1
|
Shen J, Jiang Y, Bu W, Yu M, Huang R, Tang C, Yang Z, Gao H, Su L, Cheng D, Zhao X. Protein Ubiquitination Modification in Pulmonary Fibrosis. Compr Physiol 2025; 15:e70013. [PMID: 40312137 DOI: 10.1002/cph4.70013] [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: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive fibrotic interstitial lung disease characterized by a high incidence and mortality rate, which encompasses features, such as diffuse alveolar inflammation, invasive fibroblast activation, and uncontrolled extracellular matrix (ECM) deposition. Beyond the local pathological processes, PF can be better understood in light of interorgan communication networks that are involved in its progression. Notably, pulmonary inflammation can affect cardiovascular, renal, hepatic, and neural functions, highlighting the importance of understanding these systemic interactions. Posttranslational modifications play a crucial role in regulating protein function, localization, stability, and activity. Specifically, protein ubiquitination modifications are involved in PF induced by various stimuli, involving a range of ubiquitin-modifying enzymes and substrates. In this review, we provide an overview of how E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) modulate PF through several signaling pathways, such as TGF-β, Wnt, metabolic activity, aging, ferroptosis, endoplasmic reticulum stress, and inflammatory responses. This perspective includes the role of ubiquitin-proteasome systems in interorgan communication, affecting the progression of PF and related systemic conditions. Additionally, we also summarize the currently available therapeutic compounds targeting protein ubiquitination-related enzymes or ubiquitination substrates for the treatment of PF. Understanding the interplay between ubiquitination and interorgan communication may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Jinping Shen
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Yuling Jiang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Wenxia Bu
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Mengjiao Yu
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Ruiyao Huang
- Department of Clinical Medicine, Nantong University Xinglin College, Nantong, China
| | - Can Tang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Zeyun Yang
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Haiping Gao
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, China
| | - Demin Cheng
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Xinyuan Zhao
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
2
|
Li X, Ding H, Jing J, Qian S, Ma Y, Lv M, Gao Y, Zhang Y, Li T. Sulfasalazine improves neuronal function in mice with ischemic stroke by inhibiting the STING/NF-κB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5797-5810. [PMID: 39612000 DOI: 10.1007/s00210-024-03656-w] [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: 06/06/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Inflammation plays an essential role in the pathological processes of ischemic stroke (IS). Sulfasalazine is used in clinical practice for the treatment of inflammatory diseases. This study investigated the effects of sulfasalazine in mice with IS and its underlying mechanisms. We employed an in vivo mice model of middle cerebral artery occlusion (MCAO)/reperfusion, investigating the impact of sulfasalazine on MCAO mice using tetrazolium chloride (TTC) staining, behavioral experiments, and pathological staining. Utilizing of network pharmacology methodologies, we speculated that the protective effect of sulfasalazine against IS may be related to inflammation. The effects of sulfasalazine on inflammation and polarization in oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced BV2 cells were examined through immunofluorescence and PCR techniques. Additionally, the influence of sulfasalazine on the STING/NF-κB pathway was assessed using Western blot and immunofluorescence techniques. Sulfasalazine significantly reduced the infarct area in MCAO mice, restored cerebral blood flow, and improved motor function in the MCAO mice. Pathological staining results indicated that sulfasalazine can mitigate neuronal damage in the cerebral cortex of MCAO mice. Immunofluorescence and PCR testing showed that sulfasalazine promotes M1 to M2 polarization, and reduces inflammation in OGD/R-induced BV2 cells. Furthermore, the Western blot and immunofluorescence results both confirmed that sulfasalazine can inhibit the activation of the STING/NF-κB pathway. This study elucidated the potential therapeutic role of sulfasalazine in IS through its anti-inflammatory effects and modulation of STING/NF-κB pathway, offering novel insights into treatment strategies for IS.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, Chongqing, 401331, People's Republic of China
| | - Huamin Ding
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| | - Jing Jing
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| | - Shuyu Qian
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China
| | - Yulin Ma
- School of Medicine, Shanghai University, Shanghai, China
| | - Mengting Lv
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China.
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, China.
| |
Collapse
|
3
|
Liu K, Shi M, Li X, Zeng X, Liu X. Curcumin modulates the PTEN/PI3K/AKT pathway to alleviate inflammation and oxidative stress in PM2.5-Induced chronic obstructive pulmonary disease. Food Chem Toxicol 2025; 201:115460. [PMID: 40245976 DOI: 10.1016/j.fct.2025.115460] [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: 12/19/2024] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Ambient fine particulate matter (PM2.5) contributes to the onset and escalation of chronic obstructive pulmonary disease (COPD) through the induction of inflammatory reactions and oxidative stress. Curcumin is a natural polyphenolic compound renowned for the potent antioxidant and anti-inflammatory properties. This research utilized a PM2.5-induced COPD mouse model and BEAS-2B cell line to explore the protective mechanisms of curcumin. The results showed that PM2.5 impaired pulmonary function, exacerbated airway inflammation, and caused structural damage to lung tissue. Elevated levels of inflammatory cytokines such as IL-6, IL-1β, and TNF-α, increased malondialdehyde, and reduced activities of antioxidant enzymes catalase and superoxide dismutase were observed in both mice and BEAS-2B cell line. PM2.5 exposure also suppressed PTEN expression and activated PI3K/AKT signal, and the downstream molecule NF-κB was activated and FoxO1 activity was inhibited. PTEN overexpression partially reversed PM2.5-induced inflammation and oxidative stress in vitro. Curcumin enhanced PTEN expression, inhibited PI3K/AKT and NF-κB activation, and restored FoxO1 activity, alleviating airway inflammation and oxidative stress, while PTEN inhibition attenuated the ameliorating effects of curcumin in vitro and in vivo. In summary, PM2.5 exposure induces COPD inflammation and oxidative stress by disrupting PTEN/PI3K/AKT signaling and curcumin significantly alleviates these effects partially through PTEN/PI3K/AKT signal.
Collapse
Affiliation(s)
- Kai Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Meng Shi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xin Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiaoli Zeng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Xiaoju Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Li J, Wei H, Zhang Y, Wang N, Chen J, An Z, Song J, Wu W. PM 2.5 exacerbates nasal epithelial barrier damage in allergic rhinitis mice: A crosstalk between gut microbiota and NLRP3 inflammsome. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:118140. [PMID: 40185032 DOI: 10.1016/j.ecoenv.2025.118140] [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: 12/11/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Numerous studies have demonstrated a positive correlation between the frequency and severity of allergic rhinitis (AR) with fine particulate matter (PM2.5) exposure, although the exact mechanisms remain poorly understood. This study aimed to investigate the role of gut microbiota disorder and NLRP3 pathway activation in PM2.5-induced nasal epithelial barrier damage in AR mice. The results indicated that PM2.5 could exacerbate rhinitis symptoms and epithelial barrier damage in nasal mucosa. The NLRP3 pathway-related proteins including NLRP3, Caspase-1, GSDMD, and IL-1β were elevated. Additionally, nasal mucosa injury was significantly worsen in AR mice with gut microbiota disorder. Gut Microbiomic studies indicated the Ileibacterium and Alistipes are associated with nasal injury exacerbation. Metabolomic analysis suggested that bile acid metabolism disorder is a potential contributor to aggravate nasal mucosa damage. The correlation analysis revealed that IL-1β was positively associated with Alistipes, Ileibacterium, cholic acid and PC (15:0/15:0). Alistipes was positively correlated with LPE18:2 and negatively correlated with zonula occludens-1 (ZO-1) and Claudin-1 proteins. In summary, gut microbiota disorder may cause abnormal bile acid metabolism and NLRP3 inflammasome activation, which participate in PM2.5 exposure-induced exacerbation of epithelial barrier damage in nasal mucosa. This study supplied a new insight and potential targets for prevention and treatment of AR.
