1
|
Afthab M, Hambo S, Kim H, Alhamad A, Harb H. Particulate matter-induced epigenetic modifications and lung complications. Eur Respir Rev 2024; 33:240129. [PMID: 39537244 PMCID: PMC11558539 DOI: 10.1183/16000617.0129-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Air pollution is one of the leading causes of early deaths worldwide, with particulate matter (PM) as an emerging factor contributing to this trend. PM is classified based on its physical size, which ranges from PM10 (diameter ≤10 μm) to PM2.5 (≤2.5 μm) and PM0.5 (≤0.5 μm). Smaller-sized PM can move freely through the air and readily infiltrate deep into the lungs, intensifying existing health issues and exacerbating complications. Lung complications are the most common issues arising from PM exposure due to the primary site of deposition in the respiratory system. Conditions such as asthma, COPD, idiopathic pulmonary fibrosis, lung cancer and various lung infections are all susceptible to worsening due to PM exposure. PM can epigenetically modify specific target sites, further complicating its impact on these conditions. Understanding these epigenetic mechanisms holds promise for addressing these complications in cases of PM exposure. This involves studying the effect of PM on different gene expressions and regulation through epigenetic modifications, including DNA methylation, histone modifications and microRNAs. Targeting and manipulating these epigenetic modifications and their mechanisms could be promising strategies for future treatments of lung complications. This review mainly focuses on different epigenetic modifications due to PM2.5 exposure in the various lung complications mentioned above.
Collapse
Affiliation(s)
- Muhammed Afthab
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Shadi Hambo
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Hyunji Kim
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Ali Alhamad
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Hani Harb
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| |
Collapse
|
2
|
Gavito-Covarrubias D, Ramírez-Díaz I, Guzmán-Linares J, Limón ID, Manuel-Sánchez DM, Molina-Herrera A, Coral-García MÁ, Anastasio E, Anaya-Hernández A, López-Salazar P, Juárez-Díaz G, Martínez-Juárez J, Torres-Jácome J, Albarado-Ibáñez A, Martínez-Laguna Y, Morán C, Rubio K. Epigenetic mechanisms of particulate matter exposure: air pollution and hazards on human health. Front Genet 2024; 14:1306600. [PMID: 38299096 PMCID: PMC10829887 DOI: 10.3389/fgene.2023.1306600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024] Open
Abstract
Environmental pollution nowadays has not only a direct correlation with human health changes but a direct social impact. Epidemiological studies have evidenced the increased damage to human health on a daily basis because of damage to the ecological niche. Rapid urban growth and industrialized societies importantly compromise air quality, which can be assessed by a notable accumulation of air pollutants in both the gas and the particle phases. Of them, particulate matter (PM) represents a highly complex mixture of organic and inorganic compounds of the most variable size, composition, and origin. PM being one of the most complex environmental pollutants, its accumulation also varies in a temporal and spatial manner, which challenges current analytical techniques used to investigate PM interactions. Nevertheless, the characterization of the chemical composition of PM is a reliable indicator of the composition of the atmosphere, the quality of breathed air in urbanized societies, industrial zones and consequently gives support for pertinent measures to avoid serious health damage. Epigenomic damage is one of the most promising biological mechanisms of air pollution-derived carcinogenesis. Therefore, this review aims to highlight the implication of PM exposure in diverse molecular mechanisms driving human diseases by altered epigenetic regulation. The presented findings in the context of pan-organic cancer, fibrosis, neurodegeneration and metabolic diseases may provide valuable insights into the toxicity effects of PM components at the epigenomic level and may serve as biomarkers of early detection for novel targeted therapies.
Collapse
Affiliation(s)
- Dulcemaría Gavito-Covarrubias
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
- Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Josué Guzmán-Linares
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Dulce María Manuel-Sánchez
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alejandro Molina-Herrera
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Miguel Ángel Coral-García
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Estela Anastasio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Arely Anaya-Hernández
- Centro de Investigación en Genética y Ambiente, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Primavera López-Salazar
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gabriel Juárez-Díaz
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Javier Martínez-Juárez
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julián Torres-Jácome
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alondra Albarado-Ibáñez
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Ygnacio Martínez-Laguna
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| |
Collapse
|
3
|
Xie J, Wu Y, Tao Q, Liu H, Wang J, Zhang C, Zhou Y, Wei C, Chang Y, Jin Y, Ding Z. The role of lncRNA in the pathogenesis of chronic obstructive pulmonary disease. Heliyon 2023; 9:e22460. [PMID: 38034626 PMCID: PMC10687241 DOI: 10.1016/j.heliyon.2023.e22460] [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: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive and irreversible airflow obstruction with abnormal lung function. Because its pathogenesis involves multiple aspects of oxidative stress, immunity and inflammation, apoptosis, airway and lung repair and destruction, the clinical approach to COPD treatment is not further updated. Therefore, it is crucial to discover a new means of COPD diagnosis and treatment. COPD etiology is associated with complex interactions between environmental and genetic determinants. Numerous genes are involved in the pathogenic process of this illness in research samples exposed to hazardous environmental conditions. Among them, Long non-coding RNAs (lncRNAs) have been reported to be involved in the molecular mechanisms of COPD development induced by different environmental exposures and genetic susceptibility encounters, and some potential lncRNA biomarkers have been identified as early diagnostic, disease course determination, and therapeutic targets for COPD. In this review, we summarize the expression profiles of the reported lncRNAs that have been reported in COPD studies related to environmental risk factors such as smoking and air pollution exposure and provided an overview of the roles of those lncRNAs in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hua Liu
- Anhui Institute for Food and Drug Control, Hefei, Anhui, China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Zhen Ding
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| |
Collapse
|
4
|
Alluli A, Rijnbout St James W, Eidelman DH, Baglole CJ. Dynamic relationship between the aryl hydrocarbon receptor and long noncoding RNA balances cellular and toxicological responses. Biochem Pharmacol 2023; 216:115745. [PMID: 37597813 DOI: 10.1016/j.bcp.2023.115745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a cytosolic transcription factor activated by endogenous ligands and xenobiotic chemicals. Once the AhR is activated, it translocates to the nucleus, dimerizes with the AhR nuclear translator (ARNT) and binds to xenobiotic response elements (XRE) to promote gene transcription, notably the cytochrome P450 CYP1A1. The AhR not only mediates the toxic effects of environmental chemicals, but also has numerous putative physiological functions. This dichotomy in AhR biology may be related to reciprocal regulation of long non-coding RNA (lncRNA). lncRNA are defined as transcripts more than 200 nucleotides in length that do not encode a protein but are implicated in many physiological processes such as cell differentiation, cell proliferation, and apoptosis. lncRNA are also linked to disease pathogenesis, particularly the development of cancer. Recent studies have revealed that AhR activation by environmental chemicals affects the expression and function of lncRNA. In this article, we provide an overview of AhR signaling pathways activated by diverse ligands and highlight key differences in the putative biological versus toxicological response of AhR activation. We also detail the functions of lncRNA and provide current data on their regulation by the AhR. Finally, we outline how overlap in function between AhR and lncRNA may be one way in which AhR can be both a regulator of endogenous functions but also a mediator of toxicological responses to environmental chemicals. Overall, more research is still needed to fully understand the dynamic interplay between the AhR and lncRNA.
Collapse
Affiliation(s)
- Aeshah Alluli
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - Willem Rijnbout St James
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, McGill University, Canada; Department of Medicine, McGill University, Canada
| | - Carolyn J Baglole
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada; Department of Medicine, McGill University, Canada; Department of Pharmacology and Therapeutics, McGill University, Canada.
| |
Collapse
|
5
|
Noah TL, Worden CP, Rebuli ME, Jaspers I. The Effects of Wildfire Smoke on Asthma and Allergy. Curr Allergy Asthma Rep 2023; 23:375-387. [PMID: 37171670 PMCID: PMC10176314 DOI: 10.1007/s11882-023-01090-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 05/13/2023]
Abstract
PURPOSE OF REVIEW To review the recent literature on the effects of wildfire smoke (WFS) exposure on asthma and allergic disease, and on potential mechanisms of disease. RECENT FINDINGS Spatiotemporal modeling and increased ground-level monitoring data are allowing a more detailed picture of the health effects of WFS exposure to emerge, especially with regard to asthma. There is also epidemiologic and some experimental evidence to suggest that WFS exposure increases allergic predisposition and upper airway or sinonasal disease, though much of the literature in this area is focused more generally on PM2.5 and is not specific for WFS. Experimental evidence for mechanisms includes disruption of epithelial integrity with downstream effects on inflammatory or immune pathways, but experimental models to date have not consistently reflected human disease in this area. Exposure to WFS has an acute detrimental effect on asthma. Potential mechanisms are suggested by in vitro and animal studies.
