1
|
Wang S, Gong X, Yuan J, Huang J, Zhao R, Ji J, Wang M, Shi X, Xin W, Zhong Y, Zheng Y, Jiang Q. Iron-doped diesel exhaust early-in-life inhalation-induced cardiopulmonary toxicity in chicken embryo: Roles of ferroptosis and acyl hydrocarbon signaling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125085. [PMID: 39374763 DOI: 10.1016/j.envpol.2024.125085] [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: 07/28/2024] [Revised: 09/14/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Diesel exhaust (DE) is a major contributor to air pollution. Iron-doping could improve diesel burning efficacy and decrease emission, however, it will also change the composition of DE, potentially enhancing the toxicities. This study is aimed to assess iron-doped DE-induced cardiopulmonary toxicity in an established in ovo early-in-life inhalation exposure chicken embryo model, and to explore potential mechanisms. Ferrocene (205, 410, 820,1640 mg/L, equivalent to 75, 150, 300, 600 ppm iron mass) was added to diesel fuel, DE was collected from a diesel generator, and then exposed to embryonic day 18-19 chicken embryo via in ovo inhalation. Hatched chickens were kept for 0, 1, or 3 months, and then sacrificed. Histopathology, electrocardiography along with biochemical methods were used to assess cardiopulmonary toxicities. For mechanistic investigation, inhibitor for ferroptosis (ferrostatin-1) or Acyl hydrocarbon receptor (PDM2) were administered before DE (with or without iron-doping), and the cardiopulmonary toxicities were compared. Characterization of DE particles indicated that addition of ferrocene significantly elevated iron content. Additionally, the contents of major toxic polycyclic aromatic hydrocarbons decreased following addition of 820 mg/L ferrocene, but increased at other doses. Remarkable cardiopulmonary toxicities, in the manifestation of elevated heart rates, cardiac remodeling and cardiac/pulmonary fibrosis were observed in animals exposed to iron-doped DEs, in which the addition of ferrocene significantly enhanced the toxicities. Both ferrostatin-1 and PDM2 pretreatment could effectively alleviate the observed effects in animals exposed to iron-doped DE. Inhibition of AhR signaling seems to be capable of alleviating changes to ferroptosis related molecules following exposure to iron-doped DE, while inhibition of ferroptosis does not seem to affect AhR signaling molecules. In summary, iron-doping with ferrocene to diesel enhanced DE-induced cardiopulmonary toxicities in chicken embryos. Ferroptosis and AhR signaling both seem to participate in this process, in which AhR signaling seems to affect ferroptosis.
Collapse
Affiliation(s)
- Siyi Wang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Xinxian Gong
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Huang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Rui Zhao
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Meinan Wang
- Qingdao Product Quality Testing Research Institute, 77 Keyuanweisi Road, Qingdao, China
| | - Xiaoyu Shi
- Qingdao Product Quality Testing Research Institute, 77 Keyuanweisi Road, Qingdao, China
| | - Wenya Xin
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
2
|
Li H, Wang J, Li Z, Wu Z, Zhang Y, Kong L, Yang Q, Wang D, Shi H, Shen G, Zou S, Zhu W, Fan K, Xu Z. Quantitative proteomics reveals the mechanism of endoplasmic reticulum stress-mediated pulmonary fibrosis in mice. Heliyon 2024; 10:e39150. [PMID: 39640640 PMCID: PMC11620036 DOI: 10.1016/j.heliyon.2024.e39150] [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: 07/11/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Pulmonary fibrosis is a progressive disease that can lead to respiratory failure. Many types of cells are involved in the progression of pulmonary fibrosis. This study utilized quantitative proteomics to investigate the mechanism of TGF-β-induced fibrosis-like changes in mouse epithelial cells. Our findings revealed that TGF-β significantly impacted biological processes related to the endoplasmic reticulum, mitochondrion, and ribonucleoprotein complex. Pull-down assay coupled with proteomics identified 114 proteins that may directly interact with TGF-β, and their functions were related to mitochondria, translation, ubiquitin ligase conjugation, mRNA processing, and actin binding. Among them, 17 molecules were also found in different expression proteins (DEPs) of quantitative proteomic, such as H1F0, MED21, SDF2L1, DAD1, and TMX1. Additionally, TGF-β decreased the folded structure and the number of ribosomes in the endoplasmic reticulum and increased the expression of key proteins in the unfolded protein response, including HRD1, PERK, and ERN1. Overall, our study suggested that TGF-β induced fibrotic changes in mouse lung epithelial cells by ER stress and initiated the unfolded protein response through the PRKCSH/IRE1 and PERK/GADD34/CHOP signaling pathways.