Collapse
Affiliation(s)
- Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Huai Wei
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Ying Zhang
- Qinyang People's Hospital, Qinyang, Henan 454550, China
| | - Ning Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Jing Chen
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China.
| |
Collapse
|
5
|
Yu K, Yang S, Song H, Sun Z, Wang K, Zhu Y, Yang C, Hao R, Cao Y. High-Resolution Tracking of Aging-Related Small Molecules: Bridging Pollutant Exposure, Brain Aging Mechanisms, and Detection Innovations. BIOSENSORS 2025; 15:242. [PMID: 40277555 PMCID: PMC12024821 DOI: 10.3390/bios15040242] [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: 02/09/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025]
Abstract
Brain aging is a complex process regulated by genetic, environmental, and metabolic factors, and increasing evidence suggests that environmental pollutants can significantly accelerate this process by interfering with oxidative stress, neuroinflammation, and mitochondrial function-related signaling pathways. Traditional studies have focused on the direct damage of pollutants on macromolecules (e.g., proteins, DNA), while the central role of senescence-associated small molecules (e.g., ROS, PGE2, lactate) in early regulatory mechanisms has been long neglected. In this study, we innovatively proposed a cascade framework of "small molecule metabolic imbalance-signaling pathway dysregulation-macromolecule collapse", which reveals that pollutants exacerbate the dynamics of brain aging through activation of NLRP3 inflammatory vesicles and inhibition of HIF-1α. Meanwhile, to address the technical bottleneck of small molecule spatiotemporal dynamics monitoring, this paper systematically reviews the cutting-edge detection tools such as electrochemical sensors, genetically encoded fluorescent probes and antioxidant quantum dots (AQDs). Among them, AQDs show unique advantages in real-time monitoring of ROS fluctuations and intervention of oxidative damage by virtue of their ultra-high specific surface area, controllable surface modification, and free radical scavenging ability. By integrating multimodal detection techniques and mechanism studies, this work provides a new perspective for analyzing pollutant-induced brain aging and lays a methodological foundation for early intervention strategies based on small molecule metabolic networks.
Collapse
Affiliation(s)
- Keying Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Sirui Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Hongxu Song
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Zhou Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Kaichao Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuqi Zhu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Chengkai Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing 100069, China;
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| |
Collapse
|
6
|
Wang M, Kim RY, Kohonen-Corish MRJ, Chen H, Donovan C, Oliver BG. Particulate matter air pollution as a cause of lung cancer: epidemiological and experimental evidence. Br J Cancer 2025:10.1038/s41416-025-02999-2. [PMID: 40185876 DOI: 10.1038/s41416-025-02999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Air pollution has a significant global impact on human health. Epidemiological evidence strongly suggests that airborne particulate matter (PM), the dust components of polluted air, is associated with increased incidence and mortality of lung cancer. PM2.5 (PM less than 2.5 µm) from various sources carries different toxic substances, such as sulfates, organic compounds, polycyclic aromatic hydrocarbons, and heavy metals, which are considered major carcinogens that increase lung cancer risk. The incidence and mortality of lung cancer caused by PM2.5 exposure may be due to significant geographical differences, and can be influenced by various factors, including local sources of air pollution, socioeconomic conditions, and public health measures. This review aims to provide comprehensive insights into the health implications of air pollution and to inform strategies for lung cancer prevention, by summarising the relationship between exposure to PM2.5 and lung cancer development. We explore the different sources of PM2.5 and relevant carcinogenic mechanisms in the context of epidemiological studies on the development of lung cancer from various geographical regions worldwide.
Collapse
Affiliation(s)
- Meng Wang
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
| | - Richard Y Kim
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Maija R J Kohonen-Corish
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
- Sydney Local Health District, Sydney, NSW, Australia
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chantal Donovan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
7
|
Xiao X, Huang G, Yu X, Tan Y. Advances in Selenium and Related Compounds Inhibiting Multi-Organ Fibrosis. Drug Des Devel Ther 2025; 19:251-265. [PMID: 39830783 PMCID: PMC11742456 DOI: 10.2147/dddt.s488226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Selenium (Se), a critically essential trace element, plays a crucial role in diverse physiological processes within the human body, such as oxidative stress response, inflammation regulation, apoptosis, and lipid metabolism. Organ fibrosis, a pathological condition caused by various factors, has become a significant global health issue. Numerous studies have demonstrated the substantial impact of Se on fibrotic diseases. This review delves into the latest research advancements in Se and Se-related biological agents for alleviating fibrosis in the heart, liver, lungs, and kidneys, detailing their mechanisms of action within fibrotic pathways. Additionally, the article summa-rizes some of the anti-fibrotic drugs currently in clinical trials for the aforementioned organ fibroses.
Collapse
Affiliation(s)
- Xixi Xiao
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei Minzu University, Enshi, 445000, People’s Republic of China
| | - Guoquan Huang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bioapplications, Enshi, 445000, People’s Republic of China
| | - Xinqiao Yu
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, People’s Republic of China
| | - Yong Tan
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, People’s Republic of China
- Hubei Provincial Key Laboratory of Selenium Resources and Bioapplications, Enshi, 445000, People’s Republic of China
| |
Collapse
|
8
|
Li C, Zhu L, Yang Y, Zhang T, Chen C, Zhang Y, Ji W, Duan X, Xue W, Li L, Zhao J. Overexpression of FBP1 enhances dendritic cell activation and maturation by inhibiting glycolysis and promoting the secretion of IL33 in lung adenocarcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167559. [PMID: 39486659 DOI: 10.1016/j.bbadis.2024.167559] [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: 03/23/2024] [Revised: 09/20/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Fructose 1,6-diphosphatase 1 (FBP1) is an enzyme involved in gluconeogenesis and glycolysis inhibition. Dendritic cells (DCs) are antigen-presenting cells, and antigens presented to T cells activate the immune response. FBP1 inhibits the development of several tumors, and high FBP1 expression inhibits the proliferation, migration, and invasion of lung cancer cells. However, the mechanism through which FBP1 mediates the tumor immune microenvironment is unclear. This study mainly analyzed the role of FBP1 in regulating the function of DCs through metabolic reprogramming and immune microenvironment using in vitro and in vivo experiments. The positive association of FBP1 with DCs was found by bioinformatic analysis. The in vitro experiments revealed that the extracellular acidification rate and lactate level were lower in the FBP1 overexpression cells than in the control cells and that the lower lactate level reduced the inhibition of DC function. In addition, high FBP1 expression promoted the secretion of IL33 by activating the cGAS/STING/NF-κB/IL33 pathway, which was identified and verified via high-throughput sequencing and in vitro experiments. FBP1 activated the cGAS/STING pathway by increasing the degree of DNA damage, as revealed by the level of γH2AX and comet assay. IL33 enhanced the expression of the DC costimulatory molecules CD86 and HLA-DR as well as that of the functional factor IL-1β. The results demonstrated that FBP1 promoted the activation and maturation of DCs by inhibiting glycolysis and promoting the secretion of IL33 as well as by further activating the function of CD8+T cells. Finally, the humanized immune system mouse models confirmed the above role of FBP1. Thus, FBP1 may serve as a new target to cure lung adenocarcinoma, and IL33 may improve the efficiency of immune therapy in lung adenocarcinoma.
Collapse
Affiliation(s)
- Chunwei Li
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lili Zhu
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yaqi Yang
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Tengfei Zhang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Chengxin Chen
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yixing Zhang
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wenxuan Ji
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaoran Duan
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Wenhua Xue
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lifeng Li
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Medical School, Huanghe Science and Technology University, 666 Zi Jing Shan Road, Zhengzhou 450000, Henan, China.
| | - Jie Zhao
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
9
|
Cao C, Hu B, Wang J, Li W, Guo L, Sheng J, Zhang C. Swertianin Promotes Anti-Tumor activity by facilitating Macrophage M1 polarization via STING signaling. Int Immunopharmacol 2024; 142:113182. [PMID: 39298821 DOI: 10.1016/j.intimp.2024.113182] [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: 08/26/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
To investigate the mechanism by which swertiamarin (swertianin, SWE) regulates the polarization of tumor microenvironment-associated macrophages to M1 phenotype, thereby exerting anti-tumor effects.SWE promoted the formation of M1 cells and increased the proportion of CD86 + cells in both RAW264.7 and primary monocyte-derived macrophages, while activating the STING-NF-κB pathway. When STING or P65 was knocked out, the effects of SWE were antagonized, inhibiting the formation of CD86 + M1 cells. At the animal level, SWE inhibited tumor growth, activated STING-NF-κB, and promoted the formation of CD86 + cells. STING-KO inhibited the effects of SWE.SWE can activate the STING-NF-κB signal to promote macrophage M1 polarization, playing an anti-tumor role.