Collapse
Affiliation(s)
- Terry L Noah
- Department of Pediatrics, University of North Carolina at Chapel Hill, 260 Macnider Building, 333 S. Columbia St., Chapel Hill, NC, 27599, USA.
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, USA.
| | - Cameron P Worden
- Department of Otolaryngology/Head & Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Meghan E Rebuli
- Department of Pediatrics, University of North Carolina at Chapel Hill, 260 Macnider Building, 333 S. Columbia St., Chapel Hill, NC, 27599, USA
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Ilona Jaspers
- Department of Pediatrics, University of North Carolina at Chapel Hill, 260 Macnider Building, 333 S. Columbia St., Chapel Hill, NC, 27599, USA
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|
6
|
Yi E, Lin B, Zhang Y, Wang X, Zhang J, Liu Y, Jin J, Hong W, Lin Z, Cao W, Mei X, Bai G, Bing Li B, Zhou Y, Ran P. Smad3-mediated lncRNA HSALR1 enhances the non-classic signalling pathway of TGF-β1 in human bronchial fibroblasts by binding to HSP90AB1. Clin Transl Med 2023; 13:e1292. [PMID: 37317677 PMCID: PMC10267427 DOI: 10.1002/ctm2.1292] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is one of the diseases with high mortality and morbidity with complex pathogenesis. Airway remodeling is an unavoidable pathological characteristic. However, the molecular mechanisms of airway remodeling are incompletely defined. METHODS lncRNAs highly correlated with transforming growth factor beta 1(TGF-β1) expression were chosen, the lncRNA ENST00000440406 (named HSP90AB1 Assoicated LncRNA 1, HSALR1) was chosen for further functional experiments. Dual luciferase and ChIP assay were used to detect the upstream of HSALR1, transcriptome sequencing, Cck-8, Edu, cell proliferation, cell cycle assay, and WB detection of pathway levels confirmed the effect of HSALR1 on fibroblast proliferation and phosphorylation levels of related pathways. Mice was infected with adeno-associated virus (AAV) to express HSALR1 by intratracheal instillation under anesthesia and was exposure to cigarette smoke, then mouse lung function was performed and the pathological sections of lung tissues were analyzed. RESULTS Herein, lncRNA HSALR1 was identified as highly correlated with the TGF-β1 and mainly expressed in human lung fibroblasts. HSALR1 was induced by Smad3 and promoted fibroblasts proliferation. Mechanistically, it could directly bind to HSP90AB1 protein, and acted as a scaffold to stabilize the binding between Akt and HSP90AB1 to promote Akt phosphorylation. In vivo, mice expressed HSALR1 by AAV was exposure to cigarette smoke (CS) for COPD modeling. We found that lung function was worse and airway remodeling was more pronounced in HSLAR1 mice compare to wild type (WT) mice. CONCLUSION Our results suggest that lncRNA HSALR1 binds to HSP90AB1 and Akt complex component, and enhances activity of the TGF-β1 smad3-independent pathway. This finding described here suggest that lncRNA can participate in COPD development, and HSLAR1 is a promising molecular target of COPD therapy.
Collapse
Affiliation(s)
- Erkang Yi
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Biting Lin
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yi Zhang
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiaoyu Wang
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jiahuan Zhang
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yu Liu
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jing Jin
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wei Hong
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhiwei Lin
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Weitao Cao
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xinyue Mei
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Ge Bai
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Bing Bing Li
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yumin Zhou
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Pixin Ran
- Guangzhou Institute of Respiratory Health & State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou LaboratoryBiolandGuangzhouGuangdongChina
| |
Collapse
|
7
|
He B, Xu HM, Liu HW, Zhang YF. Unique regulatory roles of ncRNAs changed by PM 2.5 in human diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114812. [PMID: 36963186 DOI: 10.1016/j.ecoenv.2023.114812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
PM2.5 is a type of particulate matter with an aerodynamic diameter smaller than 2.5 µm, and exposure to PM2.5 can adversely damage human health. PM2.5 may impair health through oxidative stress, inflammatory reactions, immune function alterations and chromosome or DNA damage. Through increasing in-depth studies, researchers have found that noncoding RNAs (ncRNAs), particularly microRNAs (miRNAs), circular RNAs (circRNAs) as well as long noncoding RNAs (lncRNAs), might play significant roles in PM2.5-related human diseases via some of the abovementioned mechanisms. Therefore, in this review, we mainly discuss the regulatory function of ncRNAs altered by PM2.5 in human diseases and summarize the potential molecular mechanisms. The findings reveal that these ncRNAs might induce or promote diseases via inflammation, the oxidative stress response, cell autophagy, apoptosis, cell junction damage, altered cell proliferation, malignant cell transformation, disruption of synaptic function and abnormalities in the differentiation and status of immune cells. Moreover, according to a bioinformatics analysis, the altered expression of potential genes caused by these ncRNAs might be related to the development of some human diseases. Furthermore, some ncRNAs, including lncRNAs, miRNAs and circRNAs, or processes in which they are involved may be used as biomarkers for relevant diseases and potential targets to prevent these diseases. Additionally, we performed a meta-analysis to identify more promising diagnostic ncRNAs as biomarkers for related diseases.
Collapse
Affiliation(s)
- Bo He
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Hai-Ming Xu
- Department of Occupational and Environmental Medicine, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| | - Hao-Wen Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
8
|
Aghaei-Zarch SM, Alipourfard I, Rasoulzadeh H, Najafi S, Aghaei-Zarch F, Partov S, Movafagh A, Jahanara A, Toolabi A, Sheikhmohammadi A, Pour NN, Neghad SK, Ashrafi-Asgarabad A. Non-coding RNAs: An emerging player in particulate matter 2.5-mediated toxicity. Int J Biol Macromol 2023; 235:123790. [PMID: 36822288 DOI: 10.1016/j.ijbiomac.2023.123790] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/17/2023] [Indexed: 02/23/2023]
Abstract
Exposure to air pollution has been connected to around seven million early deaths annually and also contributing to higher than 3 % of disability-adjusted lost life years. Particulate matters (PM) are among the key pollutants that directly discharged or formed due to atmospheric chemical interactions. Among these matters, due of its large surface area, PM2.5 may absorb a different harmful and toxic substances. One of the outcomes of such environmental disturbance is oxidative stress which affects cellular processes including apoptosis, inflammation, and epithelial mesenchymal transition. Non-coding RNAs (ncRNA) such as, miRNAs, lncRNAs, and circRNAs are classified as non-protein coding RNA's. Over the past few years these small molecules have been gaining so much attention since they participate in variety of physiological and pathological processes and their expression change during disease periods. Regarding epigenetic properties, ncRNAs play an important function in organism's response to environmental stimulus. In this manner, it was revealed that exposure to PM2.5 may cause epigenetic reprogramming, such as, ncRNAs signature's alteration, which can be effective concerning pathophysiology state. In this review, we describe PM2.5 impact on ncRNAs and excavate its roles in toxicity caused by PM2.5.
Collapse
Affiliation(s)
- Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Hassan Rasoulzadeh
- Department of Environmental Health Engineering, School of Public Health, Bam University of Medical Sciences, Bam, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Saber Partov
- Department of Clinical and Biological Sciences, Faculty of Medicine and Surgery, University of Turin, Turin, Italy
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Jahanara
- Neonatology, Bam University of Medical Sciences, Bam, Iran
| | - Ali Toolabi
- Environmental Health Research Center, School of Health and Nutrition, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Amir Sheikhmohammadi
- Department of Environmental Health Engineering, School of Health, Khoy University of Medical Sciences, Khoy, Iran
| | | | | | - Ahad Ashrafi-Asgarabad
- Department of Epidemiology, School of Health, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
9
|
Du X, Jiang Y, Zhang Q, Zhu X, Zhang Y, Liu C, Niu Y, Cai J, Kan H, Chen R. Genome-Wide Profiling of Exosomal Long Noncoding RNAs Following Air Pollution Exposure: A Randomized, Crossover Trial. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:2856-2863. [PMID: 36757895 DOI: 10.1021/acs.est.2c05956] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Changes in human genome-wide long noncoding RNAs (lncRNAs) associated with air pollution are unknown. This study aimed to investigate the effect of air pollution on human exosomal lncRNAs. A randomized, crossover trial was conducted among 35 healthy adults. Participants were allocated to 4 h exposure in road (high air pollution) and park (low air pollution) sessions in random order with a 2 week washout period. RNA sequencing was performed to measure lncRNAs. Differential lncRNAs were identified using a linear mixed-effect model. Mean concentrations of air pollutants such as ultrafine particles (UFP), black carbon (BC), carbon monoxide (CO), and nitrogen dioxide (NO2) were 2-3 times higher in the road than those in the park. Fifty-five lncRNAs [false discovery rate (FDR) < 0.05] including lncRNA NORAD, MALAT1, and H19 were changed in response to air pollution exposure. We found that 54 lncRNAs were associated with CO, 49 lncRNAs with UFP, 49 lncRNAs with BC, 48 lncRNAs with NO2, and 4 lncRNAs with PM2.5 (FDR < 0.05). These differential lncRNAs participated in dozens of pathways including cardiovascular signaling, epithelial cell proliferation, inflammation, and transforming growth factor. This trial for the first time profiled changes of human exosomal lncRNAs following air pollution. Our findings revealed multiple biological processes moderated by lncRNAs and provided epigenetic insights into cardiovascular effects of air pollution.
