1
|
Kim JM, Lee HL, Go MJ, Sung MJ, Heo HJ. Green tea enriched with catechins prevents particulate matter 2.5 exposure-induced cardiovascular cytotoxicity via regulation of ferroptosis in BALB/c mice. Food Sci Biotechnol 2025; 34:2255-2267. [PMID: 40351732 PMCID: PMC12064492 DOI: 10.1007/s10068-025-01831-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 05/14/2025] Open
Abstract
This study was conducted to assess the protective effects of the aqueous green tea extract (GTE) against particulate matter (PM)2.5-induced cardiac dysfunction in BALB/c mice. The GTE treatment ameliorated PM2.5-induced ferroptosis and vascular smooth muscle cells (VSMC) switching in cardiovascular A7r5 cells. The administration of GTE regulated the body weight change, heart index, serum biomarkers, and cardiac antioxidant system. GTE downregulated the inflammatory reaction by inhibiting the protein expression levels of TLR2, TLR4, NOX4, p-Akt, p-JNK, p-IκB-α, Cas-1, iNOS, Ptgs2, HO-1, TNF-α, and IL-1β. In addition, the supplement of GTE ameliorated cardiac damage by regulating the ferroptotic biomarkers such as p53, xCT, GPX4, TFR, and FtH, and mitochondrial apoptosis indicators such as Cas-3, BCl-2, and BAX. It also protected VSMC phenotype levels of SM22α, αSMA, and calponin. This study suggests that GTE might be a potential material to protect PM2.5-induced cardiac damage via ferroptosis and inflammation pathway.
Collapse
Affiliation(s)
- Jong Min Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
- Korea Food Research Institute, Wanju‑gun, 55365 Republic of Korea
| | - Hyo Lim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Min Ji Go
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Mi Jeong Sung
- Korea Food Research Institute, Wanju‑gun, 55365 Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| |
Collapse
|
2
|
Cheng FJ, Huang CE, Chen PS, Tseng YL, Yuan CS, Lai CS. New evidence on the nephrotoxicity of fine particulate matter: Potential toxic components from different emission sources. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117808. [PMID: 39904257 DOI: 10.1016/j.ecoenv.2025.117808] [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: 09/19/2024] [Revised: 12/30/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Associations exist between fine particulate matter (PM2.5) exposure and impaired kidney function. However, the specific mechanisms and components causing renal damage remain unclear. PM2.5 was collected from an industrial and a rural area. Mice were categorized according to exposure, and biochemical, western blotting, histological, and immunohistochemical analyses were performed to evaluate the impact of PM2.5 constituents on their kidneys. To assess the impact of different PM2.5 components on inflammatory responses, a study was conducted by exposing the murine macrophage cell line (RAW 264.7). The study used a chelating resin to remove the influence of heavy metals from the water extract and employed a Toll-like receptor 4 (TLR4) antagonist to eliminate the effects of endotoxin, thereby evaluating the cellular inflammatory responses induced by various PM2.5 components. The major metallic elements at the industrial site were Fe, Mg, Zn, and Ca, whereas those at site Rural were Ca, K, and Mg. PM2.5 water extracts from both sites induced inflammatory cytokine upregulation in the lungs and kidneys, and inflammatory cell infiltration, antioxidant activity downregulation, and elevated levels of kidney injury molecule 1 in the kidneys. Exposure to PM2.5 water extract increased the mRNA levels of tumor necrosis factor-α, interleukin-6, and nitrite production in RAW264.7 macrophages. The inflammatory response and nitrite production induced by the industrial-site PM2.5 water extract were significantly suppressed after treatment with a chelating resin, whereas those from the rural area were suppressed by the Toll-like receptor 4 (TLR4) antagonist. These results suggest that heavy metals are crucial factors in PM2.5-induced cellular inflammatory responses in industrial areas, whereas endotoxin receptor--TLR4 mediated inflammatory pathways are the primary factor responsible for this response in rural areas. Furthermore, at equivalent dosages, the renal toxicity induced by the water-soluble components of rural-site PM2.5 may exceed that from industrial areas.
