1
|
Zhang K, Honda A, Sagawa T, Miyasaka N, Qiu B, Ishikawa R, Okuda T, Kameda T, Sadakane K, Ichinose T, Takano H. Complement system is activated in acute inflammatory response to environmental particulates in the lungs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118259. [PMID: 40315749 DOI: 10.1016/j.ecoenv.2025.118259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 04/01/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Short-term exposure (typically ranging from a few hours to 7 days) to particulate matter (PM) elicits acute inflammatory responses with significant activation of complement component C5. However, the relationship between PM-induced complement system activation, acute inflammation, and the contributing factors remains unclear. In this study, we aimed to investigate the intensity of acute inflammatory responses, as well as the activation levels of complement C5 and its related products, C5aR1 and C5b-9, following exposure to different types of PM. Acute inflammatory responses and complement activation were assessed in mice intratracheally administered four types of PM: titanium dioxide (TiO₂), diesel exhaust particles (DEP), Asian sand dust (ASD), and ambient PM with an aerodynamic diameter ≤ 2.5 μm (PM2.5). Complement system and inflammatory markers were evaluated by analyzing increased C5 protein levels, C5b-9 deposition, and C5aR1 expression. Additionally, dark-field microscopy and Raman microscopy were used to detect PM components adjacent to infiltrating neutrophils, and elemental composition was quantified using ICP-MS and EDXRF. ASD, DEP, and PM2.5 exposure significantly increased C5b-9 deposition in lung tissues. All PM-exposed groups exhibited substantial upregulation of C5aR1 expression, primarily in neutrophils. Raman spectroscopic analysis revealed Si, K, Mg, Al, and Fe adjacent to infiltrating neutrophils in ASD-exposed lungs. Furthermore, elemental analysis identified Si, Mg, Al, and K as the most potent contributors to complement activation and inflammatory responses. Of the four types of PM, ASD induced the most severe acute inflammatory response and complement system activation. Therefore, ASD-induced complement system activation, driven at least partly by its mineral components, may play a critical role in neutrophil activation and acute pulmonary inflammation. These findings highlight the differential impact of PM types on complement system activation and underscore the importance of PM composition in the evaluation of air pollution-related health risks, particularly acute pulmonary inflammation.
Collapse
Affiliation(s)
- Kerui Zhang
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Akiko Honda
- Graduate School of Engineering, Kyoto University, Kyoto, Japan.
| | - Tomoya Sagawa
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsuko Miyasaka
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Binyang Qiu
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Raga Ishikawa
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Tomoaki Okuda
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Takayuki Kameda
- Graduate School of Energy Science, Kyoto University, Kyoto, Japan
| | - Kaori Sadakane
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, Japan
| | - Takamichi Ichinose
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, Japan
| | - Hirohisa Takano
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Institute for International Academic Research, Kyoto University of Advanced Science, Kyoto, Japan; Research Institute for Coexistence and Health Science, Kyoto University of Advanced Science, Kyoto, Japan.
| |
Collapse
|
2
|
Saheb Sharif-Askari N, Mdkhana B, Hafezi S, Elamin OF, Eladham MW, Al-Sheakly BKS, Halwani H, Saheb Sharif-Askari F, Halwani R. Calprotectin inhibition attenuates silica-induced lung fibrosis. Inflammopharmacology 2025:10.1007/s10787-025-01771-5. [PMID: 40381145 DOI: 10.1007/s10787-025-01771-5] [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: 01/03/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Respirable silica exposure adversely affects lung tissue immunopathology, triggering oxidative bursts in macrophages and neutrophils, releasing Damage-associated molecular patterns (DAMPs), including calprotectin proteins, S100A8, and S100A9. Calprotectin constitutes up to 45% of these innate immune cells, and serum levels of these alarmins correlate with inflammation, fibrosis, remodelling, and drug response in chronic diseases, including inflammatory bowel disease, asthma, and cystic fibrosis. The consequence of releasing calprotectin protein could trigger the pro-fibrotic effect of silicosis. This study aimed to investigate the role of calprotectin (S100A8/S100A9) as a pro-inflammatory and pro-fibrotic mediator in silica-induced lung fibrosis and evaluated the therapeutic potential of the calprotectin inhibitor, paquinimod. Using a mouse model of silicosis, silica exposure significantly elevated calprotectin expression, lung inflammation, and fibrosis, as evidenced by increased levels of epithelial-to-mesenchymal transition (EMT) markers, collagen deposition, and matrix metalloproteinases (MMPs). In vitro, stimulation of human bronchial fibroblasts with S100A8/S100A9 upregulated fibrotic markers (COL1A1 and α-SMA), which were reduced by inhibitors of TLR4 and RAGE receptors, as well as by paquinimod. Treatment with paquinimod effectively reduced these pathological changes, normalized calprotectin levels, decreased fibrosis scores, and attenuated NF-κB activation. These findings highlighted calprotectin's pivotal role in silica-induced lung fibrosis and inflammation, suggesting that its inhibition could be a promising therapeutic approach for silicosis and other fibro-inflammatory lung diseases. Further research is warranted to explore the precise mechanisms linking calprotectin to lung fibrosis and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ola Faisal Elamin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mariam Wed Eladham
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Hala Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
3
|
Chang CK, Wu ZS, Niu GH, Chou YY, Tang SH, Zhang MM, Sung CS, Tung HT, Tsou LK, Tang CC, Sung PJ, Lo YH, Wen ZH. Marine-derived STING inhibitors, excavatolide B promote wound repair in full-thickness-incision rats. Int Immunopharmacol 2025; 155:114593. [PMID: 40209311 DOI: 10.1016/j.intimp.2025.114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025]
Abstract
The process of wound healing encompasses both inflammatory and proliferative stages. Excessive inflammation is known to impede the healing of chronic wounds. Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway causes inflammation during cellular stress and tissue damage. Herein, we examined the anti-inflammatory effects of the marine-derived STING antagonist excavatolide B (EXCB) and its derivatives, EXCB-61 and EXCB-79, in full-thickness-incision rats. Wound area measurements, histopathological observations, and immunohistochemical analyses were performed to evaluate the roles of these compounds in wound healing. These three compounds were found to have low toxicity, with EXCB promoting Hs68 human dermal fibroblast migration and proliferation. EXCB and EXCB61 treatments, but not EXCB79, reduced the wound area. The histopathological results showed a significant decrease in immune cell infiltration and mast cell accumulation in all compound-treated groups. Immunohistochemical analysis revealed that EXCB and its derivatives reduced cGAS-STING pathway factors such as STING, phosphorylated TANK-binding kinase 1, nuclear factor kappa-light-chain-enhancer of activated B cells, and M1 macrophages while increasing the expression of angiogenic factors vascular endothelial growth factor and CD31, as well as M2 macrophages and collagen I/III deposition. We conclude that marine-derived STING antagonists can attenuate inflammatory responses by inhibiting the cGAS-STING pathway and promoting angiogenesis, thereby aiding wound healing.
Collapse
Affiliation(s)
- Chun-Kai Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Plastic & Reconstructive Surgery, Zouying Armed Forces General Hospital, No. 553, Junxiao Rd., Zuoying Dist., Kaohsiung City 81342, Taiwan
| | - Zong-Sheng Wu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Yu-Yu Chou
- Department of Plastic & Reconstructive Surgery, Zouying Armed Forces General Hospital, No. 553, Junxiao Rd., Zuoying Dist., Kaohsiung City 81342, Taiwan
| | - Shih-Hsuan Tang
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan; School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsiang-Ting Tung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pingtung University, 91201, Taiwan
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan; Department of Nursing, Meiho University, Pingtung County, 91200, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
4
|
Wang C, Lin X, Guan S, Wu Q, Liang S. Dihydroartemisinin Attenuates Radiation-Induced Lung Injury by Inhibiting the cGAS/STING/NF-κB Signaling Pathway. Drug Dev Res 2025; 86:e70090. [PMID: 40285509 PMCID: PMC12032573 DOI: 10.1002/ddr.70090] [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: 03/26/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Dihydroartemisinin (DHA) is a derivative of artemisinin, which affects inflammation, oxidative stress, and immune regulation. However, the mechanism underlying its effects remains largely unknown. This study aims to explore the mechanism by which DHA affects radiation-induced lung injury (RILI), providing new insights for lung radiotherapy. To elucidate its mechanism of action, C57BL/6 J mice were irradiated with 15 Gy whole chest. RILI was evaluated by qRT-PCR, ELISA, histology, Western blot analysis, immunohistochemistry, and RILI signaling cascade studies. In addition, small interfering RNAs were employed to knockdown cGAS proteins in the cGAS-STING signaling pathway in the human bronchial epithelium cell line (BEAS-2B). Both In Vivo and Vitro experiments were conducted to investigate the specific mechanism by which DHA alleviated RILI. We observed the activation of the cGAS-STING pathway, along with the phosphorylation of the downstream target NF-κB and an increase in inflammatory factor levels in the mouse model following radiation exposure. In the cell model, irradiation also triggered the activation of the cGAS-STING signaling pathway and its downstream targets, leading to elevated levels of inflammatory factors. Notably, knocking down the cGAS using small interfering RNA in the BEAS-2B cells significantly alleviated RILI in the cell model. Our study elucidated the mechanism of DHA reducing RILI through the cGAS/STING/NF-κB signaling pathway, and revealed that the GAS/STING/NF-κB axis may be a potential therapeutic target for RILI.