Collapse
Affiliation(s)
- Xihao Du
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Yixuan Jiang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Qingli Zhang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Xinlei Zhu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Yang Zhang
- Department of Systems Biology for Medicine, Shanghai Medical College, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Cong Liu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Yue Niu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Jing Cai
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
- National Center for Children's Health, Children's Hospital of Fudan University, Shanghai 200032, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| |
Collapse
|
10
|
Lee KY, Ho SC, Sun WL, Feng PH, Lin CW, Chen KY, Chuang HC, Tseng CH, Chen TT, Wu SM. Lnc-IL7R alleviates PM 2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol Toxicol 2022; 38:1097-1120. [PMID: 35303175 DOI: 10.1007/s10565-022-09709-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Long-term exposure to PM2.5 (particulate matter with an aerodynamic diameter of ≤ 2.5 μm) is associated with pulmonary injury and emphysema in patients with chronic obstructive pulmonary disease (COPD). We investigated mechanisms through which the long noncoding RNA lnc-IL7R contributes to cellular damage by inducing oxidative stress in COPD patients exposed to PM2.5. METHODS Associations of serum lnc-IL7R levels with lung function, emphysema, and previous PM2.5 exposure in COPD patients were analyzed. Reactive oxygen species and lnc-IL7R levels were measured in PM2.5-treated cells. The levels of lnc-IL7R and cellular senescence-associated genes, namely p16INK4a and p21CIP1/WAF1, were determined through lung tissue section staining. The effects of p16INK4a or p21CIP1/WAF1 regulation were examined by performing lnc-IL7R overexpression and knockdown assays. The functions of lnc-IL7R-mediated cell proliferation, cell cycle, senescence, colony formation, and apoptosis were examined in cells treated with PM2.5. Chromatin immunoprecipitation assays were conducted to investigate the epigenetic regulation of p21CIP1/WAF1. RESULTS Lnc-IL7R levels decreased in COPD patients and were negatively correlated with emphysema or PM2.5 exposure. Lnc-IL7R levels were upregulated in normal lung epithelial cells but not in COPD cells exposed to PM2.5. Lower lnc-IL7R expression in PM2.5-treated cells induced p16INK4a and p21CIP1/WAF1 expression by increasing oxidative stress. Higher lnc-IL7R expression protected against cellular senescence and apoptosis, whereas lower lnc-IL7R expression augmented injury in PM2.5-treated cells. Lnc-IL7R and the enhancer of zeste homolog 2 (EZH2) synergistically suppressed p21CIP1/WAF1 expression through epigenetic modulation. CONCLUSION Lnc-IL7R attenuates PM2.5-mediated p21CIP1/WAF1 expression through EZH2 recruitment, and its dysfunction may augment cellular injury in COPD.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
11
|
Mukherjee S, Kundu U, Desai D, Pillai PP. Particulate Matters Affecting lncRNA Dysregulation and Glioblastoma Invasiveness: In Silico Applications and Current Insights. J Mol Neurosci 2022; 72:2188-2206. [PMID: 36370303 DOI: 10.1007/s12031-022-02069-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/14/2022] [Indexed: 11/15/2022]
Abstract
With a reported rise in global air pollution, more than 50% of the population remains exposed to toxic air pollutants in the form of particulate matters (PMs). PMs, from various sources and of varying sizes, have a significant impact on health as long-time exposure to them has seen a correlation with various health hazards and have also been determined to be carcinogenic. In addition to disrupting known cellular pathways, PMs have also been associated with lncRNA dysregulation-a factor that increases predisposition towards the onset or progression of cancer. lncRNA dysregulation is further seen to mediate glioblastoma multiforme (GBM) progression. The vast array of information regarding cancer types including GBM and its various precursors can easily be obtained via innovative in silico approaches in the form of databases such as GEO and TCGA; however, a need to obtain selective and specific information correlating anthropogenic factors and disease progression-in the case of GBM-can serve as a critical tool to filter down and target specific PMs and lncRNAs responsible for regulating key cancer hallmarks in glioblastoma. The current review article proposes an in silico approach in the form of a database that reviews current updates on correlation of PMs with lncRNA dysregulation leading to GBM progression.
Collapse
Affiliation(s)
- Swagatama Mukherjee
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Uma Kundu
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Dhwani Desai
- Integrated Microbiome Resource, Department of Pharmacology and Marine Microbial Genomics and Biogeochemistry lab, Department of Biology, Dalhousie University, Halifix, Canada
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India.
| |
Collapse
|
12
|
Xie W, Ling M, Xiao T, Fan Z, Chen D, Tang M, Bian Q. Tanshinone IIA-regulation of IL-6 antagonizes PM 2 .5 -induced proliferation of human bronchial epithelial cells via a STAT3/miR-21 reciprocal loop. ENVIRONMENTAL TOXICOLOGY 2022; 37:1686-1696. [PMID: 35304817 DOI: 10.1002/tox.23517] [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: 10/21/2021] [Revised: 02/21/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Particulate matter 2.5 (PM2.5 ), a component of atmospheric particulate matter, leads to changes in gene expression and cellular functions. Epidemiological evidence confirms that PM2.5 has a positive correlation with lung injury. However, the molecular mechanisms involved remain poorly understood, and preventive methods are needed. In the present study, with human bronchial epithelial (HBE) cells in culture, we showed that low concentrations of PM2.5 resulted in acceleration of the G1/S transition and cell proliferation. Consistent with these effects, expression of the pro-inflammatory factor interleukin-6 (IL-6) was elevated in HBE cells exposed to PM2.5 . Accordingly, signal transducer and activator of transcription 3 (STAT3) was activated, which down-regulated expression of cyclin D1. In addition, PM2.5 exposure led to higher levels of miR-21, and there was a reciprocal loop between miR-21 and STAT3. For HBE cells, tanshinone IIA (Tan IIA) reversed the PM2.5 -induced cell cycle alteration and cell proliferation, and reduced the expression of cytokines (IL-6, STAT3, and miR-21). These results show that, for HBE cells, Tan IIA attenuates the PM2.5 -induced G1/S alteration and cell proliferation, and indicate that it has potential clinical application for PM2.5 -induced respiratory injuries.
Collapse
Affiliation(s)
- Wenjing Xie
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
- Public Health Administration Center, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Min Ling
- Institute of Toxicology and Risk Assessment, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Tian Xiao
- Institute of Toxicology and Risk Assessment, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Zi Fan
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongya Chen
- Institute of Toxicology and Risk Assessment, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Qian Bian
- Institute of Toxicology and Risk Assessment, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Li YR, Zong RQ, Zhang HY, Meng XY, Wu FX. Mechanism Analysis of LINC00665 and Its Peptides CIP2A-BP in Hepatocellular Carcinoma. Front Genet 2022; 13:861096. [PMID: 35350239 PMCID: PMC8957827 DOI: 10.3389/fgene.2022.861096] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: More and more studies show that long non-coding RNAs (lncRNAs) have miniature open reading frames that can be translated into short peptides. Here, we identify the long non-coding gene LINC00665 and its short peptides (CIP2A-BP) in hepatocellular carcinoma (HCC) and explore how they contribute to HCC progression. Materials and methods: First, GSE101728 data were acquired through the Gene Expression Omnibus for identification of differentially expressed genes (DEGs), and gene set enrichment analysis (GSEA) was conducted to find enriched biological pathways. Then, further bioinformatics analysis was carried out on the screened long non-coding genes, and LINC00665 expression was detected in HCC and normal liver samples. The relations between LINC00665 expression, HCC prognosis, and clinical characteristics were studied. Receiver operating characteristic (ROC) analysis was also applied to verify the LINC00665 prediction in HCC prognosis. In addition, pertinent experiments on LINC00665 and CIP2A-BP were also carried out to explore their roles in the progression of HCC. Results: As a result, we screened out 332 DEGs in total, including 130 upregulated and 202 downregulated DEGs. These DEGs were mainly enriched in posttranscriptional regulation of gene expression, RNA processing, nucleolus, and gene silencing biological pathways. In addition, we found that LINC00665 was increased in HCC samples, which substantially indicated its poor prognosis. Compared with normal tissues, LINC00665 had higher expression in the pathological stages III and IV, tumor-free groups, people no more than 60 years old, and stages T3, T4, N0, N1, and M1. ROC curve indicated that the variable INC00665 had certain accuracy in predicting overall survival (OS). Moreover, in functional experiments, LINC00665 knockdown could significantly decrease HCC cell proliferation, migration, and invasion, while overexpressed CIP2A-BP could markedly increase HCC cell proliferation, invasion, and migration. Conclusion: Our findings not only disclose a unique mechanism by which CIP2A-BP encoded by LINC00665 promotes HCC carcinogenesis but suggest that these long non-coding genes and short peptides could be used as biomarkers for HCC diagnosis and prognosis and new targets for HCC therapy.
