmic RNA 1 (BCYRN1) has been reported to act as an oncogene in non‑small‑cell lung cancer (NSCLC). The present study investigated the role of BCYRN1 in NSCLC glycolysis. BCYRN1 expression was detected in NSCLC cells and tissues using reverse transcription‑quantitative PCR. The effect of BCYRN1 on aerobic glycolysis was examined by measuring NSCLC cell glucose catabolism and lactate synthesis. The relationships between BCYRN1 and microRNA (miR)‑149, and between miR‑149 and pyruvate kinase M1/2 (PKM2) were measured using a dual‑luciferase reporter assay. Cell proliferation and invasion were analyzed by the Cell Counting kit‑8 assay and the Matrigel invasion assay, respectively. High BCYRN1 expression was observed in NSCLC tissues and cells compared with the corresponding controls. BCYRN1 induced glycolysis and upregulated the expression levels of PKM2 in NSCLC cells. In addition, BCYRN1 regulated miR‑149 expression levels, and miR‑149 inhibitor rescued the effects of si‑BCYRN1 on glucose consumption and lactate production. miR‑149 knockdown significantly enhanced the expression of PKM2. Furthermore, PKM2 inhibition significantly reversed the effects of miR‑149 inhibitor on glucose catabolism and lactate synthesis. Furthermore, PKM2 was involved in NSCLC cell proliferation and invasion, and BCYRN1 knockdown and miR‑149 overexpression inhibited both processes. The present study suggested that BCYRN1 was involved in cell glycolysis, proliferation and invasion during NSCLC via regulating miR‑149 and PKM2.
Collapse
Affiliation(s)
- Ning Lang
- Department of Preventive Health, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chunyang Wang
- Department of Thoracic Surgery, First Hospital of Qiqihar City, Qiqihar, Heilongjiang 161000, P.R. China
| | - Jiangyang Zhao
- Department of Thoracic Surgery, First Hospital of Qiqihar City, Qiqihar, Heilongjiang 161000, P.R. China
| | - Feng Shi
- Department of Respiratory Diseases, First Hospital of Qiqihar City, Qiqihar, Heilongjiang 161000, P.R. China
| | - Tong Wu
- Department of Respiratory Diseases, First Hospital of Qiqihar City, Qiqihar, Heilongjiang 161000, P.R. China
| | - Hongyan Cao
- Department of Oncology, First Hospital of Qiqihar City, Qiqihar, Heilongjiang 161000, P.R. China
| |
Collapse
|
26
|
Zhao K, Zhang Y, Yuan H, Zhao M, Zhao D. Long noncoding RNA LINC00958 accelerates the proliferation and matrix degradation of the nucleus pulposus by regulating miR-203/SMAD3. Aging (Albany NY) 2019; 11:10814-10825. [PMID: 31804973 PMCID: PMC6932897 DOI: 10.18632/aging.102436] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggests that long noncoding RNAs (lncRNAs) play important roles in the development of intervertebral disc degeneration (IDD). LncRNA LINC00958 has recently been shown to play crucial roles in the development of tumors. However, the role of LINC00958 in IDD remains unclear. We showed that the expression of lncRNA LINC00958 was upregulated in degenerative NP samples, and LINC00958 expression increased gradually along with the grade of exacerbation of disc degeneration. Ectopic expression of LINC00958 promoted nucleus pulposus (NP) cell proliferation, inhibited aggrecan and Col II expression and promoted MMP-2 and MMP-13 expression. In addition, we showed that miR-203 expression was downregulated in degenerative NP samples, and miR-203 expression reduced gradually along with the grade of exacerbation of disc degeneration. Moreover, we demonstrated that the expression of miR-203 was inversely related with LINC00958 expression in NP samples. Ectopic expression of miR-203 inhibited NP cell growth and inhibited ECM degradation. Furthermore, we showed that ectopic expression of miR-203 suppressed the luciferase activity of the wild-type LINC00958 3'-UTR but not the mutant LINC00958 3'-UTR. Elevated expression of LINC00958 inhibited the expression of miR-203 and promoted the expression of SMAD3. In addition, we demonstrated that lncRNA LINC00958 exerted its function by targeting miR-203 in the NP cells. These data suggested that dysregulated lncRNA LINC00958 expression might play an important role in the development of IDD.