Collapse
Affiliation(s)
- Heng Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Jin Wang
- Department of Clinical Laboratory, Tianjin Third Central Hospital, Tianjin, 300170, People's Republic of China
| | - Ziling Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Zhidong Wu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Lingjia Kong
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Qingqing Yang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Dong Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - He Shi
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Guozheng Shen
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Wenqing Zhu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Kaiyuan Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| |
Collapse
|
3
|
Lukyanchuk A, Muraki N, Kawai T, Sato T, Hata K, Ito T, Tajima A. Long-term exposure to diesel exhaust particles induces concordant changes in DNA methylation and transcriptome in human adenocarcinoma alveolar basal epithelial cells. Epigenetics Chromatin 2024; 17:24. [PMID: 39103936 DOI: 10.1186/s13072-024-00549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Diesel exhaust particles (DEP), which contain hazardous compounds, are emitted during the combustion of diesel. As approximately one-third of the vehicles worldwide use diesel, there are growing concerns about the risks posed by DEP to human health. Long-term exposure to DEP is associated with airway hyperresponsiveness, pulmonary fibrosis, and inflammation; however, the molecular mechanisms behind the effects of DEP on the respiratory tract are poorly understood. Such mechanisms can be addressed by examining transcriptional and DNA methylation changes. Although several studies have focused on the effects of short-term DEP exposure on gene expression, research on the transcriptional effects and genome-wide DNA methylation changes caused by long-term DEP exposure is lacking. Hence, in this study, we investigated transcriptional and DNA methylation changes in human adenocarcinoma alveolar basal epithelial A549 cells caused by prolonged exposure to DEP and determined whether these changes are concordant. RESULTS DNA methylation analysis using the Illumina Infinium MethylationEPIC BeadChips showed that the methylation levels of DEP-affected CpG sites in A549 cells changed in a dose-dependent manner; the extent of change increased with increasing dose reaching the statistical significance only in samples exposed to 30 µg/ml DEP. Four-week exposure to 30 µg/ml of DEP significantly induced DNA hypomethylation at 24,464 CpG sites, which were significantly enriched for DNase hypersensitive sites, genomic regions marked by H3K4me1 and H3K27ac, and several transcription factor binding sites. In contrast, 9,436 CpG sites with increased DNA methylation levels were significantly overrepresented in genomic regions marked by H3K27me3 as well as H3K4me1 and H3K27ac. In parallel, gene expression profiling by RNA sequencing demonstrated that long-term exposure to DEP altered the expression levels of 2,410 genes, enriching 16 gene sets including Xenobiotic metabolism, Inflammatory response, and Senescence. In silico analysis revealed that the expression levels of 854 genes correlated with the methylation levels of the DEP-affected cis-CpG sites. CONCLUSIONS To our knowledge, this is the first report of genome-wide transcriptional and DNA methylation changes and their associations in A549 cells following long-term exposure to DEP.