Collapse
Affiliation(s)
- Yi-Ran Li
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Rui-Qing Zong
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hong-Yan Zhang
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiao-Yan Meng
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Fei-Xiang Wu
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Aghapour M, Ubags ND, Bruder D, Hiemstra PS, Sidhaye V, Rezaee F, Heijink IH. Role of air pollutants in airway epithelial barrier dysfunction in asthma and COPD. Eur Respir Rev 2022; 31:31/163/210112. [PMID: 35321933 PMCID: PMC9128841 DOI: 10.1183/16000617.0112-2021] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic exposure to environmental pollutants is a major contributor to the development and progression of obstructive airway diseases, including asthma and COPD. Understanding the mechanisms underlying the development of obstructive lung diseases upon exposure to inhaled pollutants will lead to novel insights into the pathogenesis, prevention and treatment of these diseases. The respiratory epithelial lining forms a robust physicochemical barrier protecting the body from inhaled toxic particles and pathogens. Inhalation of airborne particles and gases may impair airway epithelial barrier function and subsequently lead to exaggerated inflammatory responses and airway remodelling, which are key features of asthma and COPD. In addition, air pollutant-induced airway epithelial barrier dysfunction may increase susceptibility to respiratory infections, thereby increasing the risk of exacerbations and thus triggering further inflammation. In this review, we discuss the molecular and immunological mechanisms involved in physical barrier disruption induced by major airborne pollutants and outline their implications in the pathogenesis of asthma and COPD. We further discuss the link between these pollutants and changes in the lung microbiome as a potential factor for aggravating airway diseases. Understanding these mechanisms may lead to identification of novel targets for therapeutic intervention to restore airway epithelial integrity in asthma and COPD. Exposure to air pollution induces airway epithelial barrier dysfunction through several mechanisms including increased oxidative stress, exaggerated cytokine responses and impaired host defence, which contributes to development of asthma and COPD. https://bit.ly/3DHL1CA
Collapse
Affiliation(s)
- Mahyar Aghapour
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Epalinges, Switzerland
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Venkataramana Sidhaye
- Pulmonary and Critical Care Medicine, Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Fariba Rezaee
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children's, Cleveland, OH, USA.,Dept of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Depts of Pathology and Medical Biology and Pulmonology, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| |
Collapse
|
15
|
Olmedo-Suárez MÁ, Ramírez-Díaz I, Pérez-González A, Molina-Herrera A, Coral-García MÁ, Lobato S, Sarvari P, Barreto G, Rubio K. Epigenetic Regulation in Exposome-Induced Tumorigenesis: Emerging Roles of ncRNAs. Biomolecules 2022; 12:513. [PMID: 35454102 PMCID: PMC9032613 DOI: 10.3390/biom12040513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
Environmental factors, including pollutants and lifestyle, constitute a significant role in severe, chronic pathologies with an essential societal, economic burden. The measurement of all environmental exposures and assessing their correlation with effects on individual health is defined as the exposome, which interacts with our unique characteristics such as genetics, physiology, and epigenetics. Epigenetics investigates modifications in the expression of genes that do not depend on the underlying DNA sequence. Some studies have confirmed that environmental factors may promote disease in individuals or subsequent progeny through epigenetic alterations. Variations in the epigenetic machinery cause a spectrum of different disorders since these mechanisms are more sensitive to the environment than the genome, due to the inherent reversible nature of the epigenetic landscape. Several epigenetic mechanisms, including modifications in DNA (e.g., methylation), histones, and noncoding RNAs can change genome expression under the exogenous influence. Notably, the role of long noncoding RNAs in epigenetic processes has not been well explored in the context of exposome-induced tumorigenesis. In the present review, our scope is to provide relevant evidence indicating that epigenetic alterations mediate those detrimental effects caused by exposure to environmental toxicants, focusing mainly on a multi-step regulation by diverse noncoding RNAs subtypes.
Collapse
Affiliation(s)
- Miguel Ángel Olmedo-Suárez
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Facultad de Biotecnología, Campus Puebla, Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla 72410, Mexico
| | - Andrea Pérez-González
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
| | - Alejandro Molina-Herrera
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
| | - Miguel Ángel Coral-García
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Decanato de Ciencias de la Salud, Campus Puebla, Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla 72410, Mexico
| | - Sagrario Lobato
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
| | - Pouya Sarvari
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
| | - Guillermo Barreto
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Laboratoire IMoPA, CNRS, Université de Lorraine, UMR 73635 Nancy, France
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico; (M.Á.O.-S.); (I.R.-D.); (A.P.-G.); (A.M.-H.); (M.Á.C.-G.); (S.L.); (P.S.); (G.B.)
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
- Laboratoire IMoPA, CNRS, Université de Lorraine, UMR 73635 Nancy, France
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
16
|
Li J, Zhang L, Wang J, Jia R, Zhang X, Li X, Fu Y, Song L. Differential expression of long non-coding RNAs in the hippocampus of mice exposed to PM 2.5 in Dalian, China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:12136-12146. [PMID: 34561797 DOI: 10.1007/s11356-021-16496-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
Evidence is mounting that PM2.5 exposure could lead to learning disability, memory deficits, and cognitive impairment; however, the underlying mechanisms are still not well demonstrated yet. Long non-coding RNAs (LncRNAs) play a crucial role in many human diseases. Although the relationship of Alzheimer's disease (AD) and lncRNAs have been discovered, the role of lncRNA in AD-like phenotype induced by PM2.5 needs further exploration. In this study, we profiled the expression of messenger RNAs (mRNAs) and lncRNAs in hippocampus after confirming the AD-like changes in mice. Compared with the control group, a total of 478 mRNAs and 151 lncRNAs were dysregulated after PM2.5 exposure. ECM-receptor interaction, focal adhesion, complement and coagulation cascades, and AGE-RAGE signaling pathway were found dysregulated through lncRNA-co-expressed genes analysis based on the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Meanwhile, the genes related to microglia were significantly altered, such as CX3CR1, CD163, lncRNA Gm44750, and lncRNA Gm43509. Above evidences suggested that microglia-related lncRNAs dysregulation probably plays a crucial role in PM2.5exposure-associated learning and memory deficits.
Collapse
Affiliation(s)
- Jie Li
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Longying Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, People's Republic of China
| | - Jiaqi Wang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
- The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, Liaoning Province, People's Republic of China
| | - Ruxue Jia
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Xiao Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Xiaojing Li
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Ying Fu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Laiyu Song
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, Liaoning Province, People's Republic of China.
| |
Collapse
|
17
|
Cao L, Ping F, Zhang F, Gao H, Li P, Ning X, Cui G, Ma Z, Jiang X, Li S, Han S. Tissue-Protective Effect of Erdosteine on Multiple-Organ Injuries Induced by Fine Particulate Matter. Med Sci Monit 2021; 27:e930909. [PMID: 34873140 PMCID: PMC8665604 DOI: 10.12659/msm.930909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background Fine particulate matter (PM2.5) is the air pollutant that most threatens global public health. The purpose of this study was to observe the inflammatory and oxidative stress injury of multiple organs induced by PM2.5 in rats and to explore the tissue-protective effect of erdosteine. Material/Methods We randomly divided 40 male Wistar rats into a blank control group, a saline group, a PM2.5 exposure group, and an erdosteine intervention group. We assessed changes in organs tissue homogenate and biomarkers of inflammation and oxidative stress in serum and bronchoalveolar lavage fluid (BALF). Results (1) The expressions of IL-6, IL-1β, TNF-α, 8-OHdG, 4-HNE, and PCC in serum and BALF of the PM2.5 exposure group increased, but decreased after treatment with erdosteine, suggesting that erdosteine treatment attenuates inflammatory and oxidative stress injury. (2) The expression of γ-GCS in serum and lungs in the PM2.5 exposure group increased, but did not change significantly after treatment with erdosteine. This suggests that PM2.5 upregulates the level of γ-GCS, while erdosteine does not affect this protective response. (3) The expression of T-AOC in serum, lungs, spleens, and kidneys of the PM2.5 exposure group decreased, but increased after treatment with erdosteine. Our results suggest that PM2.5 can cause imbalance of oxidation/anti-oxidation in multiple organs, and erdosteine can alleviate this imbalance. Conclusions PM2.5 exposure can lead to inflammatory and oxidative stress damage in serum and organ tissues of rats. Erdosteine may be an effective anti-inflammatory and antioxidant that can reduce this injury.