Collapse
Affiliation(s)
- Kunchi Zhao
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yang Zhang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongping Yuan
- Department of Nephrology, Jilin FAW General Hospital, Changchun, Jilin 130011, P.R. China
| | - Mingming Zhao
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Dongxu Zhao
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
27
|
Yang Q, Sun J, Ma Y, Zhao C, Song J. LncRNA DLX6-AS1 promotes laryngeal squamous cell carcinoma growth and invasion through regulating miR-376c. Am J Transl Res 2019; 11:7009-7017. [PMID: 31814904 PMCID: PMC6895517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/01/2019] [Indexed: 06/10/2023]
Abstract
Accumulating evidence showed that lncRNAs play important roles in tumour development. Recently, a novel lncRNA DLX6-AS1 was found to be overexpressed in some tumors such as lung adenocarcinoma, renal cell carcinoma and hepatocellular carcinoma. However, the functional roles of DLX6-AS1 in laryngeal squamous cell carcinoma (LSCC) are still unclear. In the study, we showed that the expression level of DLX6-AS1 was upregulated in the LSCC samples compared to the noncancerous tissues. By using CCK-8 analysis, we demonstrated that knockdown expression of DLX6-AS1 suppressed the Hep2 cell growth. DLX6-AS1 knockdown inhibited the Hep2 cell cycle and invasion. MiR-376c was identified to have the complementary binding sites with the DLX6-AS1. By luciferase reporter assay, we indicated that overexpression of miR-376c inhibited the luciferase activity of wild-type DLX6-AS1, but it failed to suppress luciferase activity of mutated one. DLX6-AS1 knockdown enhanced the expression of miR-376c in the Hep2 cell. Moreover, we showed that the expression level of miR-376c was lower in the LSCC samples than in the noncancerous tissues and the expression of miR-376c was negatively correlated with expression of DLX6-AS1 in LSCC tissues. Ectopic expression of miR-376c suppressed cell proliferation, cycle and invasion of LSCC cell. DLX6-AS1 knockdown suppressed cell proliferation, cycle and invasion via regulating miR-376c expression. These data proved that lncRNA DLX6-AS1 might play as an oncogene in LSCC development and tumorigenesis.
Collapse
Affiliation(s)
- Qingjun Yang
- Department of Otolaryngology, Head and Neck Surgery, Zhoukou Central HospitalZhoukou 466000, Henan, China
| | - Jin Sun
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, Henan, China
| | - Yifei Ma
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
| | - Chunjie Zhao
- Department of Otolaryngology, Head and Neck Surgery, Zhoukou Central HospitalZhoukou 466000, Henan, China
| | - Jijun Song
- Department of Otolaryngology, Head and Neck Surgery, Zhoukou Central HospitalZhoukou 466000, Henan, China
| |
Collapse
|
28
|
Wang Z, Ji N, Chen Z, Wu C, Sun Z, Yu W, Hu F, Huang M, Zhang M. Next Generation Sequencing for Long Non-coding RNAs Profile for CD4 + T Cells in the Mouse Model of Acute Asthma. Front Genet 2019; 10:545. [PMID: 31231429 PMCID: PMC6565949 DOI: 10.3389/fgene.2019.00545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/22/2019] [Indexed: 01/13/2023] Open
Abstract
Background and Aims Although long non-coding RNAs (lncRNAs) have been linked to many diseases including asthma, little is known about lncRNA transcriptomes of CD4+ T cells in asthma. The present study aimed to explore the lncRNAs profile in the CD4+T cells from the mouse model of acute asthma. Methods Next generation sequencing for lncRNAs and mRNAs was performed on CD4+ T cells from asthma and control mice. Gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) pathway analyses were performed to predict the functions and signal pathways for the aberrant lncRNAs. The selected lncRNAs were further measured using quantitative real-time PCR (polymerase chain reaction) and observed in the fluorescence in situ hybridization (FISH). The lncRNA–mRNA co-expression network was constructed via Pearson’s correlation coefficient and Cytoscape 3.6. Results Next generation sequencing revealed 36 up-regulated lncRNAs and 98 down-regulated lncRNAs in acute asthma compared with controls. KEGG pathway analysis showed that cytokine-cytokine receptor interaction had the highest enrichment scores. A co-expression network was constructed in which 23 lncRNAs and 301 mRNAs altered formed a total of 12424 lncRNA and mRNA pairs. To validate the RNA sequencing results, we measured the 4 different lncRNAs using qPCR. The lncRNA fantom3_9230106C11 was significantly reduced in CD4+ T cells of asthma. Bioinformatics analysis showed that lncRNA fantom3_9230106C11 had the potential to interact with many miRNAs and transcription factors related to Th2 differentiation. Conclusion This study provided the first evidence for different expression of lncRNAs of CD4+T cells in asthma and may serve as a template for further, larger functional in-depth analyses regarding asthma molecular lncRNAs.