Collapse
Affiliation(s)
- Cailan Wang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Xinyi Lin
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shichun Guan
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Qiaoyuan Wu
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shixiong Liang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| |
Collapse
|
5
|
Triaille C, Terrier B, Hadchouel A, Haddad E, Vaglio A, Frémond ML. The emerging concept of ANCA-associated vasculitis related to inborn errors of immunity. Autoimmun Rev 2025; 24:103824. [PMID: 40294837 DOI: 10.1016/j.autrev.2025.103824] [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: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
ANCA-associated vasculitis (AAV) is a group of rare small vessels vasculitis that preferentially affect the kidneys, lungs and upper airways. Although the detailed pathophysiology remains unclear, genetic background has been shown to play a role in sporadic forms of AAV. The discovery of these susceptibility genes (and associated biological pathways) involved in AAV have shaped the current understanding of AAV pathophysiology. In addition to common genetic polymorphisms, specific rare inborn errors of immunity (IEI) have been described with a high frequency of ANCA (antineutrophil cytoplasmic antibodies) positivity and vasculitis features in young individuals (in addition to other manifestations). A systematic literature search revealed that patients with pathogenic variants in COPA, STING1, DNASE1L3, and PIK3CD are at increased risk of developing ANCA and AAV features, including alveolar hemorrhage, interstitial lung disease, pauciimmune glomerulonephritis, and upper airways involvement (septum perforation, saddle-nose deformity, chronic nasal/sinuses ulceration). Some of these IEI may also present with a mixed phenotype and/or auto-antibodies profile associating features of AAV and other autoimmune diseases (in particular systemic lupus erythematosus). Notably, a proportion of reports and series lack serological (ANCA specificity and titers) and/or histopathological data, making challenging to assess the likelihood for ANCA pathogenicity in some patients with IEI (as opposed to unspecific signs of biologic autoimmunity). This point is nonetheless essential to make appropriate therapeutic decisions. In addition, since most of the genes mentioned above are involved in the type 1 interferon signaling, the role of this pathway in AAV etiopathogenesis deserves further investigation. In this review, we will describe these IEI, their overlap with sporadic AAV, and their evocative features. Next, we will discuss how these monogenic conditions might inform our general understanding of AAV pathophysiology. We also propose some directions for future research in order to better define the link between ANCA and IEI. Finally, we will consider how making the diagnosis of an IEI in a patient with AAV features might impact individual management.
Collapse
Affiliation(s)
- Clément Triaille
- Pôle de Pathologies Rhumatismales Systémiques et Inflammatoires (RUMA), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium; Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.
| | - Benjamin Terrier
- Department of Internal Medicine, Hospital Cochin, AP-HP, Paris, France; Université Paris Cité, Paris, France
| | - Alice Hadchouel
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France; INSERM U1151, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | - Elie Haddad
- Department of Pediatrics, Department of Microbiology, Immunology and Infectious Diseases, CHU Sainte-Justine Azrieli Research Center, Université de Montréal, Montreal, Quebec, Canada
| | - Augusto Vaglio
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Firenze, Italy; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Marie-Louise Frémond
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France; Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| |
Collapse
|
6
|
Giordano L, Ware SA, Lagranha CJ, Kaufman BA. Mitochondrial DNA signals driving immune responses: Why, How, Where? Cell Commun Signal 2025; 23:192. [PMID: 40264103 PMCID: PMC12012978 DOI: 10.1186/s12964-025-02042-0] [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: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
There has been a recent expansion in our understanding of DNA-sensing mechanisms. Mitochondrial dysfunction, oxidative and proteostatic stresses, instability and impaired disposal of nucleoids cause the release of mitochondrial DNA (mtDNA) from the mitochondria in several human diseases, as well as in cell culture and animal models. Mitochondrial DNA mislocalized to the cytosol and/or the extracellular compartments can trigger innate immune and inflammation responses by binding DNA-sensing receptors (DSRs). Here, we define the features that make mtDNA highly immunogenic and the mechanisms of its release from the mitochondria into the cytosol and the extracellular compartments. We describe the major DSRs that bind mtDNA such as cyclic guanosine-monophosphate-adenosine-monophosphate synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), NOD-, LRR-, and PYD- domain-containing protein 3 receptor (NLRP3), absent in melanoma 2 (AIM2) and toll-like receptor 9 (TLR9), and their downstream signaling cascades. We summarize the key findings, novelties, and gaps of mislocalized mtDNA as a driving signal of immune responses in vascular, metabolic, kidney, lung, and neurodegenerative diseases, as well as viral and bacterial infections. Finally, we define common strategies to induce or inhibit mtDNA release and propose challenges to advance the field.
Collapse
Affiliation(s)
- Luca Giordano
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, Giessen, Germany.
| | - Sarah A Ware
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia J Lagranha
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Jiao B, An C, Du H, Tran M, Yang D, Zhao Y, Wang P, Hu Z, Zhou D, Wang Y. Genetic deficiency or pharmacological inhibition of cGAS-STING signalling suppresses kidney inflammation and fibrosis. Br J Pharmacol 2025; 182:1741-1762. [PMID: 39833988 DOI: 10.1111/bph.17412] [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: 10/24/2023] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Chronic kidney disease (CKD) is characterised by inflammation, which can lead to tubular atrophy and fibrosis. The molecular mechanisms are not well understood. In this study, we investigated the functional role of the cyclic GMP-AMP synthase (cGAS)- stimulator of interferon genes (STING) signalling in renal inflammation and fibrosis. EXPERIMENTAL APPROACH Mice with global cGAS deficiency or global or myeloid cell-specific STING deficiency or wild-type mice treated with RU.521, a selective cGAS inhibitor, were used to examine the role of cGAS-STING signalling in renal inflammation and fibrosis in a preclinical model of obstructive nephropathy in vivo. Bone marrow-derived macrophages were used to determine whether tubular epithelial cell-derived DNA can activate cGAS-STING signalling in vitro. KEY RESULTS Following obstructive injury, cGAS-STING signalling was activated in the kidneys during the development of renal fibrosis. Mice with deficiency of cGAS or STING exhibited significantly less macrophage proinflammatory activation, myofibroblast formation, total collagen deposition, and extracellular matrix (ECM) protein production in the kidneys following obstructive injury. Pharmacological inhibition of cGAS with RU.521 reduced macrophage proinflammatory activation, suppressed myofibroblast formation, and attenuated kidney fibrosis following obstructive injury. Mechanistically, cGAS-STING signalling in macrophages is activated by double-stranded DNA released from damaged tubular epithelial cells, which induces inflammatory responses. CONCLUSIONS AND IMPLICATIONS Our study identifies the cGAS-STING signalling pathway as a critical regulator of macrophage proinflammatory activation during the development of renal fibrosis. Therefore, inhibition of cGAS-STING signalling may represent a novel therapeutic strategy for CKD.
Collapse
Affiliation(s)
- Baihai Jiao
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Changlong An
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Hao Du
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Melanie Tran
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Duomeng Yang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California, USA
| | - Penghua Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Zhaoyong Hu
- Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA
- Renal Section, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
8
|
Wu Q, Zhang Q, Jin C, Liu X, Yu H. CircHOMER1 promotes silica-induced pulmonary fibrosis by binding to HuR and stabilizing NOX4 mRNA. Cell Signal 2025; 128:111638. [PMID: 39909178 DOI: 10.1016/j.cellsig.2025.111638] [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: 11/21/2024] [Revised: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Silicosis, one of the serious occupational diseases, is mainly manifested by pulmonary fibrosis induced by long-term exposure to silica particles in workplace. Evidence demonstrates that circular RNAs (circRNAs) are interesting regulators of pulmonary fibrosis process. So, further elucidation of the role of circRNAs may provide a new perspective into mechanisms driving pulmonary fibrosis and silicosis. METHODS The characteristics of circRNA homer scaffold protein 1 (hsa_circ_0006916, circHOMER1) was assessed using Actinomycin D, RNase R, and nucleoplasmic separation assay. The histopathological examination and Enzyme-linked immunosorbent assay (ELISA) were used to confirm circHOMER1 function in mouse lung tissues under silica particle exposure. The expression of circHOMER1, human antigen R (HuR) and NADPH oxidase 4 (NOX4) was identified by western blot or RT-qPCR assay. The RNA immunoprecipitation (RIP) assay and plasmid co-transfection were used to analyze the interaction between circHOMER1, HuR and NOX4. RESULTS We confirmed an upregulated circHOMER1 in silicosis fibrosis. Functional assays showed that the knockdown of circHOMER1 suppressed the viability of fibroblasts and the production of fibrotic molecules and alleviated the histology fibrotic changes in lung tissues from mouse exposed to silica particles. Mechanistically, we found that circHOMER1 directly bound to HuR and promoted its protein expression in fibroblasts. And, circHOMER1 further regulated HuR/NOX4 signaling axis through HuR to stabilize NOX4 mRNA, which enhanced the production of reactive oxygen species (ROS), thereby promoting the silicosis fibrosis process. CONCLUSION This study revealed the role of circHOMER1 in silica-induced pulmonary fibrosis, suggesting that the inhibition of circHOMER1 may be a potential therapeutic approach to relieve the pathological process of silicosis.