Collapse
Affiliation(s)
- Fu-Jen Cheng
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung, Kaohsiung City 833, Taiwan, ROC; Chang Gung University College of Medicine, 259, Wenhua 1st Road, Guishan District, Taoyuan City 333, Taiwan, ROC
| | - Chien-Er Huang
- Department of Chemical and Materials Engineering, Cheng Shiu University, No. 840 Chengcing Rd., Kaohsiung City 833, Taiwan, ROC; Super Micro Mass Research and Technology Center, Cheng Shiu University, No. 840 Chengcing Rd., Kaohsiung City 833, Taiwan, ROC
| | - Pei-Shih Chen
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung City 807, Taiwan, ROC
| | - Yu-Lun Tseng
- Institute of Environmental Engineering, National Sun Yat-Sen University, 70, Lian-Hai Road, Kaohsiung City 804, Taiwan, ROC
| | - Chung-Shin Yuan
- Institute of Environmental Engineering, National Sun Yat-Sen University, 70, Lian-Hai Road, Kaohsiung City 804, Taiwan, ROC; Aerosol Science Research Center, National Sun Yat-sen University, 70, Lian-Hai Road, Kaohsiung City 804, Taiwan, ROC.
| | - Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung City 811, Taiwan, ROC.
| |
Collapse
|
3
|
Wang L, Li Z, He H, Qin L, Xu W, Tian H, Liu R, Lian X, Li W, Qi Y, Wang Z. Low-dose radiation ameliorates PM2.5-induced lung injury through non-canonical TLR1/TLR2-like receptor pathways modulated by Akkermansia muciniphila. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117625. [PMID: 39752914 DOI: 10.1016/j.ecoenv.2024.117625] [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: 08/07/2024] [Revised: 12/17/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025]
Abstract
Exposure of PM2.5 can cause different degrees of lung injury, which is referred with inflammatory response. Some evidences showed that low-dose radiation (LDR) induces hormesis in immune, however, it is unknown if LDR ameliorates the PM2.5-induced lung injury. Additionally, gut microbiota and inflammation are crucial in lung injury and the health benefits of LDR through gut microbiota need further exploration. Here, we aim to investigate the impact of LDR on PM2.5-induced lung injury in vivo and in vitro, and elucidated the potential mechanisms of anti-inflammation activated by gut microbiota. We observed that LDR ameliorated the lung damage induced by PM2.5 in mice. Additionally, after PM2.5 exposure, M1 polarization of macrophages in alveolar lavage fluid and Th1 polarization in spleen increased, pro-inflammatory cytokines (IL-1, IL-6 and TNF-α) increased and anti-inflammatory cytokines (IL-4, IL-10 and TGF-β) decreased in lung and serum. LDR could deteriorate the changes described as above. Intriguingly, Akkermansia muciniphila (Akk) differed most significantly in the gut microbiota of mice. Notably, PM2.5 activated the Toll-like receptors-induced MyD88/NF-κB pathways to mediate the pro-inflammation, and LDR could inhibited the pathway. However, the TLR1 and TLR2 continuously increased after LDR, indicating the downstream non-canonical TLR1/TLR2 pathway of Akk was activated to blunt the pro-inflammation of PM2.5. Our results strongly indicate that LDR-induced activation of gut Akk-dependent non-canonical TLR1/TLR2-like receptor pathway ameliorates lung injury and inflammation resulted from PM2.5.
Collapse
Affiliation(s)
- Li Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Zhipeng Li
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing, China
| | - Huan He
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Lijing Qin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Weiqiang Xu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Hongyuan Tian
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Rongrong Liu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Xinru Lian
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Wen Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China
| | - Yali Qi
- Department of Epidemiology, School of Public Health, Jilin Medical College, Jilin, Jilin 132013, PR China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, PR China.