Collapse
Affiliation(s)
- Lei Cao
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Fen Ping
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Fengrui Zhang
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Haixiang Gao
- Department of Respiratory Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Ping Li
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Xiaohui Ning
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Guohuan Cui
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Zheng Ma
- International Department, Children's Hospital of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Xin Jiang
- Third Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Suyan Li
- Department of General Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Shuzhi Han
- Third Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
18
|
Lv LX, Wen M, Lv F, Ji TB, Fu HL, Man N. Knockdown of long noncoding RNA growth arrest-specific transcript 5 regulates forkhead box O3 to inhibit lipopolysaccharide-induced human bronchial epithelial cell pyroptosis. Kaohsiung J Med Sci 2021; 38:87-96. [PMID: 34529353 DOI: 10.1002/kjm2.12452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/14/2021] [Accepted: 08/22/2021] [Indexed: 12/17/2022] Open
Abstract
Pyroptosis is a novel proinflammatory programmed cell death process. This study was designed to investigate the functional mechanisms of long noncoding RNA growth arrest-specific transcript 5 (lncRNA GAS5) on lipopolysaccharide (LPS)-induced human bronchial epithelial cell (HBEC) pyroptosis. LPS was used to induce pyroptosis in HBECs, followed by the detection of the expression of GAS5, forkhead box O3 (FOXO3), and nuclear factor E2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling pathway-related factors. Cell viability was evaluated using CCK-8 assay, lactate dehydrogenase (LDH) release was assessed by LDH assay kit and caspase-1 activity by flow cytometry. Furthermore, expression of NOD-like receptor family pyrin domain containing 3 and pyroptosis-related proteins was evaluated using Western blot analysis, while enzyme-linked immunosorbent assay was used to determine the levels of inflammatory factors. The interaction between GAS5 and FOXO3 was confirmed using bioinformatic prediction, RNA immunoprecipitation assay, RNA pull-down, and dual-luciferase reporter gene assay. Treatment of HBECs with LPS upregulated the expression of GAS5 and FOXO3, resulting in the inactivation of the Nrf2/HO-1 signaling pathway. On the other hand, inhibition of both GAS5 and FOXO3 promoted cell viability, reduced LDH release, pyroptosis, and inflammatory response in LPS-induced HBECs. Furthermore, FOXO3 could interact with GAS5, while FOXO3 overexpression reversed the inhibitory effect of GAS5 knockdown on cell pyroptosis. Thus, mechanistically, inhibition of FOXO3 activates the Nrf2/HO-1 pathway to suppress LPS-induced pyroptosis in HBECs. This study revealed that GAS5 knockdown attenuates FOXO3 expression thereby activating the Nrf2/HO-1 pathway to inhibit LPS-induced pyroptosis in HBECs. These findings may contribute to identifying novel targets that inhibit pyroptosis in HBECs.
Collapse
Affiliation(s)
- Ling-Xia Lv
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Mei Wen
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Fei Lv
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Tai-Bing Ji
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Hua-Li Fu
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| | - Ning Man
- Respiratory and Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, Hubei, China
| |
Collapse
|
19
|
Abstract
The adverse effects of polluted air on human health have been increasingly appreciated worldwide. It is estimated that outdoor air pollution is associated with the death of 4.2 million people globally each year. Accumulating epidemiological studies indicate that exposure to ambient fine particulate matter (PM2.5), one of the important air pollutants, significantly contributes to respiratory mortality and morbidity. PM2.5 causes lung damage mainly by inducing inflammatory response and oxidative stress. In this paper, we reviewed the research results of our group on the effects of PM2.5 on chronic obstructive pulmonary disease, asthma, and lung cancer. And recent research progress on epidemiological studies and potential mechanisms were also discussed. Reducing air pollution, although remaining a major challenge, is the best and most effective way to prevent the onset and progression of respiratory diseases.
Collapse
|
20
|
Li B, Huang N, Wei S, Xv J, Meng Q, Aschner M, Li X, Chen R. lncRNA TUG1 as a ceRNA promotes PM exposure-induced airway hyper-reactivity. JOURNAL OF HAZARDOUS MATERIALS 2021; 416:125878. [PMID: 34492818 PMCID: PMC8432742 DOI: 10.1016/j.jhazmat.2021.125878] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/01/2021] [Accepted: 04/11/2021] [Indexed: 05/29/2023]
Abstract
With the increased appreciation for the significance of noncoding RNAs (ncRNAs), the present research aimed to determine the role of competing endogenous RNA (ceRNA) in the process of particulate matter (PM) exposure-induced pulmonary damage. Alterations in messenger RNA (RNA), microRNA and long non-coding RNA (lncRNA) profiles of human bronchial epithelial (HBE) cells treated with PM were analyzed by microarray assays. Next, we identified that lncRNA taurine upregulated gene 1 (TUG1) acted as a competing endogenous RNA for microRNA-222-3p (miR-222-3p) and subsequently attenuated the inhibitory effect of miR-222-3p on CUGBP elav-like family member 1 (CELF1). The binding potency among ceRNAs was verified by RNA immunoprecipitation (RIP) assay and dual-luciferase reporter assay. Knockdown of TUG1 attenuated HBE cell apoptosis and cell cycle arrest by downregulation of CELF1 and protein 53 (p53). Further, we confirmed that Tug1/mir-222-3p/CELF1/p53 network aggravated PM-induced airway hyper-reactivity (AHR) in mice. In summary, our novel findings revealed that TUG1 triggered dysfunction of pulmonary cells followed by PM exposure by serving as a sponge for miR-222-3p and thereby upregulating the expression of CELF1and p53.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Nannan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Shengnan Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Jie Xv
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Qingtao Meng
- Department of Toxicology and Sanitary chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Rui Chen
- Department of Toxicology and Sanitary chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
21
|
Fu Y, Li B, Yun J, Xu J, Meng Q, Li X, Chen R. lncRNA SOX2-OT ceRNA network enhances the malignancy of long-term PM 2.5-exposed human bronchial epithelia. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 217:112242. [PMID: 33895495 DOI: 10.1016/j.ecoenv.2021.112242] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 06/12/2023]
Abstract
Exposure to fine particulate matter (PM2.5) in outdoor air is carcinogenic and associated with the development of lung cancer; however, the underlying mechanism remains unclear. In the present study, the profiles of lncRNA, microRNA and mRNA expression profiles in human bronchial epithelia (HBE) following exposure to PM2.5, diesel exhaust particles (DEPs), or aluminum oxide nanoparticles (Al2O3 NPs) were explored by microarray to reveal the lncRNA-microRNA-mRNA network participating in the malignant transformation of HBE cells following long-term PM2.5 exposure. The results showed that lncRNA SOX2 overlapping transcript (SOX2-OT) was significantly induced in HBE cells exposed to PM2.5, DEPs, or Al2O3 NPs, acting as a sponge to microRNA-345-5p, which subsequently increased the expression levels of epidermal growth factor receptor (EGFR). EGFR is a therapeutic target in non-small cell lung cancer. Here, we found that SOX2-OT is an upstream trigger of EGFR in HBE cells during long-term PM2.5 exposure. Importantly, SOX2-OT knockdown effectively reduced the colony formation and migration capacities of HBE cells, compared to the wild type control. Collectively, SOX2-OT/microRNA-345-5p/EGFR is a ceRNA mechanism underlying the malignant transformation of bronchial epithelia exposed to PM2.5, which improves our understanding of the association between ambient PM2.5 exposure and the development of lung cancer.