Collapse
Affiliation(s)
- Qiuyun Wu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou 221004, China.
| | - Qianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Chunmeng Jin
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Xue Liu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Hongmin Yu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
9
|
Lam M, Barry KT, Hodges CJ, Harpur CM, Ong JDH, Rosli S, West AC, Dousha L, Hertzog PJ, Mansell A, Tate MD. NLRP3 deficiency abrogates silica-induced neutrophil infiltration, pulmonary damage and fibrosis. Respir Res 2025; 26:109. [PMID: 40119408 PMCID: PMC11929224 DOI: 10.1186/s12931-025-03192-y] [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: 12/30/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Silicosis is a progressive and often fatal occupational lung disease. The NLRP3 inflammasome is an innate immune sensor that is activated by silica. Accumulating evidence has implicated a role for NLRP3 in silicosis pathogenesis. In this study, we mechanistically elucidated the contribution of NLRP3 to silica-induced pulmonary disease. METHODS The in vivo role of NLRP3 was investigated following intranasal delivery of 2 mg of silica or diluent alone to wildtype, NLRP3 reporter, and NLRP3-deficient mice. Protein expression, inflammation, and histopathology were analyzed in the lung. RESULTS Intranasal administration of silica recapitulated the key pathological features of human silicosis, including nonresolving inflammation, the formation of silicotic nodules, and diffuse lung fibrosis. A reporter mouse placed under the native NLRP3 promoter revealed silica rapidly upregulated NLRP3 expression throughout the lung. NLRP3-deficient mice displayed marked early reductions in silica-induced IL-1β and IL-18 levels in the airways. Additionally, NLRP3 deficiency impaired the rapid infiltration of conventional Siglec-F- and fibrotic Siglec-F+ neutrophils, which correlated with reduced levels of neutrophil elastase. Deficiency in acute NLRP3-mediated inflammation correlated with significantly reduced pulmonary transforming growth factor beta and alpha smooth muscle actin expression, tissue damage, and fibrosis in the chronic phase of disease progression. Importantly, this included reduced silicotic nodule size and cellularity. CONCLUSIONS These findings highlight a major detrimental role for the NLRP3 inflammasome in driving silica-induced pulmonary neutrophil infiltration, TGFβ-mediated myofibroblast activation, tissue damage, and fibrosis.
Collapse
Affiliation(s)
- Maggie Lam
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Kristian T Barry
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher J Hodges
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher M Harpur
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - James D H Ong
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Sarah Rosli
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Alison C West
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Lovisa Dousha
- Monash Lung, Sleep, Allergy, and Immunology, Monash Medical Centre, Clayton, Melbourne, VIC, 3168, Australia
- Department of Medicine, Monash University, Clayton, Melbourne, VIC, 3168, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia.
- Department of Molecular Translational Sciences, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
10
|
Li X, Tan J, Xiong W, Feng Y, Zhang Z. Silica-induced ferroptosis activates retinoic acid signaling in dendritic cells to drive inflammation and fibrosis in silicosis. Int Immunopharmacol 2025; 149:114244. [PMID: 39938311 DOI: 10.1016/j.intimp.2025.114244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Silicosis, a chronic lung disease caused by inhalation of silica (SiO2) particles from environmental contamination or industrial exposure, is characterized by persistent inflammation and fibrosis. This study elucidates a novel mechanism where SiO2 exposure triggers ferroptosis, a lipid peroxidation-dependent form of cell death, in dendritic cells (DCs), thereby activating retinoic acid (RA) signaling. The RA response amplifies inflammatory pathways, including cGAS-STING-IFN-I and IL-1β signaling, exacerbating lung inflammation and fibrosis. The study uses murine models to demonstrate that ferroptosis inhibitors, such as ferrostatin-1, mitigate SiO2-induced inflammation and collagen deposition. Furthermore, systemic administration of the synthetic retinoid AM80 reduces pulmonary damage by modulating immune cell distribution and promoting lymphocyte homing. These findings reveal the interplay between ferroptosis and RA signaling as a pivotal driver of silicosis pathology and suggest therapeutic avenues targeting ferroptosis and RA modulation for disease management.
Collapse
Affiliation(s)
- Xingjie Li
- Department of Clinical Laboratory, Guangyuan Central Hospital, Guangyuan, Sichuan 628000, China
| | - Jinzhuo Tan
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Clinical Laboratory, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Wenyan Xiong
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yingna Feng
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zongde Zhang
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
11
|
Feng Y, Su Q, Zhu J, Cui X, Guo J, Yang J, Zhang S. cGAS-STING mediates neutrophil extracellular traps-induced EMT in myositis-associated interstitial lung disease: STING as a potential therapeutic target. Int Immunopharmacol 2025; 149:114144. [PMID: 39904030 DOI: 10.1016/j.intimp.2025.114144] [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: 10/31/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) play crucial roles in idiopathic inflammatory myositis-associated interstitial lung disease (IIM-ILD) pathogenesis. The involvement and mechanism of alveolar epithelial cells in the pathogenesis of IIM-ILD remain unclear. This study aimed to clarify the hypothesis that NETs promote alveolar epithelial-mesenchymal transition (EMT), which contributes to IIM-ILD. METHODS Lung biopsy puncture tissue from three IIM-ILD patients was used for pathological analysis. An experimental mouse model of autoimmune myositis with ILD (EAM-ILD) was used for mechanism validation in vivo. A549 cells were treated with NETs and assessed using Western blotting, immunofluorescence, and RNA sequencing techniques. STING inhibitors were employed to assess the efficacy of stimulator of interferon gene (STING) as a therapeutic target for EAM-ILD. RESULT There was a marked EMT and the activation of cGAS-STING pathway found in the lung samples of IIM-ILD patients (N = 3) and in A549 cells in vitro (P < 0.05). RNA sequencing indicates that NETs induce an upregulation of inflammation and fibrosis-related pathways in A549 cells, with high expression of the STING-related pathway. The STING inhibitor can prevent EMT in alveolar epithelial cells both in vivo and in vitro, and reduce the inflammatory response in the lung tissue of the EAM-ILD mouse (P < 0.05). CONCLUSION These in vitro and ex vivo experiments demonstrate that NETs promote EAM-ILD by inducing EMT in alveolar epithelial cells and that the cGAS-STING signaling pathway is one of the potential mechanisms of action. Targeting STING is a potential therapeutic strategy for treating IIM-ILD.
Collapse
Affiliation(s)
- Yingyue Feng
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Qiyan Su
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China.
| | - Xi Cui
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Jin Guo
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Jumei Yang
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China.
| | - Sigong Zhang
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China; Department of Rheumatology and Immunology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
12
|
Seok JK, Jee JI, Jeon M, Kim D, Chung KH, Kim HR, Baek YW, Kang H, Lim J, Bae ON, Lee JY. cGAS/STING pathway modulation in polyhexamethyleneguanidine phosphate-induced immune dysregulation and pulmonary fibrosis using human monocytic cells (THP-1) and male C57BL/6 mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:162-174. [PMID: 39604835 DOI: 10.1080/15287394.2024.2432020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Polyhexamethyleneguanidine phosphate (PHMG), a widely used antimicrobial agent, has been implicated in humidifier disinfectant-associated lung injuries (HDLI). PHMG exposure suppressed interferon regulatory factor 3 (IRF3) activation and interferon-β (IFN-β) expression induced by a cGAS agonist or a STING agonist in human monocytic cells (THP-1), which are known to transition to alveolar macrophages during pulmonary fibrosis development. However, the mechanisms underlying PHMG-induced pulmonary toxicity in lung remain unclear. Thus, it was of interest to investigate the effects of PHMG on the innate immune system in male C57BL/6 mouse, focusing on the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway and potential role in pulmonary fibrosis. Intratracheal administration of PHMG (1 or 2 mg/kg) in mice resulted in lung fibrosis, as evidenced by H&E staining with Szapiel scoring, Masson's trichrome staining with Ashcroft scoring, and increased mRNA levels of TGF-β and collagen type I. Interestingly, lower dose of PHMG enhanced IFN-β production in the lungs, whereas higher dose decreased IFN-β levels, indicating a biphasic effect that initially promotes inflammation but ultimately impairs host defense mechanisms, leading to pulmonary fibrosis. Our findings demonstrate the critical role of the cGAS/STING pathway in PHMG-induced mouse lung injury and suggest that targeting this pathway might serve as a potential therapeutic strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Jin Kyung Seok
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jung In Jee
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Minwoo Jeon
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, Republic of Korea
| | - Kyu Hyuck Chung
- College of Pharmacy, Kyungsung University, Busan, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ha Ryong Kim
- College of Pharmacy, Korea University, Sejong, Republic of Korea
| | - Yong-Wook Baek
- Humidifier Disinfectant Health Center, Environmental Health Research Department, National Institute of Environmental Research, Incheon, Republic of Korea
| | - HanGoo Kang
- Humidifier Disinfectant Health Center, Environmental Health Research Department, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Jungyun Lim
- Humidifier Disinfectant Health Center, Environmental Health Research Department, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| |
Collapse
|
13
|
Kim EH, Wahl K, Guelfi E, Lee D. Engineering the physical characteristics of biomaterials for innate immune-mediated cancer immunotherapy. J Control Release 2025; 378:814-830. [PMID: 39719214 DOI: 10.1016/j.jconrel.2024.12.046] [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: 07/04/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
It has recently been recognized that the physical characteristics of biomaterials - such as size, structure, shape, charge, mechanical strength, hydrophobicity, and multivalency - regulate immunological functions in innate immune cells. In immuno-oncology applications, biomaterials are engineered with distinct physical properties to achieve desired innate immune responses. In this review, we discuss how physical characteristics influence effector functions and innate immune signaling pathways in distinct innate immune cell subtypes. We highlight how physical properties of biomaterials impact phagocytosis regulation, biodistribution, and innate immune cell targeting. We outline the recent advances in physical engineering of biomaterials that directly or indirectly induce desired innate immune responses for cancer immunotherapy. Lastly, we discuss the challenges in current biomaterial approaches that need to be addressed to improve clinical applicability.