Collapse
Affiliation(s)
- Chenxi Cao
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Biwen Hu
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Jin Wang
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Wenyan Li
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Li Guo
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Jian Sheng
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| | - Caiqun Zhang
- The Second Affiliated Hospital of Jiaxing University, 314001, China.
| |
Collapse
|
10
|
Xiong Z, Wang Y, Li Z, Li C, Tu C, Li Z. A review on the crosstalk between non-coding RNAs and the cGAS-STING signaling pathway. Int J Biol Macromol 2024; 283:137748. [PMID: 39566795 DOI: 10.1016/j.ijbiomac.2024.137748] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
In the innate immune system, the cyclic GMP-AMP synthase (cGAS)-interferon gene stimulator (STING) pathway activates the type I interferon (IFN) response and the NF-κB pathway by recognizing double-stranded DNAs, the imbalance of which plays a pivotal role in human diseases, including cancer, autoimmune and inflammatory diseases. Non-coding RNAs (ncRNAs) are a diverse group of transcripts that do not code for proteins but regulate various targets and signaling pathways in physiological and pathological processes. Recently, there has been increasing interest in investigating the interplay between the cGAS-STING pathway and ncRNAs. In this review, we provide a concise overview of the cGAS-STING pathway and ncRNAs. Then, we specifically delve into the regulation of the cGAS-STING pathway by long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), the three major classes of ncRNAs, and the influence of the cGAS-STING pathway on the expression of ncRNAs. Furthermore, we introduce the therapeutic applications targeting the cGAS-STING pathway and ncRNA therapy, and propose the utilization of drug delivery systems to deliver ncRNAs that influence the cGAS-STING pathway. Overall, this review highlights the emerging understanding of the intricate relationship between the cGAS-STING pathway and ncRNAs, shedding light on their potential as therapeutic targets in various diseases.
Collapse
Affiliation(s)
- Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yu Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
11
|
Chandel J, Naura AS. Dynamics of Inflammatory and Pathological Changes Induced by Single Exposure of Particulate Matter (PM 2.5) in Mice: Potential Implications in COPD. Cell Biochem Biophys 2024; 82:3463-3475. [PMID: 39031246 DOI: 10.1007/s12013-024-01433-3] [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] [Accepted: 07/11/2024] [Indexed: 07/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a progressive disorder of lungs marked by chronic bronchitis and emphysema. Particulate matter (PM2.5), a major component of air pollution has been correlated with COPD incidence. The present work aimed to understand dynamics of cellular/molecular players behind PM2.5-mediated COPD pathogenesis in mice by conducting dose and time-course studies. Single intratracheal exposure of PM2.5 at a dose of either 100 or 200 μg induced inflammatory response in lungs at 4 days. Time course studies showed that inflammation once triggered by PM2.5 is progressive in nature as reflected by data on BALF inflammatory cells at 7/14 days. Similarly, various cytokines/chemokines (KC/IL-6/TNF-α/IL-1β/G-CSF/MCP-1) peak at either 7 or 14 days. However, inflammation declined sharply at 21 days. Data on LPO/GSH and activities of SOD/Catalase show induction of continuous oxidative stress in lung tissue. Next, enhanced mtROS in the CD11b+ inflammatory cells confirms the redox imbalance in neutrophils/macrophages. A continuous decline in lung function was observed till 28 days. Further, histological analysis of lung tissues at 28 days confirmed the presence of emphysematous lesions, validating the potency of PM2.5 to cause irreversible damage to lungs through complex interplay of various cellular/molecular players which may be exploited as potential preventive/therapeutic targets.
Collapse
Affiliation(s)
- Jitender Chandel
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Amarjit S Naura
- Department of Biochemistry, Panjab University, Chandigarh, India.
| |
Collapse
|
12
|
Liu H, Wang J, Xiong J, Hu Z. cGAS deficiency mitigated PM2.5-induced lung injury by inhibiting ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117321. [PMID: 39561560 DOI: 10.1016/j.ecoenv.2024.117321] [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: 07/15/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024]
Abstract
Ferroptosis emerges as one of the pivotal types of cell death during fine particulate matter (PM2.5)-induced lung injury. The recently discovered cytosolic DNA sensor, cyclic GMP-AMP synthase (cGAS), triggers the activation of the downstream adaptor protein STING by synthesizing cyclic GMP-AMP, playing vital roles in innate immunity and cell death. Nonetheless, the specific function of cGAS in lung injury caused by PM2.5 remains to be elucidated. The present study aimed to explore the involvement of cGAS in the pathogenesis of PM2.5-induced lung injury and its potential mechanisms. The expression levels of cGAS in lung tissues and different types of cells isolated from murine lungs were detected. We generated a PM2.5-induced lung injury model with cGAS conditional knockout mice in type II alveolar epithelial (AT2) cells and investigated the roles of cGAS in ferroptosis in PM2.5-treated AT2 cells. The results demonstrated that PM2.5 could upregulate the expression of cGAS in lung tissues and AT2 cells. cGAS deficiency in AT2 cells not only improved pulmonary function, including lung compliance and oxygen saturation, but also relieved lung pathological injury in mice. In terms of mechanism, the absence of cGAS in AT2 cells notably reduced lipid peroxidation and ferroptosis in lungs exposed to PM2.5, achieved by increasing the protein level of ferritin. Meanwhile, cGAS deficiency also blocked the interaction between NCOA4 and ferritin, thus decreasing ferritinophagy. Additionally, periillaldehyde, one of the cGAS inhibitors, could protect against PM2.5-induced inflammation, oxidative stress, and edema in lung tissues by downregulating cGAS. Overall, cGAS promotes ferroptosis in PM2.5-induced lung injury by enhancing NCOA4-mediated ferritinophagy and shows promise as a therapeutic option for diseases associated with PM2.5 exposure.
Collapse
Affiliation(s)
- Huasong Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei University of Medicine, Shiyan, 442000, PR China
| | - Juan Wang
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Juan Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.
| |
Collapse
|
13
|
Chen X, Wang Y, Zhang M, Du Y, He Y, Li S. Selenomethionine alleviates kidney necroptosis and inflammation by restoring lipopolysaccharide-mediated mitochondrial dynamics imbalance via the TLR4/RIPK3/DRP1 signaling pathway in laying hens. Poult Sci 2024; 103:104439. [PMID: 39504830 PMCID: PMC11577205 DOI: 10.1016/j.psj.2024.104439] [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: 08/02/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Selenomethionine (SeMet) is a beneficial organic source of selenium that is extensively used as a food additive owing to its antioxidant and anti-inflammatory properties. Due to the sensitivity of the kidneys to noxious stimuli, they are more susceptible to various injuries. To investigate the protective mechanisms of SeMet supplementation against kidney injury, we established an in vivo experimental model using laying hens treated with SeMet (0.5 mg/kg diet) and/or lipopolysaccharide (LPS) (0.2 mg/kg. BW) and an in vitro model of chicken embryo primary kidney (CEK) cells treated with SeMet (0.075 mM) and with/ without LPS (60 μg/mL). SeMet treatment alleviated the LPS-induced kidney insufficiency and mitochondrial damage. Furthermore, it reduced the expression of TLR4, RIPK3, MLKL, DRP1, NLRP3, and IL-1β in the kidneys of laying hens. RIPK3 is known to induced necroptosis and inflammation by activating of the downstream factors DRP1 and MLKL. To investigate the mechanism whereby SeMet alleviates LPS-induced necroptosis in the kidney, we pretreated CEK cells with TLR4, RIPK3, and DRP1 inhibitors. The results demonstrated that RIPK3 inhibition resulted in a significantly increased in the mitochondrial membrane potential and downregulation of DRP1. Upon the inhibition of DRP1 expression, MLKL, NLRP3, and IL-1β expression also decreased. In summary, SeMet regulates the TLR4/RIPK3/DRP1 signaling pathway to restore the LPS-induced imbalances in mitochondrial dynamics, thereby alleviating necroptosis and inflammation in the kidneys of laying hen. Selenium also increases the expression of selenoproteins. This study provides valuable information for the development of new therapeutic strategies using SeMet to alleviate kidney injury.