Collapse
Affiliation(s)
- Alexandra Lukyanchuk
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Krasnoyarsk State Medical University Named After Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Naomi Muraki
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Department of Human Biology and Anatomy, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tsuyoshi Ito
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
4
|
Singh N, Nagar E, Roy D, Arora N. NLRP3/GSDMD mediated pyroptosis induces lung inflammation susceptibility in diesel exhaust exposed mouse strains. Gene 2024; 918:148459. [PMID: 38608794 DOI: 10.1016/j.gene.2024.148459] [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: 12/14/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Genetic diversity among species influences the disease severity outcomes linked to air pollution. However, the mechanism responsible for this variability remain elusive and needs further investigation. OBJECTIVE To investigate the genetic factors and pathways linked with differential susceptibility in mouse strains associated with diesel exhaust exposure. METHODS C57BL/6 and Balb/c mice were exposed to diesel exhaust (DE) for 5 days/week for 30 min/day for 8 weeks. Body weight of mice was recorded every week and airway hyperresponsiveness towards DE exposure was recorded after 24 h of last exposure. Mice were euthanised to collect BALF, blood, lung tissues for immunobiochemical assays, structural integrity and genetic studies. RESULTS C57BL/6 mice showed significantly decreased body weight in comparison to Balb/c mice (p < 0.05). Both mouse strains showed lung resistance and damage to elastance upon DE exposure compared to respective controls (p < 0.05) with more pronounced effects in C57BL/6 mice. Lung histology showed increase in bronchiolar infiltration and damage to the wall in C57BL/6 mice (p < 0.05). DE exposure upregulated pro-inflammatory and Th2 cytokine levels in C57BL/6 in comparison to Balb/c mice. C57BL/6 mice showed increase in Caspase-1 and ASC expression confirming activation of downstream pathway. This showed significant activation of inflammasome pathway in C57BL/6 mice with ∼2-fold increase in NLRP3 and elevated IL-1β expression. Gasdermin-D levels were increased in C57BL/6 mice demonstrating induction of pyroptosis that corroborated with IL-1β secretion (p < 0.05). Genetic variability among both species was confirmed with sanger's sequencing suggesting presence of SNPs in 3'UTRs of IL-1β gene influencing expression between mouse strains. CONCLUSIONS C57BL/6 mice exhibited increased susceptibility to diesel exhaust in contrast to Balb/c mice via activation of NLRP3-related pyroptosis. Differential susceptibility between strains may be attributed via SNPs in the 3'UTRs of the IL-1β gene.
Collapse
Affiliation(s)
- Naresh Singh
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ekta Nagar
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepti Roy
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Naveen Arora
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Wei X, Liu N, Feng Y, Wang H, Han W, Zhuang M, Zhang H, Gao W, Lin Y, Tang X, Zheng Y. Competitive-like binding between carbon black and CTNNB1 to ΔNp63 interpreting the abnormal respiratory epithelial repair after injury. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172652. [PMID: 38653146 DOI: 10.1016/j.scitotenv.2024.172652] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Airway epithelium is extraordinary vulnerable to damage owning to continuous environment exposure. Subsequent repair is therefore essential to restore the homeostasis of respiratory system. Disruptions in respiratory epithelial repair caused by nanoparticles exposure have been linked to various human diseases, yet implications in repair process remain incompletely elucidated. This study aims to elucidate the key stage in epithelial repair disturbed by carbon black (CB) nanoparticles, highlighting the pivotal role of ΔNp63 in mediating the epithelium repair. A competitive-like binding between CB and beta-catenin 1 (CTNNB1) to ΔNp63 is proposed to elaborate the underlying toxicity mechanism. Specifically, CB exhibits a remarkable inhibitory effect on cell proliferation, leading to aberrant airway epithelial repair, as validated in air-liquid culture. ΔNp63 drives efficient epithelial proliferation during CB exposure, and CTNNB1 was identified as a target of ΔNp63 by bioinformatics analysis. Further molecular dynamics simulation reveals that oxygen-containing functional groups on CB disrupt the native interaction of CTNNB1 with ΔNp63 through competitive-like binding pattern. This process modulates CTNNB1 expression, ultimately restraining proliferation during respiratory epithelial repair. Overall, the current study elucidates that the diminished interaction between CTNNB1 and ΔNp63 impedes respiratory epithelial repair in response to CB exposure, thereby enriching the public health risk assessment on CB-related respiratory diseases.