Collapse
Affiliation(s)
- Eun-Hye Kim
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Katelyn Wahl
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Erica Guelfi
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - DaeYong Lee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
14
|
Wang Y, Zhang X, Wang W, Zhang Y, Fleishman JS, Wang H. cGAS-STING targeting offers therapy choice in lung diseases. Biol Direct 2025; 20:20. [PMID: 39920718 PMCID: PMC11806777 DOI: 10.1186/s13062-025-00611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Cyclic GMP/AMP (cGAMP) synthase (cGAS), along with the endoplasmic reticulum (ER)-associated stimulator of interferon genes (STING), are crucial elements of the type 1 interferon response. cGAS senses microbial DNA and self-DNA, labeling cGAS-STING as a crucial mechanism in autoimmunity, sterile inflammatory responses, and cellular senescence. However, chronic and aberrant activation of the cGAS-STING axis results in inflammatory and autoimmune diseases. cGAS-STING has emerged as a vital mechanism driving inflammation-related diseases, including lung diseases. Insights into the biology of the cGAS-STING pathway have enabled the discovery of small-molecule agents which have the potential to inhibit the cGAS-STING axis in lung diseases. In this review, we first outline the principal components of the cGAS-STING signaling cascade. Then, we discuss recent research that highlights general mechanisms by which cGAS-STING contributes to lung diseases. Then, we focus on summarizing a list of bioactive small-molecule compounds which inhibit the cGAS-STING pathway, reviewing their potential mechanisms.These review highlights a novel groundbreaking therapeutic possibilities through targeting cGAS-STING in lung diseases.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Xuan Zhang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Yi Zhang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
15
|
Yang X, Qi Y, Wang S. DKN-01 Suppresses Gastric Cancer Progression Through Activating cGAS-STING Pathway to Block Macrophage M2 Polarization. Appl Biochem Biotechnol 2025; 197:1025-1038. [PMID: 39352454 DOI: 10.1007/s12010-024-05073-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 02/13/2025]
Abstract
Dickkopf-1 (DKK1) is a secretory antagonist that can bind with the Wnt coreceptor to desensitize cells to canonical Wnt ligands. DKN-01 is a specific antibody targeting secreted DKK1, which has been investigated as a monotherapy or combination therapy for various malignant tumors, including gastric cancer (GC). Tumor-associated macrophages (TAMs) with high plasticity usually present M2 phenotype, which can promote tumor progression. The aim of this study was to investigate the effect of DKN-01 on macrophage polarization in GC and the underlying molecular mechanism. To ascertain the effect of DKN-01 on GC tumor growth, we established a tumor-bearing mouse model and found that DKN-01 treatment suppressed tumor growth efficiently. Through RNA-seq and pathway enrichment analysis, we identified that the differentially expressed genes after DKN-01 treatment are associated with tumor immune-related pathways. Macrophage polarization was assessed using immunohistochemistry and quantitative real-time polymerase chain reaction. DKN-01 and knockdown of DKK1 promoted M1 polarization and inhibited M2 polarization of macrophages, while DKK1 overexpression got the opposite results. Moreover, DKN-01 activated the cGAS/STING pathway, while the inactivation of cGAS-STING pathway using RU.521 reversed the inhibition of tumor growth in vivo and macrophage M2 polarization caused by DKN-01. This study reveals that DKN-01 suppresses GC tumor growth through activating cGAS-STING pathway to block macrophage M2 polarization.
Collapse
Affiliation(s)
- Xiaohuan Yang
- Department of Ultrasound, Central Hospital Affiliated To Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Yingying Qi
- Department of Ultrasound, Central Hospital Affiliated To Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Sisi Wang
- Department of Ultrasound, Shandong Public Health Clinical Center, No.2999 Gangxing West Road, High Tech Zone, Jinan, 250000, Shandong, China.
| |
Collapse
|
16
|
Kawasaki H. A mechanistic review-regulation of silica-induced pulmonary inflammation by IL-10 and exacerbation by Type I IFN. Inhal Toxicol 2025; 37:59-73. [PMID: 39955624 DOI: 10.1080/08958378.2025.2465378] [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: 04/12/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Occupational exposure to crystalline silica (CS) is known to induce silicosis, a chronic lung disease characterized by the formation of granulomas and severe lung fibrosis. Specifically, individuals exposed to low doses of CS may develop silicosis after a decade or more of exposure. Similarly, in rat silicosis models exposed to occupationally relevant doses of α-quartz, there is an initial phase characterized by minimal and well-controlled pulmonary inflammation, followed by the development of robust and persistent inflammation. During the initial phase, the inflammation provoked by α-quartz is subdued by two mechanisms. Firstly, α-quartz particles are engulfed by alveolar macrophages (AMs) of the alternatively activated (M2) subtype and interstitial macrophages (IMs), limiting their interaction with other lung cells. Secondly, the anti-inflammatory cytokine, interleukin (IL)-10, is constitutively expressed by these macrophages, further dampening the inflammatory response. In the later inflammatory phase, IL-10-dependent anti-inflammatory state is disrupted by Type I interferons (IFNs), leading to the production of pro-inflammatory cytokines in response to α-quartz, aided by lipopolysaccharides (LPS). This review delves into the complex pathways involving IL-10, LPS, and Type I IFNs in α-quartz-induced pulmonary inflammation, offering a detailed analysis of the underlying mechanisms and identifying areas for future research.
Collapse
|
17
|
Zhou Q, Chang M, Guo S, Zhang Y, Qu Q, Zhou Q, Li Z, Yao S. Honokiol ameliorates silica-induced lung fibrosis by inhibiting macrophage pyroptosis via modulating cGAS/STING signaling. Int Immunopharmacol 2025; 146:113812. [PMID: 39681061 DOI: 10.1016/j.intimp.2024.113812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Silicosis is a life-threatening occupational disease because of inhaling silica dust, leading to chronic inflammation, pyroptosis, and fibrosis. Unfortunately, it is still lacking effective pharmacological intervention currently. Honokiol (HKL), a natural extract with biological activity from Magnolia bark, is known for its antioxidant and anti-inflammatory biological effects. The current work aimed to investigate the therapeutic potential of HKL in mitigating silica-induced lung fibrosis and pyroptosis, particularly focusing on the cGAS/STING signaling pathway. The pulmonary pathological results shown in H&E and Masson's trichrome staining images confirmed the protective effects of HKL on lung tissue structure. In addition, HKL significantly reduced lung inflammation, collagen deposition, and oxidative stress compared to mice in the silicosis group. HKL treatment also alleviated silica-induced pyroptosis by suppressing the activation of the cGAS/STING signaling pathway in lung tissues. Moreover, the in vitro experiments using J774A.1 macrophages demonstrated that HKL reduced pyroptosis and improved cell viability under exposure to silica combined lipopolysaccharide (LPS). These were attributed to HKL downregulating the activation of the cGAS/STING signaling pathway in pyroptotic J774A.1 cells induced by silica combined with LPS. Meanwhile, inhibition of STING signaling induced by DNase I significantly enhanced the protective effects of HKL on the inflammatory and pyroptotic processes induced by silica. Overall, HKL could attenuate silica-induced pyroptosis by modulating the cGAS/STING signaling pathway against pulmonary fibrosis. The current study offers a promising approach for treating silicosis and related inflammatory responses.