Collapse
Affiliation(s)
- Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chaojie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixiao Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenqin Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, China
| | - Fan Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China.,Department of Immunology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
29
|
Dong Y, Chen H, Gao J, Liu Y, Li J, Wang J. Bioactive Ingredients in Chinese Herbal Medicines That Target Non-coding RNAs: Promising New Choices for Disease Treatment. Front Pharmacol 2019; 10:515. [PMID: 31178721 PMCID: PMC6537929 DOI: 10.3389/fphar.2019.00515] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Chinese herbal medicines (CHMs) are widely used in China and have long been a powerful method to treat diseases in Chinese people. Bioactive ingredients are the main components extracted from herbs that have therapeutic properties. Since artemisinin was discovered to inhibit malaria by Nobel laureate Youyou Tu, extracts from natural plants, particularly bioactive ingredients, have aroused increasing attention among medical researchers. The bioactive ingredients of some CHMs have been found to target various non-coding RNA molecules (ncRNAs), especially miRNAs, lncRNAs, and circRNAs, which have emerged as new treatment targets in numerous diseases. Here we review the evidence that, by regulating the expression of ncRNAs, these ingredients exert protective effects, including pro-apoptosis, anti-proliferation and anti-migration, anti-inflammation, anti-atherosclerosis, anti-infection, anti-senescence, and suppression of structural remodeling. Consequently, they have potential as treatment agents in diseases such as cancer, cardiovascular disease, nervous system disease, inflammatory bowel disease, asthma, infectious diseases, and senescence-related diseases. Although research has been relatively limited and inadequate to date, the promising choices and new alternatives offered by bioactive ingredients for the treatment of the above diseases warrant serious investigation.
Collapse
Affiliation(s)
- Yan Dong
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hengwen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialiang Gao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongmei Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
Liu Z, Mei L, He Z. Long non-coding RNA00882 contributes to platelet-derived growth factor-induced proliferation of human fetal airway smooth muscle cells by enhancing Wnt/β-catenin signaling via sponging miR-3619-5p. Biochem Biophys Res Commun 2019; 514:9-15. [PMID: 31014672 DOI: 10.1016/j.bbrc.2019.04.106] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/14/2019] [Indexed: 12/19/2022]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as novel and critical regulators in the pathogenesis of asthma. However, the precise role of lncRNAs in pediatric asthma remains largely unknown. In this study, we aimed to investigate the biological function of lncRNA00882 (LINC00882) in regulating the proliferation of fetal airway smooth muscle (ASM) cells, which play an important role in airway remodeling during asthma development. Herein, we found that LINC00882 expression was significantly up-regulated in ASM cells stimulated with platelet-derived growth factor (PDGF). Functional experiments showed that the knockdown of LINC00882 markedly reduced the proliferation of fetal ASM cells induced by PDGF, while the overexpression of LINC00882 exhibited the opposite effect. Bioinformatics analysis, the luciferase reporter assay and the RNA pull-down assay revealed that LINC00882 directly interacted with microRNA-3619-5p (miR-3619-5p). LINC00882 negatively regulated miR-3619-5p expression in fetal ASM cells. Notably, β-catenin was identified as a target gene of miR-3619-5p. miR-3619-5p overexpression restricted PDGF-induced cell proliferation through inhibiting Wnt/β-catenin signaling. Moreover, miR-3619-5p overexpression significantly attenuated the LINC00882-induced promotion effect on PDGF-induced cell proliferation and Wnt/β-catenin signaling in fetal ASM cells. In contrast, miR-3619-5p inhibition significantly reversed the LINC00882 knockdown-mediated inhibitory effect on PDGF-induced cell proliferation and Wnt/β-catenin signaling. Taken together, our results demonstrate that LINC00882 promotes PDGF-induced cell proliferation of ASM cells by enhancing Wnt/β-catenin signaling via sponging miR-3619-5p, suggesting a potential role for LINC00882 in airway remodeling in pediatric asthma.