Collapse
Affiliation(s)
- Xiaoran Wei
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Nan Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yawen Feng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongmei Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Weizhong Han
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Min Zhuang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Hongna Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wei Gao
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Tang
- Department of Medical Chemistry, School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
6
|
Sharma S, Koshy R, Kumar R, Mohammad G, Thinlas T, Graham BB, Pasha Q. Hypobaric hypoxia drives selection of altitude-associated adaptative alleles in the Himalayan population. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 913:169605. [PMID: 38159773 PMCID: PMC11285711 DOI: 10.1016/j.scitotenv.2023.169605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Genetic variants play a crucial role in shaping the adaptive phenotypes associated with high-altitude populations. Nevertheless, a comprehensive understanding of the specific impacts of different environments associated with increasing altitudes on the natural selection of these genetic variants remains undetermined. Hence, this study aimed to identify genetic markers responsible for high-altitude adaptation with specific reference to different altitudes, majorly focussing on an altitude elevation range of ∼1500 m and a corresponding decrease of ≥5 % in ambient oxygen availability. We conducted a comprehensive genome-wide investigation (n = 192) followed by a validation study (n = 514) in low-altitude and three high-altitude populations (>2400 m) of Nubra village (NU) (3048 m), Sakti village (SKT) (3812 m), and Tso Moriri village (TK) (4522 m). Extensive genetic analysis identified 86 SNPs that showed significant associations with high-altitude adaptation. Frequency mapping of these SNPs revealed 38 adaptive alleles and specific haplotypes that exhibited a strong linear correlation with increasing altitude. Notably, these SNPs spanned crucial genes, such as ADH6 and NAPG along with the vastly studied genes like EGLN1 and EPAS1, involved in oxygen sensing, metabolism, and vascular homeostasis. Correlation analyses between these adaptive alleles and relevant clinical and biochemical markers provided evidence of their functional relevance in physiological adaptation to hypobaric hypoxia. High-altitude population showed a significant increase in plasma 8-isoPGF2α levels as compared to low-altitude population. Similar observation showcased increased blood pressure in NU as compared to TK (P < 0.0001). In silico analyses further confirmed that these alleles regulate gene expression of EGLN1, EPAS1, COQ7, NAPG, ADH6, DUOXA1 etc. This study provides genetic insights into the effects of hypobaric-hypoxia on the clinico-physiological characteristics of natives living in increasing high-altitude regions. Overall, our findings highlight the synergistic relationship between environment and evolutionary processes, showcasing physiological implications of genetic variants in oxygen sensing and metabolic pathway genes in increasing high-altitude environments.
Collapse
Affiliation(s)
- Samantha Sharma
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Department of Medical and Molecular Genetics, Indiana University, Indianapolis 46202, IN, USA
| | - Remya Koshy
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Ghulam Mohammad
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh, Ladakh 194101, India
| | - Tashi Thinlas
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh, Ladakh 194101, India
| | - Brian B Graham
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Qadar Pasha
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Institute of Hypoxia Research, New Delhi 110067, India.
| |
Collapse
|
7
|
Fang ZF, Wang ZN, Chen Z, Peng Y, Fu Y, Yang Y, Han HL, Teng YB, Zhou W, Xu D, Liu XY, Xie JX, Zhang JJ, Zhong NS. Fine particulate matter contributes to COPD-like pathophysiology: experimental evidence from rats exposed to diesel exhaust particles. Respir Res 2024; 25:14. [PMID: 38178075 PMCID: PMC10765838 DOI: 10.1186/s12931-023-02623-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Ambient fine particulate matter (PM2.5) is considered a plausible contributor to the onset of chronic obstructive pulmonary disease (COPD). Mechanistic studies are needed to augment the causality of epidemiologic findings. In this study, we aimed to test the hypothesis that repeated exposure to diesel exhaust particles (DEP), a model PM2.5, causes COPD-like pathophysiologic alterations, consequently leading to the development of specific disease phenotypes. Sprague Dawley rats, representing healthy lungs, were randomly assigned to inhale filtered clean air or DEP at a steady-state concentration of 1.03 mg/m3 (mass concentration), 4 h per day, consecutively for 2, 4, and 8 weeks, respectively. Pulmonary inflammation, morphologies and function were examined. RESULTS Black carbon (a component of DEP) loading in bronchoalveolar lavage macrophages demonstrated a dose-dependent increase in rats following DEP exposures of different durations, indicating that DEP deposited and accumulated in the peripheral lung. Total wall areas (WAt) of small airways, but not of large airways, were significantly increased following DEP exposures, compared to those following filtered air exposures. Consistently, the expression of α-smooth muscle actin (α-SMA) in peripheral lung was elevated following DEP exposures. Fibrosis areas surrounding the small airways and content of hydroxyproline in lung tissue increased significantly following 4-week and 8-week DEP exposure as compared to the filtered air controls. In addition, goblet cell hyperplasia and mucus hypersecretions were evident in small airways following 4-week and 8-week DEP exposures. Lung resistance and total lung capacity were significantly increased following DEP exposures. Serum levels of two oxidative stress biomarkers (MDA and 8-OHdG) were significantly increased. A dramatical recruitment of eosinophils (14.0-fold increase over the control) and macrophages (3.2-fold increase) to the submucosa area of small airways was observed following DEP exposures. CONCLUSIONS DEP exposures over the courses of 2 to 8 weeks induced COPD-like pathophysiology in rats, with characteristic small airway remodeling, mucus hypersecretion, and eosinophilic inflammation. The results provide insights on the pathophysiologic mechanisms by which PM2.5 exposures cause COPD especially the eosinophilic phenotype.