Collapse
Affiliation(s)
- Qiang Zhou
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; School of Public Health, North China University of Science and Technology, Tangshan 063000, China
| | - Meiyu Chang
- School of Public Health, North China University of Science and Technology, Tangshan 063000, China
| | - Shuhan Guo
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Yiming Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Qiufang Qu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Qingnan Zhou
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhiheng Li
- Institution for Occupational Disease Prevention and Treatment of China Pingmei Shenma Group, Pingdingshan 462500, China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; School of Public Health, North China University of Science and Technology, Tangshan 063000, China.
| |
Collapse
|
18
|
Zhao X, Du L, Ma N, Tan X, Lei X, Zhang P, Qu B. The role of cGAS-STING pathway in the development of radiation-induced lung injury. J Cancer Res Clin Oncol 2025; 151:48. [PMID: 39856464 PMCID: PMC11761121 DOI: 10.1007/s00432-025-06088-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND AND PURPOSE Radiation-induced lung injury (RILI) limits the efficacy of thoracic radiotherapy. However, the underlying mechanism of RILI remains unclear. cGAS-STING pathway is reported to be involved in the recognization of cytosolic dsDNA and various inflammatory diseases. This study aimed to investigate the role of cGAS-STING pathway in the development of RILI. MATERIALS AND METHODS A pre-clinical mouse model of RILI was established by whole thorax irradiation and confirmed using H&E and Masson's trichrome staining. STING agonist (DMXAA) and antagonist(C-176) were administrated to modulate cGAS-STING pathway in vivo. Western blot and ELISA were used to determine the expression levels of different proteins. RESULTS Quantitation analysis showed dsDNA accumulation in lung tissue and western blot showed the up-regulation of cGAS and STING protein level post-irradiation, indicating pathway activation. Histological evaluation showed that C-176 administration ameliorated radiation-induced pulmonary inflammation and fibrosis, while DMXAA exhibited contrary effects. In further in vitro study, the release of dsDNA induced by radiation led to the activation of cGAS-STING pathway in RAW 264.7 cells, resulting in the polarization into M1 phenotype and pro-inflammatory production. CONCLUSION In summary, our data demonstrated a link between cGAS-STING pathway and the development of RILI, indicating its potential application in clinic.
Collapse
Affiliation(s)
- Xinyao Zhao
- Department of Radiation Oncology, Shijingshan Teaching Hospital of Capital Medical University, Shijingshan Hospital, Beijing, China
| | - Lehui Du
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Na Ma
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xin Tan
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pei Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
19
|
Hou X, Wei Z, Jiang X, Wei C, Dong L, Li Y, Liang R, Nie J, Shi Y, Qin X. A comprehensive retrospect on the current perspectives and future prospects of pneumoconiosis. Front Public Health 2025; 12:1435840. [PMID: 39866352 PMCID: PMC11757636 DOI: 10.3389/fpubh.2024.1435840] [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: 05/21/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Pneumoconiosis is a widespread occupational pulmonary disease caused by inhalation and retention of dust particles in the lungs, is characterized by chronic pulmonary inflammation and progressive fibrosis, potentially leading to respiratory and/or heart failure. Workers exposed to dust, such as coal miners, foundry workers, and construction workers, are at risk of pneumoconiosis. This review synthesizes the international and national classifications, epidemiological characteristics, strategies for prevention, clinical manifestations, diagnosis, pathogenesis, and treatment of pneumoconiosis. Current research on the pathogenesis of pneumoconiosis focuses on the influence of autophagy, apoptosis, and pyroptosis on the progression of the disease. In addition, factors such as lipopolysaccharide and nicotine have been found to play crucial roles in the development of pneumoconiosis. This review provides a comprehensive summary of the most fundamental achievements in the treatment of pneumoconiosis with the purpose of indicating the future direction of its treatment and control. New technologies of integrative omics, artificial intelligence, systemic administration of mesenchymal stromal cells have proved useful in solving the conundrum of pneumoconiosis. These directional studies will provide novel therapeutic targets for the treatment of pneumoconiosis.
Collapse
Affiliation(s)
- Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
- Environmental Exposures Vascular Disease Institute, Shanxi Medical University, Taiyuan, Shanxi, China
- China Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhengqian Wei
- Department of General Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xuelu Jiang
- Environmental Exposures Vascular Disease Institute, Shanxi Medical University, Taiyuan, Shanxi, China
- Academy of Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chengjie Wei
- Environmental Exposures Vascular Disease Institute, Shanxi Medical University, Taiyuan, Shanxi, China
- Academy of Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lin Dong
- Environmental Exposures Vascular Disease Institute, Shanxi Medical University, Taiyuan, Shanxi, China
- Academy of Medical Science, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhua Li
- Department of Foreign Languages, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruifeng Liang
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jisheng Nie
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, Shanxi, China
| | - Yiwei Shi
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, Shanxi, China
- Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- NHC Key Laboratory of Pneumoconiosis, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaojiang Qin
- Environmental Exposures Vascular Disease Institute, Shanxi Medical University, Taiyuan, Shanxi, China
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, Shanxi, China
- NHC Key Laboratory of Pneumoconiosis, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
20
|
Chen Y, Wu X, Jiang Z, Li X. KAE ameliorates LPS-mediated acute lung injury by inhibiting PANoptosis through the intracellular DNA-cGAS-STING axis. Front Pharmacol 2025; 15:1461931. [PMID: 39840115 PMCID: PMC11747328 DOI: 10.3389/fphar.2024.1461931] [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: 07/09/2024] [Accepted: 12/19/2024] [Indexed: 01/23/2025] Open
Abstract
Background Acute lung injury (ALI) is a severe condition characterized by inflammation, tissue damage, and persistent activation of the cyclic GMP-AMP (cGAS)-stimulator of interferon genes (STING) pathway, which exacerbates the production of pro-inflammatory mediators and promotes the progression of ALI. Specific inhibition of this pathway has been shown to alleviate ALI symptoms. Kaempferol-3-O-α-L-(4″-E-p-coumaroyl)-rhamnoside (KAE), an active compound found in the flowers of Angelica acutiloba Kitagawa, exhibits anti-inflammatory and antioxidant properties. This study aimed to investigate the molecular mechanisms through which KAE regulates the cGAS-STING pathway in the context of ALI. Methods ALI was induced using LPS. Lung damage and anti-inflammatory/antioxidant effects were assessed by H&E staining, lung edema index, and SOD, MDA, and ELISA assays. NO release and mitochondrial membrane potential (MMP) were measured by JC-1 and Griess methods. The impact of KAE on the cGAS-STING pathway and PANoptosis was analyzed using flow cytometry, Western blot, and immunofluorescence. Results KAE significantly alleviated lipopolysaccharide-induced pulmonary injury by reducing inflammatory cell infiltration, alleviating pulmonary edema, enhancing antioxidant capacity, and decreasing levels of inflammatory cytokines in mouse lung tissues. In both in vitro and in vivo analyses, KAE downregulated the expression of key components of the cGAS-STING pathway, including cGAS, STING, p-TBK1, and nuclear factor-κB. KAE also reduced the assembly and activation of the PANoptosome, thereby attenuating apoptosis, necroptosis, and pyroptosis. Additionally, KAE inhibited cGAS activation by restoring the MMP, which reduced the release of cytosolic DNA. Conclusion KAE improve ALI by inhibiting the release of cytosolic DNA and suppressing cGAS-STING pathway activation, thereby protecting cells from PANoptosis. Our findings provide valuable insights for the development and application of novel therapeutic strategies for ALI.
Collapse
Affiliation(s)
| | | | | | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, China
| |
Collapse
|
21
|
Chen Y, Yang C, Miao Y, Shi D, Li X, Tian S, Zhang Y, Xu C, Dong Y, Han C, Shi H, Bai C. Macrophage STING signaling promotes fibrosis in benign airway stenosis via an IL6-STAT3 pathway. Nat Commun 2025; 16:289. [PMID: 39753529 PMCID: PMC11698984 DOI: 10.1038/s41467-024-55170-5] [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: 05/17/2024] [Accepted: 12/04/2024] [Indexed: 01/06/2025] Open
Abstract
Acute and chronic inflammation are important pathologies of benign airway stenosis (BAS) fibrosis, which is a frequent complication of critically ill patients. cGAS-STING signalling has an important role in inflammation and fibrosis, yet the function of STING in BAS remains unclear. Here we demonstrate using scRNA sequencing that cGAS‒STING signalling is involved in BAS, which is accompanied by increased dsDNA, expression and activation of STING. STING inhibition or deficiency effectively alleviates tracheal fibrosis of BAS mice by decreasing both acute and chronic inflammation. Macrophage depletion also effectively ameliorates BAS. Mechanistically, dsDNA from damaged epithelial cells activates the cGAS-STING pathway of macrophages and induces IL-6 to activate STAT3 and promote fibrosis. In summary, the present results suggest that cGAS-STING signalling induces acute inflammation and amplifies the chronic inflammation and tracheal fibrosis associated with benign airway stenosis, highlighting the mechanism and potential drug target of BAS.