| |
Collapse
|
4
|
Chen Y, Zeng M, Xie J, Xiong Z, Jin Y, Pan Z, Spanos M, Wang T, Wang H. MiR-421 mediates PM 2.5-induced endothelial dysfunction via crosstalk between bronchial epithelial and endothelial cells. Inhal Toxicol 2024; 36:501-510. [PMID: 38776440 DOI: 10.1080/08958378.2024.2356839] [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: 02/15/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE PM2.5 is closely linked to vascular endothelial injury and has emerged as a major threat to human health. Our previous research indicated that exposure to PM2.5 induced an increased release of miR-421 from the bronchial epithelium. However, the role of miR-421 in PM2.5-induced endothelial injury remains elusive. MATERIALS AND METHODS We utilized a subacute PM2.5-exposure model in mice in vivo and an acute injury cell model in vitro to simulate PM2.5-associated endothelial injury. We also used quantitative real-time polymerase chain reaction, western blot, enzyme-linked immunosorbent assay, and immunohistochemistry to investigate the role of miR-421 in PM2.5-induced endothelial injury. RESULTS Our findings reveal that inhibition of miR-421 attenuated PM2.5-induced endothelial injury and hypertension. Mechanistically, miR-421 inhibited the expression of angiotensin-converting enzyme 2 (ACE2) in human umbilical vein endothelial cells and upregulated the expression of the downstream molecule inducible nitric oxide synthase (iNOS), thereby exacerbating PM2.5-induced endothelial injury. CONCLUSIONS Our results indicate that PM2.5 exposure facilitates crosstalk between bronchial epithelial and endothelial cells via miR-421/ACE2/iNOS signaling pathway, mediating endothelial damage and hypertension. MiR-421 inhibition may offer a new strategy for the prevention and treatment of PM2.5-induced vascular endothelial injury.
Collapse
Affiliation(s)
- Yiqing Chen
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengting Zeng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
| | - Jinxin Xie
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
| | - Zhihao Xiong
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
| | - Yuxin Jin
- QianWeiChang College, Shanghai University, Shanghai, China
| | - Zihan Pan
- QianWeiChang College, Shanghai University, Shanghai, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tianhui Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Nantong, China
| | - Hongyun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Nantong, China
| |
Collapse
|
5
|
Ding R, Huang L, Yan K, Sun Z, Duan J. New insight into air pollution-related cardiovascular disease: an adverse outcome pathway framework of PM2.5-associated vascular calcification. Cardiovasc Res 2024; 120:699-707. [PMID: 38636937 DOI: 10.1093/cvr/cvae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| |
Collapse
|
6
|
Xu D, Xie Y, Cheng J, He D, Liu J, Fu S, Hu G. Amygdalin Alleviates DSS-Induced Colitis by Restricting Cell Death and Inflammatory Response, Maintaining the Intestinal Barrier, and Modulating Intestinal Flora. Cells 2024; 13:444. [PMID: 38474407 PMCID: PMC10931221 DOI: 10.3390/cells13050444] [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: 01/18/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Inflammatory bowel disease (IBD) refers to a cluster of intractable gastrointestinal disorders with an undetermined etiology and a lack of effective therapeutic agents. Amygdalin (Amy) is a glycoside extracted from the seeds of apricot and other Rosaceae plants and it exhibits a wide range of pharmacological properties. Here, the effects and mechanisms of Amy on colitis were examined via 16S rRNA sequencing, ELISA, transmission electron microscopy, Western blot, and immunofluorescence. The results showed that Amy administration remarkably attenuated the signs of colitis (reduced body weight, increased disease activity index, and shortened colon length) and histopathological damage in dextran sodium sulfate (DSS)-challenged mice. Further studies revealed that Amy administration significantly diminished DSS-triggered gut barrier dysfunction by lowering pro-inflammatory mediator levels, inhibiting oxidative stress, and reducing intestinal epithelial apoptosis and ferroptosis. Notably, Amy administration remarkably lowered DSS-triggered TLR4 expression and the phosphorylation of proteins related to the NF-κB and MAPK pathways. Furthermore, Amy administration modulated the balance of intestinal flora, including a selective rise in the abundance of S24-7 and a decline in the abundance of Allobaculum, Oscillospira, Bacteroides, Sutterella, and Shigella. In conclusion, Amy can alleviate colitis, which provides data to support the utility of Amy in combating IBD.