Collapse
Affiliation(s)
- Xinzhang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yixuan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Muyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yongzhen Du
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yujiao He
- Kekedala Animal Husbandry and Veterinary Workstation of the Fourth Division of Xinjiang Construction Corps, Kekedala 831304, PR China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
14
|
Mohammadi S, Khorasani M. Implications of the cGAS-STING pathway in diabetes: Risk factors and therapeutic strategies. Int J Biol Macromol 2024; 278:134210. [PMID: 39069057 DOI: 10.1016/j.ijbiomac.2024.134210] [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: 05/12/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus is an increasingly prevalent metabolic disorder characterized by chronic hyperglycemia and impaired insulin action. Although the pathogenesis of diabetes is multifactorial, emerging evidence suggests that chronic low-grade inflammation plays a significant role in the development and progression of the disease. The cyclic GMP-AMP synthase (cGAS) and its downstream signaling pathway, the stimulator of interferon genes (STING), have recently gained attention in the field of diabetes research. This article aims to provide an overview of the role of cGAS-STING in diabetes, focusing on its involvement in the regulation of immune responses, inflammation, insulin resistance, and β-cell dysfunction. Understanding the contribution of cGAS-STING signaling in diabetes may lead to the development of targeted therapeutic strategies for this prevalent metabolic disorder. The results section presents key findings from multiple studies on the impact of STING in diabetes. It discusses the influence of STING on inflammation levels within a diabetic environment, its effect on insulin resistance, and its implications for the development and progression of diabetes. The cGAS-STING signaling pathway plays a crucial role in the development and progression of diabetes.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Natural and Medical Sciences Research Center, University of Nizwa, 611, Oman
| | - Milad Khorasani
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran; Department of Biochemistry and Nutrition, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
15
|
Zhang X, Chen H, Pang T, Liang K, Mei J, Zhu Y, Yang J. A preliminary study of sirtuin-1 on angiotensin II-induced senescence and inflammation in abdominal aortic aneurysms. Cytojournal 2024; 21:32. [PMID: 39411167 PMCID: PMC11474752 DOI: 10.25259/cytojournal_80_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Objective Recent evidence suggests the involvement of senescence and inflammation in abdominal aortic aneurysm (AAA). Considering the role of sirtuin-1 (SIRT1) in delaying senescence, we aimed to preliminarily investigate the potential mechanism underlying the effects of SIRT1 in senescence and inflammation during AAA. Material and Methods A cell AAA model was established using angiotensin II (Ang II) as the inducer, which was applied to treat human aortic vascular smooth muscle cells (HASMCs). The senescence and cell cycle of treated HASMCs were evaluated based on senescence-associated (SA)-b-galactosidase (b-gal) assay and flow cytometry, respectively. The levels of inflammatory cytokines and proteins related to senescence-associated secretory phenotype (SASP), along with nuclear factor-kappa B (NF-kB) and mitogen-activated protein kinases (MAPK) pathways, as well as SIRT1, were gauged. The correlation between SIRT1 and NF-kB and MAPK pathway-related proteins was further estimated. Results In Ang II-treated HASMCs, reduced SIRT1 and B-cell lymphoma-2 levels yet increased levels of SASP-related proteins P16 and P21, inflammatory cytokines, as well as Bax and caspases were all visible. In the meantime, Ang II exposure enhanced the number of b-gal-positive HASMCs and promoted cell cycle arrest. SIRT1 was also repressed following Ang II treatment and negatively correlated with NF-kB and MAPK pathway-related proteins (P < 0.05). Furthermore, the overexpression of SIRT1 diminished the levels of SASP-related proteins and reduced the phosphorylation of extracellular regulated kinase 1/2 and P65 in Ang II-treated HASMCs (P < 0.05). Conclusion Taken together, our results indicate that SIRT1 overexpression attenuates the inflammatory and senescent responses of HASMCs in the Ang II-induced AAA cell model. This finding suggests that SIRT1 can be a highly promising target for clinical treatment of AAA.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianshu Pang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Liang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhua Mei
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuefeng Zhu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
Liao K, Wang F, Xia C, Xu Z, Zhong S, Bi W, Ruan J. The cGAS-STING pathway in COPD: targeting its role and therapeutic potential. Respir Res 2024; 25:302. [PMID: 39113033 PMCID: PMC11308159 DOI: 10.1186/s12931-024-02915-x] [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: 03/27/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Chronic obstructive pulmonary disease(COPD) is a gradually worsening and fatal heterogeneous lung disease characterized by airflow limitation and increasingly decline in lung function. Currently, it is one of the leading causes of death worldwide. The consistent feature of COPD is airway inflammation. Several inflammatory factors are known to be involved in COPD pathogenesis; however, anti-inflammatory therapy is not the first-line treatment for COPD. Although bronchodilators, corticosteroids and roflumilast could improve airflow and control symptoms, they could not reverse the disease. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway plays an important novel role in the immune system and has been confirmed to be a key mediator of inflammation during infection, cellular stress, and tissue damage. Recent studies have emphasized that abnormal activation of cGAS-STING contributes to COPD, providing a direction for new treatments that we urgently need to develop. Here, we focused on the cGAS-STING pathway, providing insight into its molecular mechanism and summarizing the current knowledge on the role of the cGAS-STING pathway in COPD. Moreover, we explored antagonists of cGAS and STING to identify potential therapeutic strategies for COPD that target the cGAS-STING pathway.
Collapse
Affiliation(s)
- Kexin Liao
- First Clinical Medical College, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Fengshuo Wang
- College of Pharmacy, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Chenhao Xia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Ze Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Sen Zhong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Wenqi Bi
- First Clinical Medical College, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Jingjing Ruan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
17
|
Tian M, Li F, Pei H. The cGAS-STING Pathway: A New Therapeutic Target for Ischemia-Reperfusion Injury in Acute Myocardial Infarction? Biomedicines 2024; 12:1728. [PMID: 39200193 PMCID: PMC11352180 DOI: 10.3390/biomedicines12081728] [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: 07/04/2024] [Revised: 07/18/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
The innate immune system is the body's natural defense system, which recognizes a wide range of microbial molecules (such as bacterial DNA and RNA) and abnormal molecules within cells (such as misplaced DNA, self-antigens) to play its role. DNA released into the cytoplasm activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway to initiate an immune response. Ischemia-reperfusion injury (IRI) after acute myocardial infarction refers to the phenomenon where myocardial tissue suffers further damage upon the restoration of blood flow. This issue is a significant clinical problem in the treatment of myocardial infarction, as it can diminish the effectiveness of reperfusion therapy and lead to further deterioration of cardiac function. Studies have found that the cGAS-STING signaling pathway is closely related to this phenomenon. Therefore, this review aims to describe the role of the cGAS-STING signaling pathway in ischemia-reperfusion injury after myocardial infarction and summarize the current development status of cGAS-STING pathway inhibitors and the application of nanomaterials to further elucidate the potential of this pathway as a therapeutic target.