Collapse
Affiliation(s)
- You Fu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Jun Yun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Jie Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Qingtao Meng
- School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
22
|
Dolati S, Shakouri SK, Dolatkhah N, Yousefi M, Jadidi-Niaragh F, Sanaie S. The role of exosomal non-coding RNAs in aging-related diseases. Biofactors 2021; 47:292-310. [PMID: 33621363 DOI: 10.1002/biof.1715] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Aging is a biological process caused by the accumulation of senescent cells with a permanent proliferative arrest. To the influence of aging on human life expectancy, there is essential for new biomarkers which possibly will assistance in recognizing age-associated pathologies. Exosomes, which are cell-secreted nanovesicles, make available a new biomarker detection and therapeutic approach for the transfer of different molecules with high capacity. Recently, non-coding RNAs (ncRNA) which are contained in exosomes have developed as important molecules regulating the complexity of aging and relevant human diseases. The discovery of ncRNA provided perceptions into an innovative regulatory platform that could interfere with cellular senescence. The non-coding transcriptome includes a different of RNA species, spanning from short ncRNAs (<200 nucleotides) to long ncRNAs, that are >200 bp long. Upgraded evidence displays that targeting ncRNAs possibly will influence senescence pathways. In this article, we will address ncRNAs that participated in age-related and cellular senescence diseases. Growing conception of ncRNAs in the aging process possibly will be responsible for new understandings into the improvement of age-related diseases and elongated life span.
Collapse
Affiliation(s)
- Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Dolatkhah
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Qi H, Liu Y, Wang N, Xiao C. Lentinan Attenuated the PM2.5 Exposure-Induced Inflammatory Response, Epithelial-Mesenchymal Transition and Migration by Inhibiting the PVT1/miR-199a-5p/caveolin1 Pathway in Lung Cancer. DNA Cell Biol 2021; 40:683-693. [PMID: 33902331 DOI: 10.1089/dna.2020.6338] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PM2.5 plays an important role in the physiological and pathological progression of lung cancer. Lentinan exerts antitumor activity in many kinds of human cancers. Plasmacytoma variant translocation 1 (PVT1) exerts antitumor activity in many kinds of human cancers. However, the role and underlying molecular mechanism of PVT1 in the role of lentinan in PM2.5-exposed lung cancer are still largely unknown. Our study confirmed that PM2.5 exposure induced the production of inflammatory factors, epithelial-mesenchymal transition (EMT) and migration of lung cancer cells. Lentinan exerted antitumor effects by inhibiting the production of inflammatory factors, EMT, and migration of lung cancer cells. Lentinan suppressed PM2.5 exposure-induced cellular progression by inhibiting the PM2.5 exposure-induced elevation of PVT1 expression. PVT1 absorbed miR-199a, and miR-199a inhibited caveolin1 expression and thus formed the PVT1/miR-199a/caveolin1 signaling pathway in lung cancer cells. Our study revealed that silencing of the PVT1/miR-199a/caveolin1 signaling pathway affected the role of lentinan in PM2.5-exposed lung cancer cells. Thus, this study first investigated the role of lentinan in PM2.5-exposed lung cancer cells and further displayed the underlying molecular mechanism, providing a potential treatment for PM2.5-exposed lung cancer.
Collapse
Affiliation(s)
- He Qi
- Liaoning University of Traditional Chinese Medicine, Graduate School, Shenyang, People's Republic of China.,Department of Medical Technology, Liaoning Vocational College of Medicine, Shenyang, People's Republic of China
| | - Ying Liu
- Department of Medical Technology, Liaoning Vocational College of Medicine, Shenyang, People's Republic of China
| | - Nan Wang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, People's Republic of China
| | - Chunling Xiao
- Liaoning University of Traditional Chinese Medicine, Graduate School, Shenyang, People's Republic of China.,Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, People's Republic of China
| |
Collapse
|
24
|
Hu C, Li J, Du Y, Li J, Yang Y, Jia Y, Peng L, Qin Y, Wei Y. Impact of chronic intermittent hypoxia on the long non-coding RNA and mRNA expression profiles in myocardial infarction. J Cell Mol Med 2021; 25:421-433. [PMID: 33215878 PMCID: PMC7810970 DOI: 10.1111/jcmm.16097] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/28/2020] [Accepted: 10/18/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) is the primary feature of obstructive sleep apnoea (OSA), a crucial risk factor for cardiovascular diseases. Long non-coding RNAs (lncRNAs) in myocardial infarction (MI) pathogenesis have drawn considerable attention. However, whether CIH participates in the modulation of lncRNA profiles during MI is yet unclear. To investigate the influence of CIH on MI, cardiac damage was assessed by histology and echocardiography, and lncRNA and mRNA integrated microarrays were screened. MI mouse model showed myocardial hypertrophy, aggravated inflammation and fibrosis, and compromised left ventricle function under CIH. Compared with normoxia, 644 lncRNAs and 1084 differentially expressed mRNAs were identified following CIH for 4 weeks, whereas 1482 lncRNAs and 990 mRNAs were altered at 8 weeks. Strikingly, reoxygenation after CIH markedly affected 1759 lncRNAs and 778 mRNAs. Of these, 11 lncRNAs modulated by CIH were restored after reoxygenation and were validated by qPCR. The GO terms and KEGG pathways of genes varied significantly by CIH. lncRNA-mRNA correlation further showed that lncRNAs, NONMMUT032513 and NONMMUT074571 were positively correlated with ZEB1 and negatively correlated with Cmbl. The current results demonstrated a causal correlation between CIH and lncRNA alternations during MI, suggesting that lncRNAs might be responsible for MI aggravation under CIH.
Collapse
Affiliation(s)
- Chaowei Hu
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Jing Li
- Heart Center & Beijing Key Laboratory of HypertensionBeijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Yunhui Du
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Juan Li
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Remodeling‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yunyun Yang
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Remodeling‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yifan Jia
- Department of CardiologyBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Lu Peng
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Remodeling‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yongxiang Wei
- Key Laboratory of Upper Airway Dysfunction‐related Cardiovascular DiseasesBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Otolaryngological Department of Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
25
|
Omote N, Sauler M. Non-coding RNAs as Regulators of Cellular Senescence in Idiopathic Pulmonary Fibrosis and Chronic Obstructive Pulmonary Disease. Front Med (Lausanne) 2020; 7:603047. [PMID: 33425948 PMCID: PMC7785852 DOI: 10.3389/fmed.2020.603047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is a cell fate implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Cellular senescence occurs in response to cellular stressors such as oxidative stress, DNA damage, telomere shortening, and mitochondrial dysfunction. Whether these stresses induce cellular senescence or an alternative cell fate depends on the type and magnitude of cellular stress, but also on intrinsic factors regulating the cellular stress response. Non-coding RNAs, including both microRNAs and long non-coding RNAs, are key regulators of cellular stress responses and susceptibility to cellular senescence. In this review, we will discuss cellular mechanisms that contribute to senescence in IPF and COPD and highlight recent advances in our understanding of how these processes are influenced by non-coding RNAs. We will also discuss the potential therapeutic role for targeting non-coding RNAs to treat these chronic lung diseases.
Collapse
Affiliation(s)
- Norihito Omote
- Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
26
|
Long non-coding RNA maternally expressed gene regulates cigarette smoke extract induced lung inflammation and human bronchial epithelial apoptosis via miR-149-3p. Exp Ther Med 2020; 21:60. [PMID: 33365060 PMCID: PMC7716647 DOI: 10.3892/etm.2020.9492] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has become a significant public health risk. Long non-coding RNAs (lncRNAs) have been identified as important factors involved in the proliferation, apoptosis and inflammatory cytokine expression of lung cells. Peripheral blood samples from 66 subjects (18 non-smokers, 24 smokers without COPD and 28 smokers with COPD) and HBE135-E6E7 cell treated with cigarette smoke extract (CSE) or not were used as the research object. The aim of the present study was to investigate the underlying mechanism of lncRNA maternally expressed gene 3 (MEG3) in COPD. Following transfection with microRNA (miR)-149-3p mimics, miR-negative control mimics, miR-149-3p inhibitor, miR-negative control inhibitor, small interfering (si)RNA targeting MEG3 (si-MEG3) and si-negative control (si-NC), levels of MEG3 and microRNA (miR)-149-3p were detected using reverse transcription-quantitative PCR, Proliferation and apoptosis were examined using the Cell Counting Kit-8 and flow cytometry assays, respectively. Enzyme-linked immunosorbent assay (ELISA) was performed to detect the expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Protein levels of B-cell lymphoma-2 (Bcl-2), cleaved-caspase-3, cleaved-caspase-9, phosphorylated (p)-p65, total (t)-p65, p-lkBα and t-lkBα were measured by western blotting. Luciferase assay was conducted to examine the relationship between MEG3 and miR-149-3p. LncRNA MEG3 was highly expressed, whereas miR-149-3p expression was downregulated in smokers with COPD peripheral blood samples, compared with non-smokers and smokers without COPD samples. Compared with untreated human bronchial epithelial (HBE) cells, MEG3 expression was increased in cigarette smoke extract (CSE)-treated HBE cells. Compared with CSE-treated HBE cells transfected with si-NC, MEG3 knockdown promoted cell proliferation and inhibited apoptosis in CSE-treated HBE cells transfected with si-MEG3, and it also decreased the levels of IL-6, TNF-α, Bcl-2 and increased cleaved-caspase-3 and cleaved-caspase-9 in CSE-treated HBE cells transfected with si-MEG3. The luciferase assay demonstrated that miR-149-3p has target sites for MEG3. MEG3 was demonstrated to regulate the NF-κB signaling pathway by sponging miR-149-3p in CSE-treated HBE cells. In conclusion, these findings suggested that MEG3 promoted proliferation and inhibited apoptosis by regulating the NF-κB signal pathway via miR-149-3p in CSE-treated HBE cells. These results provide an insight for further verification and understanding of the molecular basis of COPD.