Collapse
Affiliation(s)
- Dianwen Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| | - Yachun Xie
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| | - Ji Cheng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| | - Dewei He
- College of Animal Science, Jilin University, Changchun 130062, China;
| | - Juxiong Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| | - Shoupeng Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| | - Guiqiu Hu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (D.X.); (Y.X.); (J.C.); (J.L.)
| |
Collapse
|
7
|
Wang S, Lin F, Zhang C, Gao D, Qi Z, Wu S, Wang W, Li X, Pan L, Xu Y, Tan B, Yang A. Xuanbai Chengqi Decoction alleviates acute lung injury by inhibiting NLRP3 inflammasome. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117227. [PMID: 37751794 DOI: 10.1016/j.jep.2023.117227] [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] [Received: 08/05/2023] [Revised: 09/20/2023] [Accepted: 09/23/2023] [Indexed: 09/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a prevalent critical respiratory disorder caused mostly by infection and other factors. However, effective drug therapies are currently lacking. Xuanbai Chengqi Decoction (XCD), a traditional Chinese medicine (TCM) prescription, is commonly employed to treat lung diseases. It has been recommended by Chinese health authorities as one of the TCM prescriptions for COVID-19. Nonetheless, its underlying mechanism for the treatment of ALI has not been fully understood. AIM OF THE STUDY The study aims to investigate the therapeutic effect of XCD on lipopolysaccharide (LPS) -induced ALI in mice and explore its anti-inflammatory mechanism involving pyroptosis. MATERIALS AND METHODS Ultra-performance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS) was employed to identify the active compounds of XCD, and quantitative analysis of the main compounds was conducted. Male C57BL/6J mice were given different doses of XCD (4.5 and 9.0 g/kg/day) or dexamethasone (5 mg/kg/day) by oral gavage for 5 consecutive days. Subsequently, ALI was induced by injecting LPS (20 mg/kg) intraperitoneally 2 h after the last administration, and serum and lung tissues were collected 8 h later. J774A.1 cells were pretreated with different doses of XCD (100, 200, 400 μg/ml) for 12 h, then incubated with LPS (1 μg/ml) for 4 h and ATP (1 mM) for 2 h to induce pyroptosis. Supernatant and cells were collected. Moreover, J774A.1 cells were transfected with an NLRP3 overexpression plasmid for 24 h, followed by subsequent experiments with XCD (400 μg/ml). Lung histopathological changes were evaluated using hematoxylin and eosin (HE) staining. To assess the efficacy of XCD on ALI/ARDS, the levels of inflammatory factors, chemokines, and proteins associated with NLRP3 inflammasome signaling pathway were evaluated. RESULTS XCD was found to ameliorate lung inflammation injury in ALI mice, and reduce the protein expression of TNF-α, IL-1β, and IL-6 in both mouse serum and J774A.1 cell supernatant. Meanwhile, XCD significantly decreased the mRNA levels of IL-1β, pro-IL-1β, CXCL1, CXCL10, TNF-α, NLRP3, NF-κB P65, and the protein expression of NLRP3, Cleaved-Caspase1, and GSDMD-N in the lung and J774A.1 cells. These effects were consistent with the NLRP3 inhibitor MCC950. Furthermore, overexpression of NLRP3 reversed the anti-inflammatory effect of XCD. CONCLUSION The therapeutic mechanism of XCD in ALI treatment may involve alleviating inflammatory responses in lung tissues by inhibiting the activation of the NLRP3 inflammasome-mediated pyroptosis in macrophages.
Collapse
Affiliation(s)
- Shun Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Feifei Lin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Chengxi Zhang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Dan Gao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Zhuocao Qi
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Suwan Wu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Wantao Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Xiaoqian Li
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| | - Lingyun Pan
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 210203, China.
| | - Yanwu Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bo Tan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Aidong Yang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 201203, China.