Collapse
Affiliation(s)
- Zhang-Fu Fang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, 518020, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhao-Ni Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhe Chen
- Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, Jiangsu, China
| | - Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yu Fu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hai-Long Han
- Global Health Research Center, Duke Kunshan University, Kunshan, 215316, Jiangsu Province, China
| | - Yan-Bo Teng
- Global Health Research Center, Duke Kunshan University, Kunshan, 215316, Jiangsu Province, China
| | - Wei Zhou
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, 518061, China
| | - Damo Xu
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, 518061, China
| | - Xiao-Yu Liu
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, 518061, China
| | - Jia-Xing Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Junfeng Jim Zhang
- Global Health Research Center, Duke Kunshan University, Kunshan, 215316, Jiangsu Province, China.
- Nicholas School of the Environment and Global Health Institute, Duke University, Durham, NC, 27708, USA.
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou Laboratory, Guangzhou, 510000, China.
| |
Collapse
|
8
|
Wu T, Liu B, Wei Y, Li Z. TGF-β Regulates m 6A RNA Methylation after PM 2.5 Exposure. TOXICS 2023; 11:1026. [PMID: 38133427 PMCID: PMC10747615 DOI: 10.3390/toxics11121026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
PM2.5 exposure leads to a variety of respiratory diseases, including pulmonary fibrosis, metastatic lung cancer, etc. Exposure to PM2.5 results in the alteration of epigenetic modification. M6A RNA methylation is an essential epigenetic modification that regulates gene expression at the post-transcriptional level. Our previous study found that PM2.5 exposure up-regulated m6A RNA methylation and TGF-β expression level in the lung, but the mechanisms and pathways of PM2.5 regulation of m6A RNA methylation are still unclear. Moreover, a previous study reported that the TGF-β signal pathway could regulate m6A RNA methylation. Based on this evidence, we investigate the role of the TGF-β signaling pathway in PM2.5-induced m6A RNA methylation with the A549 cell line. Our results showed that PM2.5 could induce upregulation of m6A RNA methylation, accompanied by increased expression of TGF-β, Smad3, methyltransferase-like 3 (METTL3), methyltransferase-like 14 (METTL14). Furthermore, these alterations induced by PM2.5 exposure could be reversed by treatment with TGF-β inhibitor. Therefore, we speculated that the TGF-β signal pathway plays an indispensable role in regulating m6A RNA methylation after PM2.5 exposure. Our study demonstrates that PM2.5 exposure influences m6A RNA methylation by inducing the alteration of the TGF-β signal pathway, which could be an essential mechanism for lung-related diseases induced by PM2.5 exposure.
Collapse
Affiliation(s)
| | | | | | - Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; (T.W.); (B.L.); (Y.W.)