Collapse
Affiliation(s)
- Zengfang Liu
- Yulin No.2 Hospital, Yulin City, Shaanxi, 719000, China
| | - Linghua Mei
- Yulin No.2 Hospital, Yulin City, Shaanxi, 719000, China.
| | - Zhaoping He
- Yulin No.2 Hospital, Yulin City, Shaanxi, 719000, China
| |
Collapse
|
31
|
Zhang L, Shao L, Hu Y. Long noncoding RNA LINC00961 inhibited cell proliferation and invasion through regulating the Wnt/β‐catenin signaling pathway in tongue squamous cell carcinoma. J Cell Biochem 2019; 120:12429-12435. [PMID: 30854692 DOI: 10.1002/jcb.28509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/12/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Affiliation(s)
| | | | - Ying Hu
- Jinan Stomatological Hospital Jinan China
| |
Collapse
|
32
|
Long non-coding RNA TCF7 contributes to the growth and migration of airway smooth muscle cells in asthma through targeting TIMMDC1/Akt axis. Biochem Biophys Res Commun 2018; 508:749-755. [PMID: 30528236 DOI: 10.1016/j.bbrc.2018.11.187] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have been revealed to participate in cellular biological processes in multiple diseases, including asthma. Nevertheless, the role of lncRNA TCF7 (lncTCF7) in airway smooth muscle cells (ASMCs) is still covered. METHODS The expression of lncTCF7 and TIMMDC1 in ASMCs from 12 asthma patients and 12 healthy controls were detected using qRT-PCR. Then MTT assay, EdU assay and transwell assay were conducted respectively to assess the impact of lncTCF7 on ASMCs viability, proliferation and migration. Besides, western blotting was performed to determine the protein levels of TIMMDC1 and AKT/p-AKT. RESULTS We discovered that lncTCF7 and TIMMDC1 were upregulated in asthma groups and lncTCF7 improved ASMCs viability/proliferation and migration. In addition, lncTCF7 regulated TIMMDC1 expression indeed and PDGF-BB treated ASMCs exhibited elevated levels of lncTCF7 and TIMMDC1. Moreover, lncTCF7 suppression diminished both the mRNA and protein levels of TIMMDC1 and markedly reduced p-AKT level which could be enhanced under TIMMDC1 overexpression. Finally, both TIMMDC1 overexpression and AKT activator could restored the inhibitory impacts of lncTCF7 silence on PDGF-BB treated ASMCs. CONCLUSION Our study uncovered that lncTCF7 facilitated human ASMCs growth and migration via targeting TIMMDC1 thus activating AKT signaling, providing a novel possible target for asthma therapy.
Collapse
|
33
|
Kaczmarek KA, Clifford RL, Knox AJ. Epigenetic Changes in Airway Smooth Muscle as a Driver of Airway Inflammation and Remodeling in Asthma. Chest 2018; 155:816-824. [PMID: 30414795 DOI: 10.1016/j.chest.2018.10.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/10/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Epigenetic changes are heritable changes in gene expression, without changing the DNA sequence. Epigenetic processes provide a critical link between environmental insults to the airway and functional changes that determine how airway cells respond to future stimuli. There are three primary epigenetic processes: histone modifications, DNA modification, and noncoding RNAs. Airway smooth muscle has several important roles in the development and maintenance of the pathologic processes occurring in asthma, including inflammation, remodeling, and contraction/hyperresponsiveness. In this review, we describe the evidence for the role of epigenetic changes in driving these processes in airway smooth muscle cells in asthma, with a particular focus on histone modifications. We also discuss how existing therapies may target some of these changes and how epigenetic processes provide targets for the development of novel asthma therapeutics. Epigenetic marks may also provide a biomarker to assess phenotype and treatment responses.
Collapse
Affiliation(s)
- Klaudia A Kaczmarek
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node
| | - Rachel L Clifford
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node
| | - Alan J Knox
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node.