| |
Collapse
|
8
|
Wang BH, Tang LL, Sun XH, Zhang Q, Liu CY, Zhang XN, Yu KY, Yang Y, Hu J, Shi XL, Wang Y, Liu L. Qufeng Xuanbi Formula inhibited benzo[a]pyrene-induced aggravated asthma airway mucus secretion by AhR/ROS/ERK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117203. [PMID: 37734473 DOI: 10.1016/j.jep.2023.117203] [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: 05/30/2023] [Revised: 08/12/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Excessive secretion of airway mucus may be an important pathological factor of air pollution-induced acute asthma attacks. Treatment of airway mucus hypersecretion improves asthma aggravated by air pollutants. Qufeng Xuanbi Formula (QFXBF) has been used to treat asthma for more than 30 years. However, whether QFXBF inhibits asthmatic mucus secretion exacerbated by air pollutants has not yet been established. AIM OF THE STUDY This study aimed to evaluate the effect of QFXBF on airway mucus secretion and the mechanism of action in an air pollutant benzo[a]pyrene (BaP)-induced mouse model of aggravated asthma. MATERIALS AND METHODS Ovalbumin (OVA) and BaP co-exposure were used to establish the aggravated asthma model. The average enhanced pause (Penh), serum OVA-specific IgE, and changes in lung histopathology were determined. 16HBE cells exposed to BaP, treatment with QFXBF, arylhydrocarbon receptor (AhR) signal antagonist SR1, reactive oxygen species (ROS) antagonist NAC, or extracellular signal-regulated kinase (ERK1/2) signal antagonist U0126 were established to investigate the effect of QFXBF on BaP-induced mucus secretion and its target. The mRNA and protein expression levels of MUC5AC in the lung tissue and 16HBE cells were examined. We also studied the effect of QFXBF on ROS production. Finally, the protein expression of AhR, phospho-extracellular signal-regulated kinases (p-ERK1/2), and ERK1/2 in 16HBE cells and lung tissues was determined by western blotting. RESULTS Administration of QFXBF significantly alleviated the pathological symptoms, including Penh, serum OVA-specific IgE, and changes in lung histopathology in a BaP-induced mouse model of aggravated asthma. QFXBF inhibited MUC5AC expression in asthmatic mice and 16HBE cells exposed to BaP. ROS production, AhR expression, and ERK1/2 phosphorylation were significantly increased in BaP-induced asthmatic mice and 16HBE cells. Signaling pathway inhibitors StemRegenin 1 (SR1), NAC, and U0126 significantly inhibitedBaP-induced MUC5AC expression in 16HBE cells. SR1 reversed Bap-induced ROS production and ERK activation, and NAC inhibited Bap-induced ERK activation. In addition, QFXBF regulated AhR signaling, inhibited ROS production, reversed ERK activation, and downregulated mucus secretion to improve asthma aggravated by air pollutant BaP. CONCLUSIONS QFXBF can ameliorate mucus secretion in BaP-induced aggravated asthmatic mice and 16HBE cells, and the specific mechanism may be related to the inhibition of the AhR/ROS/ERK signaling pathway.
Collapse
Affiliation(s)
- Bo-Han Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ling-Ling Tang
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xian-Hong Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Qian Zhang
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Chun-Yang Liu
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Xiao-Na Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ke-Yao Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ying Yang
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Jun Hu
- College of Acupuncture-Moxibustion and Tuina & College of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xiao-Lu Shi
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Yue Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Li Liu
- Central Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
9
|
Tang S, Wang M, Peng Y, Liang Y, Lei J, Tao Q, Ming T, Shen Y, Zhang C, Guo J, Xu H. Armeniacae semen amarum: a review on its botany, phytochemistry, pharmacology, clinical application, toxicology and pharmacokinetics. Front Pharmacol 2024; 15:1290888. [PMID: 38323080 PMCID: PMC10844384 DOI: 10.3389/fphar.2024.1290888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Armeniacae semen amarum-seeds of Prunus armeniaca L. (Rosaceae) (ASA), also known as Kuxingren in Chinese, is a traditional Chinese herbal drug commonly used for lung disease and intestinal disorders. It has long been used to treat coughs and asthma, as well as to lubricate the colon and reduce constipation. ASA refers to the dried ripe seed of diverse species of Rosaceae and contains a variety of phytochemical components, including glycosides, organic acids, amino acids, flavonoids, terpenes, phytosterols, phenylpropanoids, and other components. Extensive data shows that ASA exhibits various pharmacological activities, such as anticancer activity, anti-oxidation, antimicrobial activity, anti-inflammation, protection of cardiovascular, neural, respiratory and digestive systems, antidiabetic effects, and protection of the liver and kidney, and other activities. In clinical practice, ASA can be used as a single drug or in combination with other traditional Chinese medicines, forming ASA-containing formulas, to treat various afflictions. However, it is important to consider the potential adverse reactions and pharmacokinetic properties of ASA during its clinical use. Overall, with various bioactive components, diversified pharmacological actions and potent efficacies, ASA is a promising drug that merits in-depth study on its functional mechanisms to facilitate its clinical application.