| |
Collapse
|
9
|
Singh N, Nagar E, Gautam A, Kapoor H, Arora N. Resveratrol mitigates miR-212-3p mediated progression of diesel exhaust-induced pulmonary fibrosis by regulating SIRT1/FoxO3. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:166063. [PMID: 37544448 DOI: 10.1016/j.scitotenv.2023.166063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/19/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Diesel exhaust (DE) exposure contributes to the progression of chronic respiratory diseases and is associated with dysregulation of microRNA expression. The present study aims to investigate the involvement of miRNAs and target genes in DE-induced lung fibrosis. METHODS C57BL/6 mice were divided into three groups. Group 1 mice were exposed to filtered air (Control). Group 2 mice were exposed to DE for 30 min per day, 5 days per week, for 8 weeks (DE). Group 3 mice received DE exposure along with resveratrol on alternate days for the last 2 weeks (DE + RES). Mice were sacrificed to isolate RNA from lung tissue for miRNA microarray profiling. Bronchoalveolar lavage fluid and lung tissues were collected for cell count and biochemical analysis. RESULTS DE exposure resulted in differential expression of 28 miRNAs with fold change >2 (p < 0.05). The upregulated miR-212-3p was selected for further analysis. Consensus analysis revealed enrichment of SIRT1 in the FoxO pathway, along with a co-annotation of reduced body weight (p < 0.05). A549 cells transfected with a miR-212-3p inhibitor showed a dose-dependent increase in SIRT1 expression, indicating SIRT1 as a direct target. Treatment with resveratrol restored SIRT1 and miR-212-3p expression and led to a reduction in inflammatory cytokines (p < 0.05). The modulation of SIRT1 correlated negatively with macrophage infiltration, confirming its role in regulating cellular infiltration and lung inflammation. Fibronectin, alpha-SMA, and collagen levels were significantly decreased in DE + RES compared to DE group suggesting modulation of cellular functions and resolution of lung fibrosis. Furthermore, a significant decrease in FoxO3a and TGF-β gene expressions was observed upon resveratrol administration thereby downregulating pro-fibrotic pathway. CONCLUSIONS The present study demonstrates resveratrol treatment stabilizes SIRT1 gene expression by attenuating miR-212-3p in DE-exposed mice, leading to downregulation of TGF-β and FoxO3a expressions. The study highlights the therapeutic role of resveratrol in the treatment of DE-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Naresh Singh
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ekta Nagar
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anshu Gautam
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Himanshi Kapoor
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Naveen Arora
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
10
|
Yin Y, Mu C, Wang J, Wang Y, Hu W, Zhu W, Yu X, Hao W, Zheng Y, Li Q, Han W. CXCL17 Attenuates Diesel Exhaust Emissions Exposure-Induced Lung Damage by Regulating Macrophage Function. TOXICS 2023; 11:646. [PMID: 37624152 PMCID: PMC10459829 DOI: 10.3390/toxics11080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023]
Abstract
Exposure to diesel exhaust emissions (DEE) is strongly linked to innate immune injury and lung injury, but the role of macrophage chemoattractant CXCL17 in the lung damage caused by DEE exposure remains unclear. In this study, whole-body plethysmography (WBP), inflammatory cell differential count, and histopathological analysis were performed to assess respiratory parameters, airway inflammation, and airway injury in C57BL/6 male mice exposed to DEE for 3 months. qRT-PCR, IHC (immunohistochemistry), and ELISA were performed to measure the CXCL17 expression in airway epithelium or BALF (bronchoalveolar lavage fluid) following DEE/Diesel exhaust particle (DEP) exposure. Respiratory parameters, airway inflammation, and airway injury were assessed in CXCL17-overexpressing mice through adeno-associated virus vector Type 5 (AAV5) infection. Additionally, an in vitro THP-1 and HBE co-culture system was constructed. Transwell assay was carried out to evaluate the effect of rh-CXCL17 (recombinant human protein-CXCL17) on THP-1 cell migration. Flow cytometry and qRT-PCR were conducted to assess the impacts of rh-CXCL17 on apoptosis and inflammation/remodeling of HBE cells. We found that the mice exposed to DEE showed abnormal respiratory parameters, accompanied by airway injury and remodeling (ciliary injury in airway epithelium, airway smooth muscle hyperplasia, and increased collagen deposition). Carbon content in airway macrophages (CCAM), but not the number of macrophages in BALF, increased significantly. CXCL17 expression significantly decreased in mice airways and HBE after DEE/DEP exposure. AAV5-CXCL17 enhanced macrophage recruitment and clearance of DEE in the lungs of mice, and it improved respiratory parameters, airway injury, and airway remodeling. In the THP-1/HBE co-culture system, rh-CXCL17 increased THP-1 cell migration while attenuating HBE cell apoptosis and inflammation/remodeling. Therefore, CXCL17 might attenuate DEE-induced lung damage by recruiting and activating pulmonary macrophages, which is expected to be a novel therapeutic target for DEE-associated lung diseases.