Collapse
Affiliation(s)
- Mengxiang Tian
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410083, China; (M.T.); (H.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Fengyuan Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410083, China; (M.T.); (H.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410083, China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410083, China; (M.T.); (H.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410083, China
| |
Collapse
|
18
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [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: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
19
|
Hu R, Fan W, Li S, Zhang G, Zang L, Qin L, Li R, Chen R, Zhang L, Gu W, Zhang Y, Rajagopalan S, Sun Q, Liu C. PM 2.5-induced cellular senescence drives brown adipose tissue impairment in middle-aged mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116423. [PMID: 38705039 DOI: 10.1016/j.ecoenv.2024.116423] [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: 03/05/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Airborne fine particulate matter (PM2.5) exposure is closely associated with metabolic disturbance, in which brown adipose tissue (BAT) is one of the main contributing organs. However, knowledge of the phenotype and mechanism of PM2.5 exposure-impaired BAT is quite limited. In the study, male C57BL/6 mice at three different life phases (young, adult, and middle-aged) were simultaneously exposed to concentrated ambient PM2.5 or filtered air for 8 weeks using a whole-body inhalational exposure system. H&E staining and high-resolution respirometry were used to assess the size of adipocytes and mitochondrial function. Transcriptomics was performed to determine the differentially expressed genes in BAT. Quantitative RT-PCR, immunohistochemistry staining, and immunoblots were performed to verify the transcriptomics and explore the mechanism for BAT mitochondrial dysfunction. Firstly, PM2.5 exposure caused altered BAT morphology and mitochondrial dysfunction in middle-aged but not young or adult mice. Furthermore, PM2.5 exposure increased cellular senescence in BAT of middle-aged mice, accompanied by cell cycle arrest, impaired DNA replication, and inhibited AKT signaling pathway. Moreover, PM2.5 exposure disrupted apoptosis and autophagy homeostasis in BAT of middle-aged mice. Therefore, BAT in middle-aged mice was more vulnerable to PM2.5 exposure, and the cellular senescence-initiated apoptosis, autophagy, and mitochondrial dysfunction may be the mechanism of PM2.5 exposure-induced BAT impairment.
Collapse
Affiliation(s)
- Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200433, China
| | - Sanjay Rajagopalan
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China.
| |
Collapse
|
20
|
Zhang J, Du J, Liu D, Zhuo J, Chu L, Li Y, Gao L, Xu M, Chen W, Huang W, Xie L, Chen J, Meng X, Zou F, Cai S, Dong H. Polystyrene microplastics induce pulmonary fibrosis by promoting alveolar epithelial cell ferroptosis through cGAS/STING signaling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116357. [PMID: 38677073 DOI: 10.1016/j.ecoenv.2024.116357] [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: 12/25/2023] [Revised: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Polystyrene microplastics (PS-MPs) are new types of environmental pollutant that have garnered significant attention in recent years since they were found to cause damage to the human respiratory system when they are inhaled. The pulmonary fibrosis is one of the serious consequences of PS-MPs inhalation. However, the impact and underlying mechanisms of PS-MPs on pulmonary fibrosis are not clear. In this study, we studied the potential lung toxicity and PS-MPs-developed pulmonary fibrosis by long-term intranasal inhalation of PS-MPs. The results showed that after exposing to the PS-MPs, the lungs of model mouse had different levels of damage and fibrosis. Meanwhile, exposing to the PS-MPs resulted in a markedly decrease in glutathione (GSH), an increase in malondialdehyde (MDA), and iron overload in the lung tissue of mice and alveolar epithelial cells (AECs). These findings suggested the occurrence of PS-MP-induced ferroptosis. Inhibitor of ferroptosis (Fer-1) had alleviated the PS-MPs-induced ferroptosis. Mechanically, PS-MPs triggered cell ferroptosis and promoted the development of pulmonary fibrosis via activating the cGAS/STING signaling pathway. Inhibition of cGAS/STING with G150/H151 attenuated pulmonary fibrosis after PS-MPs exposure. Together, these data provided novel mechanistic insights of PS-MPs-induced pulmonary fibrosis and a potential therapeutic paradigm.
Collapse
Affiliation(s)
- Jinming Zhang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiangzhou Du
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinzhong Zhuo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanqun Li
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Respiratory and Critical Care Medicine, Ganzhou people's Hospital, Ganzhou, China
| | - Lin Gao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingming Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimou Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wufeng Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lingyan Xie
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junwei Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Occupational Health and Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Occupational Health and Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
21
|
An C, Li Z, Chen Y, Huang S, Yang F, Hu Y, Xu T, Zhang C, Ge S. The cGAS-STING pathway in cardiovascular diseases: from basic research to clinical perspectives. Cell Biosci 2024; 14:58. [PMID: 38720328 PMCID: PMC11080250 DOI: 10.1186/s13578-024-01242-4] [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: 10/11/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
The cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase-stimulator of interferon genes (cGAS-STING) signaling pathway, an important component of the innate immune system, is involved in the development of several diseases. Ectopic DNA-induced inflammatory responses are involved in several pathological processes. Repeated damage to tissues and metabolic organelles releases a large number of damage-associated molecular patterns (mitochondrial DNA, nuclear DNA, and exogenous DNA). The DNA fragments released into the cytoplasm are sensed by the sensor cGAS to initiate immune responses through the bridging protein STING. Many recent studies have revealed a regulatory role of the cGAS-STING signaling pathway in cardiovascular diseases (CVDs) such as myocardial infarction, heart failure, atherosclerosis, and aortic dissection/aneurysm. Furthermore, increasing evidence suggests that inhibiting the cGAS-STING signaling pathway can significantly inhibit myocardial hypertrophy and inflammatory cell infiltration. Therefore, this review is intended to identify risk factors for activating the cGAS-STING pathway to reduce risks and to simultaneously further elucidate the biological function of this pathway in the cardiovascular field, as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Zhen Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Shaojun Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Fan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ying Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China.
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
22
|
Zhang Y, Shen Z, Pei H, Wang G, Wang Z, Wei X, Yu J, Wang C, Hua J, He B. Impact of particulate-matter air pollution on 25-hydroxyvitamin D levels: a mendelian randomisation study. Public Health 2024; 230:190-197. [PMID: 38565065 DOI: 10.1016/j.puhe.2024.03.002] [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: 10/28/2023] [Revised: 02/12/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES In observational studies, the 25-hydroxyvitamin D (25(OH)D) level in body has been found to be closely related to particulate matter (PM) air pollution. In this study, we used the two-sample mendelian randomisation (MR) method to investigate and discuss the potential causal relationship and mode of influence. STUDY DESIGN MR study. METHODS PM data (PM10, PM2.5-10, PM2.5, PM2.5 absorbance) came from the UK Biobank database, and 25(OH)D data came from European Bioinformatics Institute (EBI) database. The analysis was conducted utilising three prominent methods (inverse-variance-weighted [IVW], MR-Egger, weighted median, weighted mode, and simple mode). The primary emphasis was placed on IVW, accompanied by heterogeneity and horizontal pleiotropy tests. Furthermore, sensitivity analysis was undertaken. RESULTS The MR analysis revealed a significant association between exposure to PM10 and a decrease in levels of 25(OH)D (odds ratio [OR]: 0.878, 95% confidence interval [CI]: 0.789-0.977). However, no significant relationship was observed between PM2.5 exposure and 25(OH)D (OR: 0.943, 95%CI: 0.858-1.037). Further analysis indicated that the main contributor to the decline in 25(OH)D levels is linked to PM2.5-10 exposure (OR: 0.840, 95%CI: 0.751-0.940) and PM2.5 absorbance (OR: 0.875, 95%CI: 0.824-0.929). No heterogeneity and horizontal pleiotropy existed. CONCLUSIONS The MR results suggest that PM (PM10, PM2.5-10 and PM2.5 absorbance) exposure lowers vitamin D (VD) levels, but PM2.5 was not found to have a significant effect on VD in humans.