| |
Collapse
|
34
|
Li Z, Li X, Chen C, Li S, Shen J, Tse G, Chan MTV, Wu WKK. Long non-coding RNAs in nucleus pulposus cell function and intervertebral disc degeneration. Cell Prolif 2018; 51:e12483. [PMID: 30039593 PMCID: PMC6528936 DOI: 10.1111/cpr.12483] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 05/15/2018] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is the major cause of low back pain which incurs a significant public-health and economic burden. The aetiology of IDD is complex, with developmental, genetic, biomechanical and biochemical factors contributing to the disease development. Deregulated phenotypes of nucleus pulposus cells, including aberrant differentiation, apoptosis, proliferation and extracellular matrix deposition, are involved in the initiation and progression of IDD. Non-coding RNAs, including long non-coding RNAs (lncRNAs), have recently been identified as important regulators of gene expression. Research into their roles in IDD has been very active over the past 5 years. Our review summarizes current research regarding the roles of deregulated lncRNAs (eg, RP11-296A18.3, TUG1, HCG18) in modulating nucleus pulposus cell functions in IDD. These exciting findings suggest that specific modulation of lncRNAs or their downstream signalling pathways might be an attractive approach for developing novel therapeutics for IDD.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xingye Li
- Department of Orthopedic SurgeryBeijing Jishuitan HospitalFourth Clinical College of Peking UniversityJishuitan Orthopaedic College of Tsinghua UniversityBeijingChina
| | - Chong Chen
- Department of Orthopaedic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shugang Li
- Department of Orthopaedic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianxiong Shen
- Department of Orthopaedic SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Gary Tse
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongChina
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongChina
| | - Matthew T. V. Chan
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongChina
| | - William K. K. Wu
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongChina
- Department of Anaesthesia and Intensive Care and State Key Laboratory of Digestive DiseasesThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
35
|
Qi X, Chen H, Huang Z, Fu B, Wang Y, Xie J, Zhao J, Cao Y, Xiong W. Aberrantly expressed lncRNAs identified by microarray analysis in CD4 +T cells in asthmatic patients. Biochem Biophys Res Commun 2018; 503:1557-1562. [PMID: 30049440 DOI: 10.1016/j.bbrc.2018.07.079] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 01/05/2023]
Abstract
The aim of the study was to determine the expression profiles of message RNAs and long non-coding RNAs in CD4+T cells of asthmatic patients and to explore the clinical value and biological function. Expression profiles in CD4+T cells of asthmatic patients and healthy controls were analyzed by microarray. We found 2725 lncRNAs and 3167 mRNAs differentially expressed. The data were validated by quantitative real time polymerase chain reaction, with 3 up-regulated (ENST00000444682, ENST00000566098, ENST00000583179) and 1 down-regulated (ENST00000579468) lncRNAs found. Receiver operating characteristic curve analysis showed the area under the curve was 0.7058, 0.9026, 0.8361, 0.8316, respectively. Spearman correlation analysis showed that ENST00000566098 was positively related with IL-13 and ENST00000579468 was positively related with peak expiratory flow. Bioinformatics analyses were performed to explore the function of lncRNAs. Specific lncRNAs aberrantly expressed in CD4+T cells may take part in the development of asthma and may be used as biomarkers for diagnosis.
Collapse
Affiliation(s)
- Xuefei Qi
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Huilong Chen
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Zhenli Huang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Bohua Fu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yong Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China.
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China.
| |
Collapse
|
36
|
Zhang XY, Tang XY, Ma LJ, Guo YL, Li XS, Zhao LM, Tian CJ, Cheng DJ, Chen ZC, Zhang LX. Schisandrin B down-regulated lncRNA BCYRN1 expression of airway smooth muscle cells by improving miR-150 expression to inhibit the proliferation and migration of ASMC in asthmatic rats. Cell Prolif 2017; 50. [PMID: 28960519 DOI: 10.1111/cpr.12382] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The mechanism of Schisandrin B on the proliferation and migration of airway smooth muscle cells (ASMCs) in asthmatic rats was explored. METHODS SD rats were divided into three groups: control (group 1), model (group 2) and model + Schisandrin B (group 3). miR-150 and lncRNA BCYRN1 levels were measured by qRT-PCR. The combination of BCYRN1 and miR-150 was detected by RNA pull down. ASMCs' viability/proliferation/migration were examined by WST-1 assay and 24-well Transwell system. RESULTS Schisandrin B up-regulated miR-150 expression and down-regulated BCYRN1 expression in sensitized rats. Schisandrin B reversed the expression of miR-150 and BCYRN1 in MV-treated ASMCs. In addition, Schisandrin B inhibited the viability, proliferation and migration of MV-induced ASMCs. We also found miR-150 inhibited BCYRN1 expression which was proved by experiments using ASMCs transfected with miR-150 inhibitor. CONCLUSION Schisandrin B increased miR-150 expression and decreased BCYRN1, and BCYRN1 expression was inhibited by miR-150, which indicated that Schisandrin B could regulate BCYRN1 through miR-150.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xue-Yi Tang
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li-Jun Ma
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Ya-Li Guo
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiao-Su Li
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li-Min Zhao
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Cui-Jie Tian
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Dong-Jun Cheng
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhuo-Chang Chen
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Luo-Xian Zhang
- Department of Respiratory Medicine, People's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|