Collapse
Affiliation(s)
- Yize Yin
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Public Health, Qingdao University, Qingdao 266071, China;
| | - Chaohui Mu
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China;
| | - Jiahui Wang
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
| | - Yixuan Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266071, China;
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
| | - Wenmin Hu
- School of Medicine and Pharmacy, Ocean University of China, Department of Pulmonary and Critical Care Medicine, University of Health and Rehabilitation Science, Qingdao 266071, China;
| | - Wenjing Zhu
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| | - Xinjuan Yu
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| | - Wanming Hao
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China;
| | - Qinghai Li
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
| | - Wei Han
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| |
Collapse
|
11
|
Singh N, Nagar E, Arora N. Diesel exhaust exposure impairs recovery of lung epithelial and cellular damage in murine model. Mol Immunol 2023; 158:1-9. [PMID: 37254294 DOI: 10.1016/j.molimm.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/07/2023] [Accepted: 04/04/2023] [Indexed: 06/01/2023]
Abstract
Studies have investigated the relationship between diesel exhaust (DE) exposure and lung health, highlighting the potential for DE to induce pulmonary inflammation and oxidative stress. However, the resolution of inflammation upon withdrawal of DE exposure needs further investigation. Therefore, resolution of diesel exhaust-induced lung damage was studied in the murine model. Mice (6 weeks) were divided into three groups. Group 1 (control) mice were exposed to filtered air, Group 2 (DE) mice were exposed to DE (5.1 ± 0.7 mg/m3) & Group 3 (DE-FA) mice were exposed to DE followed by filtered air exposure. Airway hyper-responsiveness was recorded after 24 h of the last exposure. BALF and lung samples were collected for cytokine estimation, immunobiological assays, and western blot analysis. DE exposure showed an increase in lung resistance thereby causing alteration in lung function parameters (p < 0.05) which was restored in the DE-FA group. BALF analysis showed a significant increase in total cell count and protein content in DE with no resolution in DE-FA groups (p < 0.05). Lung histology showed no reduction in the bronchiolar thickness and damage in the DE-FA group suggesting irreversible lung damage (p < 0.05). The significant increase in inflammatory cytokine levels, and collagen deposition showed persistent inflammatory phase and lung damage in the DE-FA group(p < 0.05). ZO-1 was significantly decreased in both test groups indicating disintegrated lung epithelium where in claudin-5 expression showed increased lung permeability. A significant increase in neutrophil elastase activity and decreased expression of, Elafin, resulted in lung epithelial damage in the DE-FA group. Lung injury marker alpha1-antitrypsin was increased in DE-FA groups indicating an immune defense mechanism against neutrophil elastase. The study showed that DE exposure causes persistent lung damage via neutrophil elastase-associated disruption of the epithelial barrier integrity and membrane dysfunction.