Collapse
|
27
|
Fan S, Ren Y, Zhang W, Zhang H, Wang C. Long non-coding maternally expressed gene 3 regulates cigarette smoke extract-induced apoptosis, inflammation and cytotoxicity by sponging miR-181a-2-3p in 16HBE cells. Oncol Lett 2020; 21:45. [PMID: 33262837 PMCID: PMC7693283 DOI: 10.3892/ol.2020.12306] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has suggested that long non-coding (lnc)RNAs are widely involved in the progression of multiple diseases, including chronic obstructive pulmonary disease (COPD). The aim of the present study was to explore the function and molecule mechanism of maternally expressed gene 3 (MEG3) in cigarette smoke extract (CSE)-treated 16HBE cells. Cell viability and apoptosis were evaluated using Cell Counting Kit-8 analysis and flow cytometry, respectively. Western blot analysis was carried out to determine the protein levels of Bcl-2, Bax and cleaved caspase-3. ELISA assays were utilized to measure the protein levels of IL-1β and IL-6 and TNF-α. Cytotoxicity was assessed using a lactate dehydrogenase release assay. The expression levels of MEG3 and microRNA (miR)-181a-2-3p were detected using reverse transcription-quantitative PCR. The interaction between miR-181a-2-3p and MEG3 was predicted using DIANA tools and verified by a dual-luciferase reporter assay and RNA Immunoprecipitation assay. MEG3 expression was enhanced while miR-181a-2-3p abundance was reduced in the serum of patients with COPD and CSE-treated 16HBE cells. MEG3-knockdown or miR-181a-2-3p-overexpression inhibited CSE-induced apoptosis, inflammation and cytotoxicity in 16HBE cells. Moreover, miR-181a-2-3p directly bind to MEG3 and its knockdown reversed the inhibitory effect of MEG3 interference on apoptosis, inflammation and cytotoxicity in CSE-treated 16HBE cells. Overall, MEG3-knockdown suppressed CSE-induced apoptosis, inflammation and cytotoxicity in 16HBE cells by upregulating miR-181a-2-3p, providing a promising therapeutic target for treatment of CSE-induced COPD.
Collapse
Affiliation(s)
- Shiming Fan
- Department of Respiratory Medicine, Changning County Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644300, P.R. China
| | - Yan Ren
- Department of Pediatrics, Changning County Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644300, P.R. China
| | - Wenli Zhang
- Department of Gastroenterology, Changning County Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644300, P.R. China
| | - Huawei Zhang
- Department of Critical Care Medicine, Changning County Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644300, P.R. China
| | - Cheng Wang
- Department of Nephrology, Changning County Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644300, P.R. China
| |
Collapse
|
28
|
Miguel V, Lamas S, Espinosa-Diez C. Role of non-coding-RNAs in response to environmental stressors and consequences on human health. Redox Biol 2020; 37:101580. [PMID: 32723695 PMCID: PMC7767735 DOI: 10.1016/j.redox.2020.101580] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Environmental risk factors, including physicochemical agents, noise and mental stress, have a considerable impact on human health. This environmental exposure may lead to epigenetic reprogramming, including changes in non-coding RNAs (ncRNAs) signatures, which can contribute to the pathophysiology state. Oxidative stress is one of the results of this environmental disturbance by modifying cellular processes such as apoptosis, signal transduction cascades, and DNA repair mechanisms. In this review, we delineate environmental risk factors and their influence on (ncRNAs) in connection to disease. We focus on well-studied miRNAs and analyze the novel roles of long-non-coding-RNAs (lncRNAs). We discuss commonly regulated lncRNAs after exposure to different stressors, such as UV, heavy metals and pesticides among others, and the potential role of these lncRNA as exposure biomarkers, epigenetic regulators and potential therapeutic targets to diminish the deleterious secondary response to environmental agents. Environmental stressors induce epigenetic changes that lead to long-lasting gene expression changes and pathology development. NcRNAs, miRNAs and lncRNAs, are epigenetic modifiers susceptible to changes in expression after environmental insults . LncRNAs influence cell function partnering with other biomolecules such as proteins, DNA, RNA or other ncRNAs. LncRNA dysregulation affects cell development, carcinogenesis, vascular disease and neurodegenerative disorders. ncRNA signatures can be potentially used as biomarkers to identify exposure to specific environmental stressors.
Collapse
Affiliation(s)
- Verónica Miguel
- Programme of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Programme of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Cristina Espinosa-Diez
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Poulet C, Njock MS, Moermans C, Louis E, Louis R, Malaise M, Guiot J. Exosomal Long Non-Coding RNAs in Lung Diseases. Int J Mol Sci 2020; 21:E3580. [PMID: 32438606 PMCID: PMC7279016 DOI: 10.3390/ijms21103580] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Within the non-coding genome landscape, long non-coding RNAs (lncRNAs) and their secretion within exosomes are a window that could further explain the regulation, the sustaining, and the spread of lung diseases. We present here a compilation of the current knowledge on lncRNAs commonly found in Chronic Obstructive Pulmonary Disease (COPD), asthma, Idiopathic Pulmonary Fibrosis (IPF), or lung cancers. We built interaction networks describing the mechanisms of action for COPD, asthma, and IPF, as well as private networks for H19, MALAT1, MEG3, FENDRR, CDKN2B-AS1, TUG1, HOTAIR, and GAS5 lncRNAs in lung cancers. We identified five signaling pathways targeted by these eight lncRNAs over the lung diseases mentioned above. These lncRNAs were involved in ten treatment resistances in lung cancers, with HOTAIR being itself described in seven resistances. Besides, five of them were previously described as promising biomarkers for the diagnosis and prognosis of asthma, COPD, and lung cancers. Additionally, we describe the exosomal-based studies on H19, MALAT1, HOTAIR, GAS5, UCA1, lnc-MMP2-2, GAPLINC, TBILA, AGAP2-AS1, and SOX2-OT. This review concludes on the need for additional studies describing the lncRNA mechanisms of action and confirming their potential as biomarkers, as well as their involvement in resistance to treatment, especially in non-cancerous lung diseases.
Collapse
Affiliation(s)
- Christophe Poulet
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
| | - Makon-Sébastien Njock
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Catherine Moermans
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Edouard Louis
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Gastroenterology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Renaud Louis
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Michel Malaise
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
| | - Julien Guiot
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| |
Collapse
|
30
|
Pei YH, Chen J, Wu X, He Y, Qin W, He SY, Chang N, Jiang H, Zhou J, Yu P, Shi HB, Chen XH. LncRNA PEAMIR inhibits apoptosis and inflammatory response in PM2.5 exposure aggravated myocardial ischemia/reperfusion injury as a competing endogenous RNA of miR-29b-3p. Nanotoxicology 2020; 14:638-653. [PMID: 32100595 DOI: 10.1080/17435390.2020.1731857] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The sensitivity of myocardium is enhanced to ischemia/reperfusion (I/R) injury under PM2.5 exposure. It is still under prelude for lncRNA-miRNA pair in the study of aggravated myocardial I/R injury under PM2.5 exposure. In this study, we first built a rat model of 30 min ischemia and 24 h reperfusion followed PM2.5 (6.0 mg/kg) exposure. We found PM2.5 exposure could obviously aggravate I/R injury in the fields of myocardium damage, apoptosis levels and cardiac function which were evaluated by TTC staining, TUNEL and echocardiography, respectively. Then, based on results of sequencing and RT-qPCR, we selected NONRATT003473.2 in the follow-up experiments and named this lncRNA as PM2.5 exposure aggravated myocardial I/R injury lncRNA (PEAMIR). Consistent with the results rat model, we confirmed PEAMIR to be a protective lncRNA against PM + HR triggered damages in H9c2 cells. Next, according to the bioinformatics analysis from miRanda database and a series of gain- and loss-of-function experiments, we proved PEAMIR to be a ceRNA for miR-29b-3p to inhibit cardiac inflammation and apoptosis. Finally, using Target-Scan database, the conserved binding sites for miR-29b-3p was identified in the 3'UTR of PI3K (p85a), a key protein of apoptosis. Our subsequent experiments validated the regulatory relationship between PEAMIR-miR-29b-3p ceRNA pair and PI3K (p85a)/Akt/GSK3b/p53 cascade pathway. In conclusion, our study demonstrated the role and mechanism of PEAMIR in the augment of I/R injury under PM2.5 exposure, suggesting a promising strategy for the prevention and treatment of I/R injury under PM2.5 exposure.