Collapse
Affiliation(s)
- Yi Zhang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zan Shen
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hang Pei
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Guanyin Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ziyue Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xinshi Wei
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jinsheng Yu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chao Wang
- Anji County Hospital of Chinese Medicine, Zhejiang, China
| | - Jiang Hua
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.
| | - Bangjian He
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.
| |
Collapse
|
23
|
Yan Q, Zheng R, Li Y, Hu J, Gong M, Lin M, Xu X, Wu J, Sun S. PM 2.5-induced premature senescence in HUVECs through the SIRT1/PGC-1α/SIRT3 pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 921:171177. [PMID: 38402989 DOI: 10.1016/j.scitotenv.2024.171177] [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: 11/21/2023] [Revised: 02/08/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Vascular endothelial cell senescence plays a pivotal role in the development of atherosclerosis. Recent studies have demonstrated that ambient fine particulate matter (PM2.5) induces stress-induced premature senescence (SIPS) in vascular endothelial cells. However, the precise mechanisms underlying this process remain to be fully elucidated. Cellular senescence is closely associated with reactive oxygen species (ROS), and emerging research has established a strong connection between the SIRT1/PGC-1α/SIRT3 signaling pathway and the antioxidant system in vascular endothelial cells. In this study, we aimed to investigate the impact of PM2.5 on vascular endothelial cell senescence and to elucidate the underlying mechanisms. Our findings revealed that PM2.5 exposure led to an increase in senescence-associated β-galactosidase (SA-β-gal) activity and the expression of the cell cycle-blocking proteins P53/P21 and P16 in human umbilical vein endothelial cells (HUVECs). Flow cytometry analysis demonstrated an elevated proportion of cells arrested in the G0/G1 phase after PM2.5 exposure. In addition, PM2.5-induced cellular senescence was attributed to the disruption of the cellular antioxidative defense system through the SIRT1/PGC-1α/SIRT3 signaling pathway. The expression of cellular senescence markers was reduced after targeted scavenging of mitochondrial ROS using MitoQ. Moreover, treatment with SRT1720, a SIRT1-specific activator, upregulated the SIRT1/PGC-1α/SIRT3 signaling pathway, restored the antioxidant system, and attenuated the expression of cellular senescence markers. Taken together, our results suggest that PM2.5 downregulates the SIRT1/PGC-1α/SIRT3 signaling pathway, resulting in impaired antioxidant defenses in HUVECs. This, in turn, allows for the accumulation of ROS, leading to inhibition of endothelial cell cycle progression and the onset of stress-induced senescence in HUVECs.
Collapse
Affiliation(s)
- Qing Yan
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Rao Zheng
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Yi Li
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Juan Hu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Meidi Gong
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Manman Lin
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xuecong Xu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jing Wu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China.
| | - Shikun Sun
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
24
|
Hou T, Zhang J, Wang Y, Zhang G, Li S, Fan W, Li R, Sun Q, Liu C. Early Pulmonary Fibrosis-like Changes in the Setting of Heat Exposure: DNA Damage and Cell Senescence. Int J Mol Sci 2024; 25:2992. [PMID: 38474239 DOI: 10.3390/ijms25052992] [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: 01/01/2024] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
It is well known that extreme heat events happen frequently due to climate change. However, studies examining the direct health impacts of increased temperature and heat waves are lacking. Previous reports revealed that heatstroke induced acute lung injury and pulmonary dysfunction. This study aimed to investigate whether heat exposure induced lung fibrosis and to explore the underlying mechanisms. Male C57BL/6 mice were exposed to an ambient temperature of 39.5 ± 0.5 °C until their core temperature reached the maximum or heat exhaustion state. Lung fibrosis was observed in the lungs of heat-exposed mice, with extensive collagen deposition and the elevated expression of fibrosis molecules, including transforming growth factor-β1 (TGF-β1) and Fibronectin (Fn1) (p < 0.05). Moreover, epithelial-mesenchymal transition (EMT) occurred in response to heat exposure, evidenced by E-cadherin, an epithelial marker, which was downregulated, whereas markers of EMT, such as connective tissue growth factor (CTGF) and the zinc finger transcriptional repressor protein Slug, were upregulated in the heat-exposed lung tissues of mice (p < 0.05). Subsequently, cell senescence examination revealed that the levels of both senescence-associated β-galactosidase (SA-β-gal) staining and the cell cycle protein kinase inhibitor p21 were significantly elevated (p < 0.05). Mechanistically, the cGAS-STING signaling pathway evoked by DNA damage was activated in response to heat exposure (p < 0.05). In summary, we reported a new finding that heat exposure contributed to the development of early pulmonary fibrosis-like changes through the DNA damage-activated cGAS-STING pathway followed by cellular senescence.
Collapse
Affiliation(s)
- Tong Hou
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Jiyang Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Yindan Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| |
Collapse
|
25
|
Wu W, Wang Z, Zhang H, Zhang X, Tian H. circGRHPR inhibits aberrant epithelial-mesenchymal transformation progression of lung epithelial cells associated with idiopathic pulmonary fibrosis. Cell Biol Toxicol 2024; 40:7. [PMID: 38267743 PMCID: PMC10808371 DOI: 10.1007/s10565-024-09839-8] [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: 09/09/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Air pollution has greatly increased the risk of idiopathic pulmonary fibrosis (IPF). Circular RNAs (circRNAs) have been found to play a significant role in the advancement of IPF, but there is limited evidence of correlation between circRNAs and lung epithelial cells (LECs) in IPF. This research aimed to explore the influence of circRNAs on the regulation of EMT progression in LECs, with the objective of elucidating its mechanism and establishing its association with IPF. Our results suggested that the downregulation of circGRHPR in peripheral blood of clinical cases was associated with the diagnosis of IPF. Meanwhile, we found that circGRHPR was downregulated in transforming growth factor-beta1 (TGF-β1)-induced A549 and Beas-2b cells. It is a valid model to study the abnormal EMT progression of IPF-associated LECs in vitro. The overexpression of circGRHPR inhibited the abnormal EMT progression of TGF-β1-induced LECs. Furthermore, as the sponge of miR-665, circGRHPR released the expression of E3 ubiquitin-protein ligase NEDD4-like (NEDD4L), thus promoting its downstream transforming growth factor beta receptor 2 (TGFBR2) ubiquitination. It is helpful to reduce the response of LECs to TGF-β1 signaling. In summary, circGRHPR/miR-665/NEDD4L axis inhibited the abnormal EMT progression of TGF-β1-induced LECs by promoting TGFBR2 ubiquitination, which provides new ideas and potential targets for the treatment of IPF.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, People's Republic of China
| | - Huiying Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| |
Collapse
|
26
|
Li Q, Wang S, Guo P, Feng Y, Yu W, Zhang H, Guo J, Li Y, Hu L, Pan J, Liao J, Tang Z. Mitochondrial DNA release mediated by TFAM deficiency promotes copper-induced mitochondrial innate immune response via cGAS-STING signalling in chicken hepatocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167315. [PMID: 37742962 DOI: 10.1016/j.scitotenv.2023.167315] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Copper (Cu) is pollution metal that is a global concern due to its toxic effects. A recent study found that the release of mitochondrial DNA (mtDNA) into the cytoplasm can activate the innate immune response, but the exact mechanisms underlying the effect of Cu exposure remains unknown. In this study, we identified that the reduction in transcription Factor A (TFAM) led to mtDNA leakage into the cytoplasm under Cu exposure in hepatocytes, accompanied by the activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway-mediated innate immunity (increased expression of cGAS, STING, TANK-binding kinase-1 (TBK1), and interferon regulatory factor-3 (IRF3)) genes and proteins, and enhanced phosphorylation levels of TBK1 and IRF3). Subsequently, silencing TFAM (siTFAM) significantly aggravated mtDNA release and the innate immune response under Cu treatment. Mitochondrial DNA depletion alleviated Cu-induced innate immunity in hepatocytes, while mtDNA transfection further enhanced the innate immune response. Notably, the inhibition of STING effectively alleviated the phosphorylation levels of the TBK1 and IRF3 proteins induced by Cu, while the upregulation of STING aggravated the Cu-induced innate immunity. Furthermore, EtBr and H-151(a STING inhibitor) treatment dramatically reversed the effect of TFAM depletion on the sharpened innate immune response induced by Cu via the cGAS-STING pathway. In general, these findings demonstrated the TFAM deficiency promotes innate immunity by activating the mtDNA-cGAS-STING signalling pathway under Cu exposure in hepatocytes, providing new insight into Cu toxicology.