Collapse
Affiliation(s)
- YiLin Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - ChengCheng Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - YuShan Miao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - DongChen Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiang Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Sen Tian
- Department of Respiratory and Critical Care Medicine, No. 906 Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, Ningbo, China
| | - YiFei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - ChengFei Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - YuChao Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - ChaoFeng Han
- Department of Histology and Embryology, Naval Medical University, Shanghai, China.
| | - Hui Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
22
|
Parks CG, Wilkerson J, Rose KM, Faiq A, Farhadi PN, Bayat N, Schiffenbauer A, Brunner HI, Goldberg B, Sandler DP, Miller FW, Rider LG. Occupational and Hobby Exposures Associated With Myositis Phenotypes in a National Myositis Patient Registry. Arthritis Care Res (Hoboken) 2025; 77:104-115. [PMID: 39530281 DOI: 10.1002/acr.25461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE The objective of this study was to investigate occupational and hobby exposures to silica, solvents, and heavy metals and the odds of having the idiopathic inflammatory myopathy (IIM) phenotypes dermatomyositis (DM) and polymyositis (PM) versus inclusion body myositis (IBM), lung disease plus fever or arthritis (LD+), and systemic autoimmune rheumatic disease-associated overlap myositis (OM). METHODS The sample included 1,390 patients (598 with DM, 409 with PM, and 383 with IBM) aged ≥18 years from a national registry. Of these, 218 (16%) were identified with LD+, and 166 (12%) with OM. Of these, 218 (16%) were identified with LD+, and 166 (12%) with OM. We calculated adjusted odds ratios (ORs) and 95% confidence intervals (CIs) and explored joint effects with smoking. RESULTS High silica exposure was associated with increased odds of having DM (OR 2.02, 95% CI 1.18-3.46, compared to no exposure; P trend = 0.004), LD+ (OR 1.75, 95% CI 1.10-2.78, vs no LD; P trend = 0.005), and OM (OR 2.07, 95% CI 1.19-3.61, P trend = 0.020). Moderate to high heavy metals exposure was associated with greater odds of having LD+ (OR 1.49, 95% CI 1.00-2.14, P trend = 0.026) and OM (OR 1.59, 95% CI 0.99-2.55, P trend = 0.051). Greater odds of having LD+ were seen among smokers with moderate to high silica exposure versus nonsmokers with low or no exposure (high-certainty assessment OR 2.53, 95% CI 1.31-4.90, P interaction = 0.061). CONCLUSION These findings, based on a systematic exposure assessment, suggest that occupational and hobby exposures to silica and heavy metals contribute to adult IIM phenotypes, including DM, OM, and LD+, a possible marker for antisynthetase syndrome or other autoantibody-associated lung diseases.
Collapse
Affiliation(s)
- Christine G Parks
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina
| | | | | | - Abdullah Faiq
- National Institute of Environmental Health Sciences, NIH, Bethesda, Maryland
| | | | - Nastaran Bayat
- National Institute of Environmental Health Sciences, NIH, Bethesda, Maryland
| | - Adam Schiffenbauer
- National Institute of Environmental Health Sciences, NIH, Bethesda, Maryland
| | | | | | - Dale P Sandler
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina
| | - Frederick W Miller
- National Institute of Environmental Health Sciences, NIH, Bethesda, Maryland
| | - Lisa G Rider
- National Institute of Environmental Health Sciences, NIH, Bethesda, Maryland
| |
Collapse
|
23
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
24
|
Sood A, Kulharia M. Inhibition of IRF3-STING axis interaction in silicosis using natural compounds: an in-silico study using molecular docking, ADMET, molecular dynamics and MMPBSA approach. In Silico Pharmacol 2024; 13:1. [PMID: 39659978 PMCID: PMC11625707 DOI: 10.1007/s40203-024-00290-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/30/2024] [Indexed: 12/12/2024] Open
Abstract
Silicosis is a chronic occupational lung disease characterized by persistent inflammation driven by the activation of the cGAS-STING pathway, leading to the downstream activation of IRF3. To develop a natural compound library of COCONUT database for this investigation, Lipinski's rule of five was used and we explored the potential of these compounds to disrupt the IRF3-STING interaction, thereby mitigating the inflammatory response. Molecular docking and molecular dynamics (MD) simulations were employed to assess the binding stability and interaction dynamics of these compounds with IRF3. The stable RMSD values indicate that the protein-ligand complexes maintained structural integrity throughout the simulation period. The compounds also demonstrated drug-like characteristics, a promising safety profile, and formed stable complexes with the target protein. Further, decomposition of binding free energy highlighted the key contributions of IRF3 residues VAL295, ASP308, PRO324, and ARG338 interacting with the selected compounds, potentially inhibiting the IRF3-STING interaction. The origin of the selected compounds was determined using ClassyFire, classifying compound CNP0310627 as a burfenolide and compound CNP0200121 as a psoralen. Both classes are recognized for their anti-inflammatory properties, reinforcing the therapeutic potential of these compounds in reducing inflammation associated with silicosis. Our findings suggest that these compounds could serve as promising candidates for further investigation in the development of anti-inflammatory therapeutic molecules in the cGAS-STING-IRF3 signaling pathway. However, to fully assess the therapeutic potential of these compounds, further in vitro and in vivo studies are required to validate their efficacy and safety in modulating the STING-IRF3 pathway. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00290-5.
Collapse
Affiliation(s)
- Ashita Sood
- Centre for Computational Biology and Bioinformatics, Central University of Himachal Pradesh, Dadroli, India
| | - Mahesh Kulharia
- Centre for Computational Biology and Bioinformatics, Central University of Himachal Pradesh, Dadroli, India
| |
Collapse
|
25
|
Huang Z, Wei C, Xie H, Xiao X, Wang T, Zhang Y, Chen Y, Hei Z, Zhao T, Yao W. Treating acute lung injury through scavenging of cell-free DNA by cationic nanoparticles. Mater Today Bio 2024; 29:101360. [PMID: 39687793 PMCID: PMC11648789 DOI: 10.1016/j.mtbio.2024.101360] [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: 09/09/2024] [Revised: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome are life-threatening conditions induced by inflammatory responses, in which cell-free DNA (cfDNA) plays a pivotal role. This study investigated the therapeutic potential of biodegradable cationic nanoparticles (cNPs) in alleviating ALI. Using a mouse model of lipopolysaccharide-induced ALI, we examined the impact of intravenously administered cNPs. Our findings indicate that cNPs possess robust DNA binding capability, enhanced accumulation in inflamed lungs, and a favorable safety profile in vivo. Furthermore, cNPs attenuate the inflammatory response in LPS-induced ALI mice by scavenging cfDNA, mainly derived from neutrophil extracellular traps, and activating the macrophage-mediated cGAS-STING pathway. The findings suggest a potential treatment for ALI by targeting cfDNA with cNPs. This approach has demonstrated efficacy in mitigating lung injury and merits further exploration.
Collapse
Affiliation(s)
- Ziyan Huang
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Cong Wei
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Hanbin Xie
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Xue Xiao
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Tienan Wang
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Yihan Zhang
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Yongming Chen
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| | - Tianyu Zhao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Weifeng Yao
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, PR China
| |
Collapse
|
26
|
Xiang M, Chen C, Chen Y, Zhang Y, Shi L, Chen Y, Li J, Li B, Zeng B, Xing HR, Wang J, Zou Z. Unexpected Inhibitory Role of Silica Nanoparticles on Lung Cancer Development by Promoting M1 Polarization of Macrophages. Int J Nanomedicine 2024; 19:11087-11104. [PMID: 39502640 PMCID: PMC11537155 DOI: 10.2147/ijn.s472796] [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: 06/18/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Inhalation exposure to silica nanoparticles (SiNPs) is frequently inevitable in modern times. Although the impact of SiNPs on the ecological niche of the lungs has been extensively explored, the role and mechanism of SiNPs in the microenvironment of lung tumors remain elusive. Methods In this investigation, Lewis lung carcinoma (LLC) was implanted into the left lung in situ after 28 days of intratracheal SiNPs injection into the lungs of mice. This study evaluates the effects of SiNPs on the tumor immune microenvironment both in vitro and in vivo. Our findings indicate that SiNPs can suppress lung cancer by modulating the immune microenvironment of tumors. Results SiNPs treatment promotes macrophage M1 polarization by activating both NF-κB pathway and glycolytic mechanisms. This phenomenon may be associated with lung inflammation and fluctuation in the pre-metastatic and metastatic microenvironments induced by SiNPs exposure in mice. Additionally, we have shown for the first time that SiNPs have an inhibitory effect on lung carcinogenesis and its progression. Conclusion This study uniquely demonstrates that SiNPs suppress lung cancer by promoting M1 polarization of macrophages in the immune microenvironment of lung tumors. Our findings are critical in exploring the interaction between SiNPs and lung cancer.