Collapse
Affiliation(s)
- Naresh Singh
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ekta Nagar
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Naveen Arora
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
12
|
Su AL, Penning TM. Role of Human Aldo-Keto Reductases and Nuclear Factor Erythroid 2-Related Factor 2 in the Metabolic Activation of 1-Nitropyrene via Nitroreduction in Human Lung Cells. Chem Res Toxicol 2023; 36:270-280. [PMID: 36693016 PMCID: PMC9974908 DOI: 10.1021/acs.chemrestox.2c00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
1-Nitropyrene (1-NP) is a constituent of diesel exhaust and classified as a group 2A probable human carcinogen. The metabolic activation of 1-NP by nitroreduction generates electrophiles that can covalently bind DNA to form mutations to contribute to cancer causation. NADPH-dependent P450 oxidoreductase (POR), xanthine oxidase (XO), aldehyde oxidase (AOX), and NAD(P)H/quinone oxidoreductase 1 (NQO1) may catalyze 1-NP nitroreduction. We recently found that human recombinant aldo-keto reductases (AKRs) 1C1-1C3 catalyze 1-NP nitroreduction. NQO1 and AKR1C1-1C3 are genes induced by nuclear factor erythroid 2-related factor 2 (NRF2). Despite this knowledge, the relative importance of these enzymes and NRF2 to 1-NP nitroreduction is unknown. We used a combination of pharmacological and genetic approaches to assess the relative importance of these enzymes and NRF2 in the aerobic nitroreduction of 1-NP in human bronchial epithelial cells, A549 and HBEC3-KT. 1-NP nitroreduction was assessed by the measurement of 1-aminopyrene (1-AP), the six-electron reduced metabolite of 1-NP, based on its intrinsic fluorescence properties (λex and λem). We found that co-treatment of 1-NP with salicylic acid, an AKR1C1 inhibitor, or ursodeoxycholate, an AKR1C2 inhibitor, for 48 h decreased 1-AP production relative to 1-NP treatment alone (control) in both cell lines. R-Sulforaphane or 1-(2-cyano-3,12,28-trioxooleana-1,9(11)-dien-28-yl)-1H-imidazole (CDDO-Im), two NRF2 activators, each increased 1-AP production relative to control only in HBEC3-KT cells, which have inducible NRF2. Inhibitors of POR, NQO1, and XO failed to modify 1-AP production relative to control in both cell lines. Importantly, A549 wild-type cells with constitutively active NRF2 produced more 1-AP than A549 cells with heterozygous expression of NFE2L2/NRF2, which were able to produce more 1-AP than A549 cells with homozygous knockout of NFE2L2/NRF2. Together, these data show dependence of 1-NP metabolic activation on AKR1Cs and NRF2 in human lung cells. This is the second example whereby NFE2L2/NRF2 is implicated in the carcinogenicity of diesel exhaust constituents.
Collapse
Affiliation(s)
- Anthony L. Su
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Trevor M. Penning
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
13
|
Zhou X, Bao WA, Zhu X, Lin J, Fan JF, Yang Y, Du XH, Wang YZ. 3,3'-Diindolylmethane attenuates inflammation and fibrosis in radiation-induced lung injury by regulating NF-κB/TGF-β/Smad signaling pathways. Exp Lung Res 2022; 48:103-113. [PMID: 35594367 DOI: 10.1080/01902148.2022.2052208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE This study aims to investigate the protective effect of 3,3'-diindolylmethane (DIM) on the radiation-induced lung injury (RILI) model and to explore its possible mechanism. Methods: A mouse model of RILI was established by thoracic irradiation, and dexamethasone was used as a positive drug to investigate the effect of DIM on RILI mice. Lung histopathology was analyzed by HE staining and Masson staining. Then the levels of inflammatory cytokines (TGF-β, TNF-α, IL-1β, and IL-6), inflammatory cell counts, and activity of MPO were detected. The expression of TGFβ1/Smad signaling pathway-related proteins was determined by immunohistochemistry. qPCR was used to analyze the mRNA expression levels of inflammatory factors, α‑SMA and COL1A1. The expression of COX-2, NF-κB, IκBα, PI3K, and Akt proteins was assessed by Western blot. Results: Histopathological staining of lung tissues showed that DIM administration alleviated the pulmonary inflammation and fibrosis caused by RILI. Moreover, the content of inflammatory factors such as IL-1β and IL-6, the expression of NF-κB pathway-related proteins, and the counts of inflammatory cells were inhibited in lung tissue, indicating that DIM can inhibit the NF-κB pathway to reduce inflammation. In addition, DIM could down-regulate the mRNA levels of α-SMA, COL1A1, and downregulate TGFβ1, Smad3, and p-Smad2/3 in lung tissues. Conclusion: Our study confirms that DIM has the potential to treat RILI in vivo by inhibiting fibrotic and inflammatory responses in lung tissue through the TGFβ/Smad and NF-κB dual pathways, respectively.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang Province, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Wu-An Bao
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang Province, China
| | - Xiang Zhu
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Juan Lin
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Ju-Fen Fan
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang Province, China
| | - Yang Yang
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Xiang-Hui Du
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang Province, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Yue-Zhen Wang
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang Province, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| |
Collapse
|