Collapse
Affiliation(s)
- Quanwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Shaofeng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Pan Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Yuanhong Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Wenlan Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
27
|
Juszczak-Czasnojć M, Tomza-Marciniak A, Pilarczyk B, Gączarzewicz D. Total Selenium Level and Its Distribution between Organs in Beef Cattle in Different Selenium Status. Animals (Basel) 2023; 13:3885. [PMID: 38136922 PMCID: PMC10740525 DOI: 10.3390/ani13243885] [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: 11/09/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
The aim of this study was to determine the Se concentration in the main tissues of beef cattle and to evaluate the differences in tissue distribution between animals with different selenium status. Selenium concentration was determined in the serum, longissimus dorsi muscle, semitendinosus muscle, kidney, heart, liver, spleen and lungs of cows, heifers and beef bulls, using spectrofluorimetric method. Despite receiving supplementation, 55.6% animals demonstrated an optimal Se level, while 44.4% were deficient. The mean serum Se concentration was significantly higher (p < 0.05) in animals with a normal Se status than in Se-deficient animals. Differences in Se tissue distribution were observed between Se-deficient animals and those with normal Se status. The organs most susceptible to Se deficiency are the semitendinosus muscle, lungs, heart and liver. In both normal and Se-deficient animals, significantly higher Se concentrations were observed in the kidney than other organs (p < 0.05), and the lowest in the muscles. As Se deficiencies can be found among supplemented animals, the level of Se should be monitored in beef cattle in order to detect possible Se deficiencies, which may have negative health effects for animals and reduce the value of animal products as a source of Se in the human diet.
Collapse
Affiliation(s)
- Marta Juszczak-Czasnojć
- Department of Animal Reproduction Biotechnology and Environmental Hygiene, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (A.T.-M.)
| | | | | | | |
Collapse
|
28
|
Wen J, Mu W, Li H, Yan Y, Zhan X, Luo W, Wang Z, Kan W, Zhao J, Hui S, He P, Qin S, Xu Y, Zhang P, Xiao X, Xu G, Bai Z. Glabridin improves autoimmune disease in Trex1-deficient mice by reducing type I interferon production. Mol Med 2023; 29:167. [PMID: 38066431 PMCID: PMC10709943 DOI: 10.1186/s10020-023-00754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The cGAS-STING signaling pathway is an essential section of the natural immune system. In recent years, an increasing number of studies have shown a strong link between abnormal activation of the cGAS-STING signaling pathway, a natural immune pathway mediated by the nucleic acid receptor cGAS, and the development and progression of autoimmune diseases. Therefore, it is important to identify an effective compound to specifically downregulate this pathway for disease. METHODS The effect of Glabridin (Glab) was investigated in BMDMs and Peripheral blood mononuclear cell (PBMC) by establishing an in vitro model of cGAS-STING signaling pathway activation. An activation model stimulated by DMXAA was also established in mice to study the effect of Glab. On the other hand, we investigated the possible mechanism of action of Glab and the effect of Glab on Trex1-deficient mice. RESULTS In this research, we report that Glab, a major component of licorice, specifically inhibits the cGAS-STING signaling pathway by inhibiting the level of type I interferon and inflammatory cytokines (IL-6 and TNF-α). In addition, Glab has a therapeutic effect on innate immune diseases caused by abnormal cytoplasmic DNA in Trex1-deficient mice. Mechanistically, Glab can specifically inhibit the interaction of STING with IRF3. CONCLUSION Glab is a specific inhibitor of the cGAS-STING signaling pathway and may be used in the clinical therapy of cGAS-STING pathway-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Jincai Wen
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
- National Key Laboratory of Kidney Diseases, Beijing, 100005, China
| | - Wenqing Mu
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, Jiangsu, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Hui Li
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Yulu Yan
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaoyan Zhan
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
- National Key Laboratory of Kidney Diseases, Beijing, 100005, China
| | - Wei Luo
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Zhongxia Wang
- Nutrition Department of the Fifth Medical Center of the PLA General Hospital, Beijing, 100039, China
| | - Wen Kan
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Jia Zhao
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Siwen Hui
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Ping He
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, People's Republic of China
| | - Yingjie Xu
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China
| | - Ping Zhang
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, 100039, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China.
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100005, China.
| | - Guang Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China.
- Fifth Medical Center of Chinese, China Military Institute of Chinese Materia, PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100005, China.
| |
Collapse
|
29
|
Wei M, Cong Y, Lei J, Du R, Yang M, Lu X, Jiang Y, Cao R, Meng X, Jiang Z, Song L. The role of ROS-pyroptosis in PM 2.5 induced air-blood barrier destruction. Chem Biol Interact 2023; 386:110782. [PMID: 37884181 DOI: 10.1016/j.cbi.2023.110782] [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: 08/24/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Fine particulate matter (PM2.5) has attracted increasing attention due to its health-threatening effects. Although numerous studies have investigated the impact of PM2.5 on lung injuries, the specific mechanisms underlying the damage to the air-blood barrier after exposure to PM2.5 remain unclear. In this study, we established an in vitro co-culture system using lung epithelial cells and capillary endothelial cells. Our findings indicated that the tight junction (TJ) proteins were up-regulated in the co-cultured system compared to the monolayer-cultured cells, suggesting the establishment of a more closely connected in vitro system. Following exposure to PM2.5, we observed damage to the air-blood barrier in vitro. Concurrently, PM2.5 exposure induced significant oxidative stress and activated the NLRP3 inflammasome-mediated pyroptosis pathway. When oxidative stress was inhibited, we observed a decrease in pyroptosis and an increase in TJ protein levels. Additionally, disulfiram reversed the adverse effects of PM2.5, effectively suppressing pyroptosis and ameliorating air-blood barrier dysfunction. Our results indicate that the oxidative stress-pyroptosis pathway plays a critical role in the disruption of the air-blood barrier induced by PM2.5 exposure. Disulfiram may represent a promising therapeutic option for mitigating PM2.5-related lung damage.
Collapse
Affiliation(s)
- Min Wei
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China; Linfen Meternity & Child Healthcare Hospital, Linfen, Shanxi Province, 041000, PR China
| | - Ying Cong
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Jinrong Lei
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Rui Du
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Mengxin Yang
- Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, PR China
| | - Xinjun Lu
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning Province, 116000, PR China
| | - Yizhu Jiang
- Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, PR China
| | - Ran Cao
- Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, PR China
| | - Xianzong Meng
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Zhenfu Jiang
- Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, PR China
| | - Laiyu Song
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China.
| |
Collapse
|
30
|
Manzano-Covarrubias AL, Yan H, Luu MDA, Gadjdjoe PS, Dolga AM, Schmidt M. Unravelling the signaling power of pollutants. Trends Pharmacol Sci 2023; 44:917-933. [PMID: 37783643 DOI: 10.1016/j.tips.2023.09.002] [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: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
Exposure to environmental pollutants contributes to diverse pathologies, including pulmonary disease, lower respiratory infections, cancer, and stroke. Pollutants' entry can occur through inhalation, traversing endothelial and epithelial barriers, and crossing the blood-brain barrier, leading to a wide distribution throughout the human body via systemic circulation. Pollutants cause cellular damage by multiple mechanisms encompassing oxidative stress, mitochondrial dysfunction, (neuro)inflammation, and protein instability/proteotoxicity. Sensing pollutants has added a new dimension to disease progression and drug failure. Understanding the molecular pathways and potential receptor binding/signaling that underpin 'sensing' could contribute to ways to combat the detrimental effects of pollutants. We highlight key points of pollutant signaling, crosstalk with receptors acting as drug targets for chronic diseases, and discuss the potential for future therapeutics.