Collapse
Affiliation(s)
- Meng Xiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Chengzhi Chen
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuting Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuhan Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Lei Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jie Li
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Bowen Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Bin Zeng
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - H Rosie Xing
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jianyu Wang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
27
|
Guirriec Y, Luque-Paz D, Bernard G, Mabo A, Kerjouan M, Ménard C, Monnier D, Nunes H, Uzunhan Y, Reynaud-Gaubert M, Bermudez J, Borie R, Crestani B, Traclet J, Wémeau-Stervinou L, Chenivesse C, Gomez E, Prévot G, Bourdin A, Bondue B, Bergeron A, Cottin V, Lederlin M, Jouneau S. Pulmonary fibrosis in patients with autoimmune pulmonary alveolar proteinosis: a retrospective nationwide cohort study. ERJ Open Res 2024; 10:00314-2024. [PMID: 39624377 PMCID: PMC11610044 DOI: 10.1183/23120541.00314-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/25/2024] [Indexed: 12/18/2024] Open
Abstract
Background Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare disease that may progress towards pulmonary fibrosis. Data about fibrosis prevalence and risk factors are lacking. Methods In this retrospective multicentre nationwide cohort, we included patients newly diagnosed with aPAP between 2008 and 2018 in France and Belgium. Data were collected from medical records using a standardised questionnaire. Results 61 patients were included in the final analysis. We identified 5 patients (8%) with fibrosis on initial computed tomography (CT) and 16 patients (26%) with fibrosis on final CT after a median time of 3.6 years. Dust exposure was associated with pulmonary fibrosis occurrence (OR 4.3; p=0.038). aPAP patients treated with whole-lung lavage, rituximab or granulocyte-monocyte colony-stimulating factor therapy did not have more fibrotic evolution than patients who did not receive these treatments (n=25 out of 45, 57% versus n=10 out of 16, 62%; p=0.69). All-cause mortality was significantly higher in fibrotic than in nonfibrotic cases (n=4 out of 16, 25% versus n=2 out of 45, 4.4%; p=0.036, respectively). Conclusion In our population, a quarter of aPAP patients progressed towards pulmonary fibrosis. Dust exposure seems to be an important factor associated with this complication. More studies are needed to analyse precisely the impact of dust exposure impact, especially silica, in patients with aPAP.
Collapse
Affiliation(s)
- Yoann Guirriec
- Pneumologie, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- These authors contributed equally
| | - David Luque-Paz
- Pneumologie, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- These authors contributed equally
| | - Gontran Bernard
- Imagerie médicale, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Axelle Mabo
- Pneumologie, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | | | - Cédric Ménard
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Delphine Monnier
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Hilario Nunes
- Centre de référence constitutif des maladies pulmonaires rares, Service de Pneumologie, AP-HP, Hôpital Avicenne, INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France
| | - Yurdagül Uzunhan
- Centre de référence constitutif des maladies pulmonaires rares, Service de Pneumologie, AP-HP, Hôpital Avicenne, INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France
| | - Martine Reynaud-Gaubert
- Centre de compétence pour les maladies pulmonaires rares, Service de Pneumologie, AP-HM, CHU Nord, Marseille, France
| | - Julien Bermudez
- Centre de compétence pour les maladies pulmonaires rares, Service de Pneumologie, AP-HM, CHU Nord, Marseille, France
| | - Raphaël Borie
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France
- Université Paris Cité, Inserm, PHERE, Université Paris Cité, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France
- Université Paris Cité, Inserm, PHERE, Université Paris Cité, Paris, France
| | - Julie Traclet
- Centre national coordonnateur de référence des maladies pulmonaires rares, Service de Pneumologie, Hôpital Louis-Pradel, Hospices Civils de Lyon, UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - Lidwine Wémeau-Stervinou
- Univ. Lille, CNRS, Inserm, CHU de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Centre de référence constitutif des maladies pulmonaires rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-allergologie, Lille, France
| | - Cécile Chenivesse
- Univ. Lille, CNRS, Inserm, CHU de Lille, U1019 – UMR 9017 – CIIL – Center for Infection and Immunity of Lille, Centre de référence constitutif des maladies pulmonaires rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-allergologie, Lille, France
| | - Emmanuel Gomez
- Centre de compétence pour les maladies pulmonaires rares, département de pneumologie, Hôpitaux de Brabois, CHRU de Nancy, Vandoeuvre-les Nancy, France
| | - Grégoire Prévot
- Centre de compétence pour les maladies pulmonaires rares, Service de Pneumologie, CHU Larrey, Toulouse, France
| | - Arnaud Bourdin
- Service de Pneumologie, Centre Hospitalier Universitaire de Montpellier, Université de Montpellier, Montpellier, France
| | - Benjamin Bondue
- Service de Pneumologie, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
| | - Anne Bergeron
- Centre de référence constitutif des maladies pulmonaires rares, Service de Pneumologie, AP-HP, Hôpital Saint Louis, Paris, France
| | - Vincent Cottin
- Centre national coordonnateur de référence des maladies pulmonaires rares, Service de Pneumologie, Hôpital Louis-Pradel, Hospices Civils de Lyon, UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - Mathieu Lederlin
- Imagerie médicale, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- These authors contributed equally
| | - Stéphane Jouneau
- Pneumologie, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Inserm UMR1085 IRSET, Université de Rennes 1, EHESP, Rennes, France
- These authors contributed equally
| | | |
Collapse
|
28
|
Tian M, Li F, Pei H, Liu X, Nie H. The role of the cGAS-STING pathway in chronic pulmonary inflammatory diseases. Front Med (Lausanne) 2024; 11:1436091. [PMID: 39540037 PMCID: PMC11557406 DOI: 10.3389/fmed.2024.1436091] [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: 05/22/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024] Open
Abstract
The innate immune system plays a vital role in the inflammatory process, serving as a crucial mechanism for the body to respond to infection, cellular stress, and tissue damage. The cGAS-STING signaling pathway is pivotal in the onset and progression of various autoimmune diseases and chronic inflammation. By recognizing cytoplasmic DNA, this pathway initiates and regulates inflammation and antiviral responses within the innate immune system. Consequently, the regulation of the cGAS-STING pathway has become a prominent area of interest in the treatment of many diseases. Chronic inflammatory lung diseases, such as chronic obstructive pulmonary disease (COPD), asthma, and pulmonary fibrosis, are characterized by persistent or recurrent lung inflammation and tissue damage, leading to diminished respiratory function. This paper explores the mechanism of action of the cGAS-STING signaling pathway in these diseases, examines the development of STING inhibitors and nanomaterial applications, and discusses the potential clinical application prospects of targeting the cGAS-STING pathway in chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Mengxiang Tian
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Fengyuan Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoling Liu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyun Nie
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing, China
| |
Collapse
|
29
|
Xu T, Zhong X, Luo N, Ma W, Hao P. Review of Excessive Cytosolic DNA and Its Role in AIM2 and cGAS-STING Mediated Psoriasis Development. Clin Cosmet Investig Dermatol 2024; 17:2345-2357. [PMID: 39464745 PMCID: PMC11512523 DOI: 10.2147/ccid.s476785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
In psoriasis, keratinocytes are triggered by factors, such as infection or tissue damage, to release DNA, which thereby activates plasmacytoid dendritic cells and macrophages to induce inflammation, thickened epidermis, and parakeratosis. The recognition of double-stranded (ds)DNA facilitates the activation of cytoplasmic DNA sensors absent in melanoma 2 (AIM2) inflammasome assembly and cyclic guanosine monophosphate adenosine monophosphate (cGAMP) synthase (cGAS) - stimulator of interferon gene (STING) pathway, both of which play a pivotal role in mediating the inflammatory response and driving the progression of psoriasis. Additionally, secreted proinflammatory cytokines can stimulate further DNA release from keratinocytes. Notably, the activation of AIM2 and cGAS-STING signaling pathways also mediates programmed cell death, potentially enhancing DNA overproduction. As a result, excessive DNA can activate these pathways, amplifying persistent inflammatory responses that contribute to the maintenance of psoriasis. Several studies have validated that targeting DNA and its mediated activation of AIM2 and cGAS-STING offers promising therapeutic strategies for psoriasis. Here, we postulate a hypothesis that excessive cytosolic DNA can activate AIM2 and cGAS-STING, mediating inflammation and programmed cell death, ultimately fostering DNA accumulation and contributing to the development of psoriasis.
Collapse
Affiliation(s)
- Tongtong Xu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaojing Zhong
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Nana Luo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenyi Ma
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Pingsheng Hao
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
30
|
Liu J, Xu F, Guo M, Gao D, Song Y. Nasal instillation of polystyrene nanoplastics induce lung injury via mitochondrial DNA release and activation of the cyclic GMP-AMP synthase-stimulator of interferon genes-signaling cascade. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174674. [PMID: 39002594 DOI: 10.1016/j.scitotenv.2024.174674] [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: 04/23/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Nanoplastics (NPs) are a common type of degraded plastic material associated with adverse health effects such as pulmonary injury. However, the molecular mechanism(s) underlying lung injury as caused by NPs remains uncertain. Thus, we herein investigated the pulmonary toxicity of NPs on RAW264.7 cells and C57BL/6 mice. Our in vitro study indicated that NPs induced oxidative stress, cell death, inflammation, and the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-signaling pathway. Mice in our in vivo study displayed significant pulmonary fibrosis, inflammation, apoptosis, necrosis, and excessive double-stranded DNA release into serum and bronchoalveolar lavage fluid. Our mechanistic exploration uncovered cGAS-STING-signaling activation as the leading cause of NPs-induced pulmonary fibrosis. The current study opens an avenue toward elucidating the role of the cGAS-STING-signaling pathway in NPs-induced pulmonary injury.