Collapse
Affiliation(s)
- Ying-Hao Pei
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Jie Chen
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Xiang Wu
- Department of Geriatrics, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Yun He
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Wei Qin
- Jiangsu Environmental Monitoring Center, Nanjing, China
| | - Shu-Yin He
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Ning Chang
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Hua Jiang
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Jiang Zhou
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Peng Yu
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Hai-Bo Shi
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Xiao-Hu Chen
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
31
|
Devadoss D, Long C, Langley RJ, Manevski M, Nair M, Campos MA, Borchert G, Rahman I, Chand HS. Long Noncoding Transcriptome in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2019; 61:678-688. [PMID: 31486667 PMCID: PMC6890411 DOI: 10.1165/rcmb.2019-0184tr] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic airway inflammation from recurring exposures to noxious environmental stimuli results in a progressive and irreversible airflow limitation and the lung parenchymal damage that characterizes chronic obstructive pulmonary disease (COPD). The large variability observed in the onset and progression of COPD is primarily driven by complex gene-environment interactions. The transcriptomic and epigenetic memory potential of lung epithelial and innate immune cells drive responses, such as mucus hyperreactivity and airway remodeling, that are tightly regulated by various molecular mechanisms, for which several candidate susceptibility genes have been described. However, the recently described noncoding RNA species, in particular the long noncoding RNAs, may also have an important role in modulating pulmonary responses to chronic inhalation of toxic substances and the development of COPD. This review outlines the features of long noncoding RNAs that have been implicated in regulating the airway inflammatory responses to cigarette smoke exposure and their possible association with COPD pathogenesis. As COPD continues to debilitate the increasingly aging population and contribute to higher morbidity and mortality rates worldwide, the search for better biomarkers and alternative therapeutic options is pivotal.
Collapse
Affiliation(s)
- Dinesh Devadoss
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Christopher Long
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Raymond J. Langley
- Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Marko Manevski
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Michael A. Campos
- Pulmonary Section, Miami Veterans Administration Medical Center, Miami, Florida
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Miller School of Medicine, University of Miami, Coral Gables, Florida; and
| | - Glen Borchert
- Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Hitendra S. Chand
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
32
|
Li N, Liu Y, Cai J. LncRNA MIR155HG regulates M1/M2 macrophage polarization in chronic obstructive pulmonary disease. Biomed Pharmacother 2019; 117:109015. [DOI: 10.1016/j.biopha.2019.109015] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/13/2019] [Accepted: 05/21/2019] [Indexed: 12/13/2022] Open
|
33
|
Wang E, Liu X, Tu W, Do DC, Yu H, Yang L, Zhou Y, Xu D, Huang S, Yang P, Ran P, Gao P, Liu Z. Benzo(a)pyrene facilitates dermatophagoides group 1 (Der f 1)-induced epithelial cytokine release through aryl hydrocarbon receptor in asthma. Allergy 2019; 74:1675-1690. [PMID: 30982974 PMCID: PMC6790621 DOI: 10.1111/all.13784] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/24/2019] [Accepted: 02/18/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Environmental pollutants, which coexist with allergens, have been associated with the exacerbation of asthma. However, the underlying molecular mechanisms remain elusive. We sought to determine whether benzo(a)pyrene (BaP) co-exposure with dermatophagoides group 1 allergen (Der f 1) can potentiate Der f 1-induced asthma and its underlying mechanisms. METHODS The effect of BaP was investigated in Der f 1-induced mouse model of asthma, including airway hyper-responsiveness, allergic inflammation, and epithelial-derived cytokines. The impact of BaP on Der f 1-induced airway epithelial cell oxidative stress (ROS) and cytokine release was further analyzed. The role of aryl hydrocarbon receptor (AhR) signaling in BaP-promoted Der f 1-induced ROS, cytokine production, and allergic inflammation was also investigated. RESULTS Compared with Der f 1, BaP co-exposure with Der f 1 led to airway hyper-responsiveness and increased lung inflammation in mouse model of asthma. Increased expression of TSLP, IL-33, and IL-25 was also found in the airways of these mice. Moreover, BaP co-exposure with Der f 1 activated AhR signaling with increased expression of AhR and CYP1A1 and promoted airway epithelial ROS generation and TSLP and IL-33, but not IL-25, expression. Interestingly, AhR antagonist CH223191 or cells with AhR knockdown abrogated the increased expression of ROS, TSLP, and IL-33. Furthermore, ROS inhibitor N-acetyl-L-cysteine (NAC) also suppressed BaP co-exposure-induced expression of epithelial TSLP, IL-33, and IL-25. Finally, AhR antagonist CH223191 and NAC inhibited BaP co-exposure with Der f 1-induced lung inflammation. CONCLUSIONS Our findings suggest that BaP facilitates Der f 1-induced epithelial cytokine release through the AhR-ROS axis.
Collapse
Affiliation(s)
- Eryi Wang
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen University School of MedicineShenzhen UniversityShenzhenChina
| | - Xiaoyu Liu
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen University School of MedicineShenzhen UniversityShenzhenChina
| | - Wei Tu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen University School of MedicineShenzhen UniversityShenzhenChina
| | - Danh C. Do
- Johns Hopkins Asthma and Allergy CenterJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Haiqiong Yu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
| | - Liteng Yang
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
| | - Yufeng Zhou
- Key Laboratory of Neonatal Disease, Ministry of Health, Children's Hospital and Institute of Biomedical SciencesFudan UniversityShanghaiChina
| | - Damo Xu
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Shau‐Ku Huang
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
- Johns Hopkins Asthma and Allergy CenterJohns Hopkins University School of MedicineBaltimoreMaryland
- National Institute of Environmental Health SciencesNational Health Research InstitutesMiaoliTaiwan
| | - Pingchang Yang
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen University School of MedicineShenzhen UniversityShenzhenChina
| | - Pixin Ran
- Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Pei‐Song Gao
- Johns Hopkins Asthma and Allergy CenterJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Zhigang Liu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital GroupShenzhenChina
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen University School of MedicineShenzhen UniversityShenzhenChina
| |
Collapse
|
34
|
Jiang L, Wang R, Fang L, Ge X, Chen L, Zhou M, Zhou Y, Xiong W, Hu Y, Tang X, Li G, Li Z. HCP5 is a SMAD3-responsive long non-coding RNA that promotes lung adenocarcinoma metastasis via miR-203/SNAI axis. Am J Cancer Res 2019; 9:2460-2474. [PMID: 31131047 PMCID: PMC6525996 DOI: 10.7150/thno.31097] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/23/2019] [Indexed: 12/24/2022] Open
Abstract
Introduction: Transforming growth factor-beta (TGFβ) signaling plays a vital role in lung adenocarcinoma (LUAD) progression. However, the involvement of TGFβ-regulated long non-coding RNAs (lncRNAs) in metastasis of LUAD remains poorly understood. Methods: We performed bioinformatic analyses to identify putative lncRNAs regulated by TGF-β/SMAD3 and validated the results by quantitative PCR in LUAD cells. We performed luciferase reporter and chromatin immunoprecipitation assays to demonstrate the transcriptional regulation of the lncRNA histocompatibility leukocyte antigen complex P5 (HCP5) we decided to focus on. Stable HCP5 knockdown and HCP5-overexpressing A549 cell variants were generated respectively, to study HCP5 function and understand its mechanism of action. We also confirmed our findings in mouse xenografts and metastasis models. We analyzed the correlation between the level of lncRNA expression with EGFR, KRAS mutations, smoke state and prognostic of LUAD patients. Results: We found that the lncRNA HCP5 is induced by TGFβ and transcriptionally regulated by SMAD3, which promotes LUAD tumor growth and metastasis. Moreover, HCP5 is overexpressed in tumor tissues of patients with LUAD, specifically in patients with EGFR and KRAS mutations and current smoker. HCP5 high expression level is positively correlated with poor prognosis of patients with LUAD. Finally, we demonstrated that upregulation of HCP5 increases the expression of Snail and Slug by sponging the microRNA-203 (miR-203) and promoting epithelial-mesenchymal transition (EMT) in LUAD cells. Conclusions: Our work demonstrates that the lncRNA HCP5 is transcriptionally regulated by SMAD3 and acts as a new regulator in the TGFβ/SMAD signaling pathway. Therefore, HCP5 can serve as a potential therapeutic target in LUAD.
Collapse
|