Collapse
Affiliation(s)
- Shun Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Minmin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhui Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanjing Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiarong Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanqiao Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Soares AG, Teixeira SA, Thakore P, Santos LG, Filho WDRP, Antunes VR, Muscará MN, Brain SD, Costa SKP. Disruption of Atrial Rhythmicity by the Air Pollutant 1,2-Naphthoquinone: Role of Beta-Adrenergic and Sensory Receptors. Biomolecules 2023; 14:57. [PMID: 38254656 PMCID: PMC10813334 DOI: 10.3390/biom14010057] [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: 11/20/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
The combustion of fossil fuels contributes to air pollution (AP), which was linked to about 8.79 million global deaths in 2018, mainly due to respiratory and cardiovascular-related effects. Among these, particulate air pollution (PM2.5) stands out as a major risk factor for heart health, especially during vulnerable phases. Our prior study showed that premature exposure to 1,2-naphthoquinone (1,2-NQ), a chemical found in diesel exhaust particles (DEP), exacerbated asthma in adulthood. Moreover, increased concentration of 1,2-NQ contributed to airway inflammation triggered by PM2.5, employing neurogenic pathways related to the up-regulation of transient receptor potential vanilloid 1 (TRPV1). However, the potential impact of early-life exposure to 1,2-naphthoquinone (1,2-NQ) on atrial fibrillation (AF) has not yet been investigated. This study aims to investigate how inhaling 1,2-NQ in early life affects the autonomic adrenergic system and the role played by TRPV1 in these heart disturbances. C57Bl/6 neonate male mice were exposed to 1,2-NQ (100 nM) or its vehicle at 6, 8, and 10 days of life. Early exposure to 1,2-NQ impairs adrenergic responses in the right atria without markedly affecting cholinergic responses. ECG analysis revealed altered rhythmicity in young mice, suggesting increased sympathetic nervous system activity. Furthermore, 1,2-NQ affected β1-adrenergic receptor agonist-mediated positive chronotropism, which was prevented by metoprolol, a β1 receptor blocker. Capsazepine, a TRPV1 blocker but not a TRPC5 blocker, reversed 1,2-NQ-induced cardiac changes. In conclusion, neonate mice exposure to AP 1,2-NQ results in an elevated risk of developing cardiac adrenergic dysfunction, potentially leading to atrial arrhythmia at a young age.
Collapse
Affiliation(s)
- Antonio G. Soares
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil; (A.G.S.); (S.A.T.); (L.G.S.); (M.N.M.)
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Simone A. Teixeira
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil; (A.G.S.); (S.A.T.); (L.G.S.); (M.N.M.)
| | - Pratish Thakore
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Cardiovascular Centre of Research Excellence, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK;
| | - Larissa G. Santos
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil; (A.G.S.); (S.A.T.); (L.G.S.); (M.N.M.)
| | - Walter dos R. P. Filho
- Fundação Jorge Duprat Figueiredo de Segurança e Medicina do Trabalho, Ministério do Trabalho e Previdência Social, Rua Capote Valente, nº 710, São Paulo 05409-002, SP, Brazil;
| | - Vagner R. Antunes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil;
| | - Marcelo N. Muscará
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil; (A.G.S.); (S.A.T.); (L.G.S.); (M.N.M.)
| | - Susan D. Brain
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Cardiovascular Centre of Research Excellence, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK;
| | - Soraia K. P. Costa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof Lineu Prestes, 1524, São Paulo 05508-000, SP, Brazil; (A.G.S.); (S.A.T.); (L.G.S.); (M.N.M.)
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Cardiovascular Centre of Research Excellence, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK;
| |
Collapse
|