Collapse
Affiliation(s)
- Ana L Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Phoeja S Gadjdjoe
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
31
|
Wan R, Wang L, Zhu M, Li W, Duan Y, Yu G. Cellular Senescence: A Troy Horse in Pulmonary Fibrosis. Int J Mol Sci 2023; 24:16410. [PMID: 38003600 PMCID: PMC10671822 DOI: 10.3390/ijms242216410] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disease characterized by myofibroblast abnormal activation and extracellular matrix deposition. However, the pathogenesis of PF remains unclear, and treatment options are limited. Epidemiological studies have shown that the average age of PF patients is estimated to be over 65 years, and the incidence of the disease increases with age. Therefore, PF is considered an age-related disease. A preliminary study on PF patients demonstrated that the combination therapy of the anti-senescence drugs dasatinib and quercetin improved physical functional indicators. Given the global aging population and the role of cellular senescence in tissue and organ aging, understanding the impact of cellular senescence on PF is of growing interest. This article systematically summarizes the causes and signaling pathways of cellular senescence in PF. It also objectively analyzes the impact of senescence in AECs and fibroblasts on PF development. Furthermore, potential intervention methods targeting cellular senescence in PF treatment are discussed. This review not only provides a strong theoretical foundation for understanding and manipulating cellular senescence, developing new therapies to improve age-related diseases, and extending a healthy lifespan but also offers hope for reversing the toxicity caused by the massive accumulation of senescence cells in humans.
Collapse
Affiliation(s)
- Ruyan Wan
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Lan Wang
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Miaomiao Zhu
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Wenwen Li
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Yudi Duan
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Guoying Yu
- Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; (R.W.); (L.W.); (M.Z.); (W.L.); (Y.D.)
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
32
|
Pei J, Tian X, Yu C, Luo J, Hong Y, Zhang J, Wen S, Hua Y, Wei G. Transcriptome-based exploration of potential molecular targets and mechanisms of selenomethionine in alleviating renal ischemia-reperfusion injury. Clin Sci (Lond) 2023; 137:1477-1498. [PMID: 37706564 DOI: 10.1042/cs20230818] [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: 07/27/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/15/2023]
Abstract
Renal ischemia-reperfusion injuries (IRIs) are one of the leading causes of acute kidney injuries (AKIs). Selenium, as an essential trace element, is able to antioxidant stress and reduces inflammatory responses. The regulation mechanism of selenomethionine, one of the major forms of selenium intake by humans, is not yet clear in renal IRIs. Therefore, we aimed to explore the key targets and related mechanisms of selenomethionine regulation in renal IRIs and provide new ideas for the treatment of selenomethionine with renal IRIs. We used transcriptome sequencing data from public databases as well as animal experiments to explore the key target genes and related mechanisms regulated by selenomethionine in renal IRI. We found that selenomethionine can effectively alleviate renal IRI by a mechanism that may be achieved by inhibiting the MAPK signaling pathway. Meanwhile, we also found that the key target of selenomethionine regulation in renal IRI might be selenoprotein GPX3 based on the PPI protein interaction network and machine learning. Through a comprehensive analysis of bioinformatic techniques and animal experiments, we found that Gpx3 might serve as a key gene for the regulation of selenomethionine in renal IRIs. Selenomethionine may exert a protective effect against renal IRI by up-regulating GPX3, inhibiting the MAPK signaling pathway, increased production of antioxidants, decreasing inflammation levels, mitigation of apoptosis in renal tubular epithelial cells, this reduces renal histopathological damage and protects renal function. Providing a theoretical basis for the mechanism of selenomethionine actions in renal IRIs.
Collapse
Affiliation(s)
- Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Xiaomao Tian
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Sheng Wen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yi Hua
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| |
Collapse
|
33
|
Bang E, Hwangbo H, Kim MY, Ji SY, Kim DH, Shim JH, Moon SK, Kim GY, Cheong J, Choi YH. Urban aerosol particulate matter promotes mitochondrial oxidative stress-induced cellular senescence in human retinal pigment epithelial ARPE-19 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104211. [PMID: 37423393 DOI: 10.1016/j.etap.2023.104211] [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: 04/05/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Environmental exposure to urban particulate matter (UPM) is a serious health concern worldwide. Although several studies have linked UPM to ocular diseases, no study has reported effects of UPM exposure on senescence in retinal cells. Therefore, this study aimed to investigate the effects of UPM on senescence and regulatory signaling in human retinal pigment epithelial ARPE-19 cells. Our study demonstrated that UPM significantly promoted senescence, with increased senescence-associated β-galactosidase activity. Moreover, both mRNA and protein levels of senescence markers (p16 and p21) and the senescence-associated secretory phenotype, including IL-1β, matrix metalloproteinase-1, and -3 were upregulated. Notably, UPM increased mitochondrial reactive oxygen species-dependent nuclear factor-kappa B (NF-κB) activation during senescence. In contrast, use of NF-κB inhibitor Bay 11-7082 reduced the level of senescence markers. Taken together, our results provide the first in vitro preliminary evidence that UPM induces senescence by promoting mitochondrial oxidative stress-mediated NF-κB activation in ARPE-19 cells.
Collapse
Affiliation(s)
- EunJin Bang
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Molecular Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Ansung 17546, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Jaehun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| |
Collapse
|
34
|
Li N, Xiong R, Li G, Wang B, Geng Q. PM2.5 contributed to pulmonary epithelial senescence and ferroptosis by regulating USP3-SIRT3-P53 axis. Free Radic Biol Med 2023; 205:291-304. [PMID: 37348684 DOI: 10.1016/j.freeradbiomed.2023.06.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Pulmonary epithelial cells act as the first line of defense against various air pollutant particles. Previous studies have reported that particulate matter 2.5 (PM2.5) could trigger pulmonary inflammation and fibrosis by inducing pulmonary epithelial senescence and ferroptosis. Sirtuin 3 (SIRT3) is one of critical the mitochondrial NAD+-dependent deacetylases, exerting antioxidant and anti-aging effects in multiple diseases. The present study aimed to explore the role of SIRT3 in PM2.5-induced lung injury as well as possible mechanisms. The role of SIRT3 in PM2.5-induced lung injury was investigated by SIRT3 genetic depletion, adenovirus-mediated overexpression in type II alveolar epithelial (AT2) cells, and pharmacological activation by melatonin. The protein level and activity of SIRT3 in lung tissues and AT2 cells were significantly downregulated after PM2.5 stimulation. SIRT3 deficiency in AT2 cells aggravated inflammatory response and collagen deposition in PM2.5-treated lung tissues. RNA-sequence and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the differentially expressed genes (DEGs) between SIRT3 flox and SIRT3 CKO mice were mainly enriched in ferroptosis and cellular longevity. Western blot further showed that SIRT3 deficiency in AT2 cells significantly upregulated the proteins associated with ferroptosis and cell senescence in PM2.5-treated lung tissues. In vitro experiments also showed that SIRT3 overexpression could decrease the levels of ferroptosis and cell senescence in PM2.5-treated AT2 cells. In addition, we found that PM2.5 could increase the acetylation of P53 via triggering DNA damage in AT2 cells. And SIRT3 could deacetylate P53 at lysines 320 (K320), thus reducing its transcriptional activity. PM2.5 decreased the protein level of SIRT3 by inducing proteasome pathway through downregulating USP3. Finally, we found that SIRT3 agonist, melatonin treatment could alleviate PM2.5-induced senescence and ferroptosis in mice. In conclusion, targeting USP3-SIRT3-P53 axis may be a novel therapeutic strategy against PM2.5-induced pulmonary inflammation and fibrosis by decreasing pulmonary epithelial senescence and ferroptosis.
Collapse
Affiliation(s)
- Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|