Collapse
Affiliation(s)
- Jing Liu
- College of Eco-Environmental Engineering, Guizhou Minzu University, GuiYang 550025, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 10085, China
| | - Fang Xu
- College of Eco-Environmental Engineering, Guizhou Minzu University, GuiYang 550025, China
| | - Mingzhu Guo
- College of Eco-Environmental Engineering, Guizhou Minzu University, GuiYang 550025, China
| | - Daxue Gao
- College of Eco-Environmental Engineering, Guizhou Minzu University, GuiYang 550025, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 10085, China.
| |
Collapse
|
31
|
Auld SC, Sheshadri A, Alexander-Brett J, Aschner Y, Barczak AK, Basil MC, Cohen KA, Dela Cruz C, McGroder C, Restrepo MI, Ridge KM, Schnapp LM, Traber K, Wunderink RG, Zhang D, Ziady A, Attia EF, Carter J, Chalmers JD, Crothers K, Feldman C, Jones BE, Kaminski N, Keane J, Lewinsohn D, Metersky M, Mizgerd JP, Morris A, Ramirez J, Samarasinghe AE, Staitieh BS, Stek C, Sun J, Evans SE. Postinfectious Pulmonary Complications: Establishing Research Priorities to Advance the Field: An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc 2024; 21:1219-1237. [PMID: 39051991 DOI: 10.1513/annalsats.202406-651st] [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: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Continued improvements in the treatment of pulmonary infections have paradoxically resulted in a growing challenge of individuals with postinfectious pulmonary complications (PIPCs). PIPCs have been long recognized after tuberculosis, but recent experiences such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic have underscored the importance of PIPCs following other lower respiratory tract infections. Independent of the causative pathogen, most available studies of pulmonary infections focus on short-term outcomes rather than long-term morbidity among survivors. In this document, we establish a conceptual scope for PIPCs with discussion of globally significant pulmonary pathogens and an examination of how these pathogens can damage different components of the lung, resulting in a spectrum of PIPCs. We also review potential mechanisms for the transition from acute infection to PIPC, including the interplay between pathogen-mediated injury and aberrant host responses, which together result in PIPCs. Finally, we identify cross-cutting research priorities for the field to facilitate future studies to establish the incidence of PIPCs, define common mechanisms, identify therapeutic strategies, and ultimately reduce the burden of morbidity in survivors of pulmonary infections.
Collapse
|
32
|
Bian Y, Deng M, Liu J, Li J, Zhang Q, Wang Z, Liao L, Miao J, Li R, Zhou X, Hou G. The glycyl-l-histidyl-l-lysine-Cu 2+ tripeptide complex attenuates lung inflammation and fibrosis in silicosis by targeting peroxiredoxin 6. Redox Biol 2024; 75:103237. [PMID: 38879894 PMCID: PMC11228880 DOI: 10.1016/j.redox.2024.103237] [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: 05/16/2024] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024] Open
Abstract
Silicosis is the most common type of pneumoconiosis, having a high incidence in workers chronically exposed to crystalline silica (CS). No specific medication exists for this condition. GHK, a tripeptide naturally occurring in human blood and urine, has antioxidant effects. We aimed to investigate the therapeutic effect of GHK-Cu on silicosis and its potential underlying molecular mechanism. An experimental silicosis mouse model was established to observe the effects of GHK-Cu on lung inflammation and fibrosis. Moreover, the effects of GHK-Cu on the alveolar macrophages (AM) were examined using the RAW264.7 cell line. Its molecular target, peroxiredoxin 6 (PRDX6), has been identified, and GHK-Cu can bind to PRDX6, thus attenuating lung inflammation and fibrosis in silicosis mice without significant systemic toxicity. These effects were partly related to the inhibition of the CS-induced oxidative stress in AM induced by GHK-Cu. Thus, our results suggest that GHK-Cu acts as a potential drug by attenuating alveolar macrophage oxidative stress. This, in turn, attenuates the progression of pulmonary inflammation and fibrosis, which provides a reference for the treatment of silicosis.
Collapse
Affiliation(s)
- Yiding Bian
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; China-Japan Friendship Hospital (Institution of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingming Deng
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiaye Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qin Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Zilin Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; China-Japan Friendship Hospital (Institution of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liwei Liao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jinrui Miao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ruixia Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoming Zhou
- Department of Pulmonary and Critical Care Medicine, Disease, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Gang Hou
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
33
|
Liu X, Jiang T, Jin H, Yan C, Tong Y, Ding J, Li Y, Huang L, Zhang Z. mtDNA amplifies beryllium sulfate-induced inflammatory responses via the cGAS-STING pathway in 16HBE cells. J Appl Toxicol 2024; 44:1403-1415. [PMID: 38778435 DOI: 10.1002/jat.4631] [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: 04/08/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
Beryllium sulfate (BeSO4) can cause inflammation through the mechanism, which has not been elucidated. Mitochondrial DNA (mtDNA) is a key contributor of inflammation. With mitochondrial damage, released mtDNA can bind to specific receptors (e.g., cGAS) and then activate related pathway to promote inflammatory responses. To investigate the mechanism of mtDNA in BeSO4-induced inflammatory response in 16HBE cells, we established the BeSO4-induced 16HBE cell inflammation model and the ethidium bromide (EB)-induced ρ016HBE cell model to detect the mtDNA content, oxidative stress-related markers, mitochondrial membrane potential, the expression of the cGAS-STING pathway, and inflammation-related factors. Our results showed that BeSO4 caused oxidative stress, decline of mitochondrial membrane potential, and the release of mtDNA into the cytoplasm of 16HBE cells. In addition, BeSO4 induced inflammation in 16HBE cells by activating the cGAS-STING pathway. Furthermore, mtDNA deletion inhibited the expression of cGAS-STING pathway, IL-10, TNF-α, and IFN-β. This study revealed a novel mechanism of BeSO4-induced inflammation in 16HBE cells, which contributes to the understanding of the molecular mechanism of beryllium and its compounds-induced toxicity.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Tianyi Jiang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Huiyun Jin
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Chenxi Yan
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuqi Tong
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiaquan Ding
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Yaqi Li
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Lian Huang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhaohui Zhang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
34
|
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
|
35
|
Zhou Q, Yi G, Chang M, Li N, Bai Y, Li H, Yao S. Activation of Sirtuin3 by honokiol ameliorates alveolar epithelial cell senescence in experimental silicosis via the cGAS-STING pathway. Redox Biol 2024; 74:103224. [PMID: 38865904 PMCID: PMC11215422 DOI: 10.1016/j.redox.2024.103224] [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: 05/08/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Silicosis, characterized by interstitial lung inflammation and fibrosis, poses a significant health threat. ATII cells play a crucial role in alveolar epithelial repair and structural integrity maintenance. Inhibiting ATII cell senescence has shown promise in silicosis treatment. However, the mechanism behind silica-induced senescence remains elusive. METHODS The study employed male C57BL/6 N mice and A549 human alveolar epithelial cells to investigate silicosis and its potential treatment. Silicosis was induced in mice via intratracheal instillation of crystalline silica particles, with honokiol administered intraperitoneally for 14 days. Silica-induced senescence in A549 cells was confirmed, and SIRT3 knockout and overexpression cell lines were generated. Various analyses were conducted, including immunoblotting, qRT-PCR, histology, and transmission electron microscopy. Statistical significance was determined using one-way ANOVA with Tukey's post-hoc test. RESULTS This study elucidates how silica induces ATII cell senescence, emphasizing mtDNA damage. Notably, honokiol (HKL) emerges as a promising anti-senescence and anti-fibrosis agent, acting through sirt3. honokiol effectively attenuated senescence in ATII cells, dependent on sirt3 expression, while mitigating mtDNA damage. Sirt3, a class III histone deacetylase, regulates senescence and mitochondrial stress. HKL activates sirt3, protecting against pulmonary fibrosis and mitochondrial damage. Additionally, HKL downregulated cGAS expression in senescent ATII cells induced by silica, suggesting sirt3's role as an upstream regulator of the cGAS/STING signaling pathway. Moreover, honokiol treatment inhibited the activation of the NF-κB signaling pathway, associated with reduced oxidative stress and mtDNA damage. Notably, HKL enhanced the activity of SOD2, crucial for mitochondrial function, through sirt3-mediated deacetylation. Additionally, HKL promoted the deacetylation activity of sirt3, further safeguarding mtDNA integrity. CONCLUSIONS This study uncovers a natural compound, HKL, with significant anti-fibrotic properties through activating sirt3, shedding light on silicosis pathogenesis and treatment avenues.
Collapse
Affiliation(s)
- Qiang Zhou
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China; School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Guan Yi
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China.
| | - Meiyu Chang
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China.
| | - Ning Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China.
| | - Yichun Bai
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Haibin Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China; School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Sanqiao Yao
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China; School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
36
|
Wang L, Zhang Z, Zhang H, Zhou M, Huang C, Xia W, Li J, You H. The effects of cGAS-STING inhibition in liver disease, kidney disease, and cellular senescence. Front Immunol 2024; 15:1346446. [PMID: 39114669 PMCID: PMC11303230 DOI: 10.3389/fimmu.2024.1346446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway is one of the fundamental mechanisms of the body's defense, which responds to the abnormal presence of double-stranded DNA in the cytoplasm to establish an effective natural immune response. In addition to detecting microbial infections, the cGAS pathway may be triggered by any cytoplasmic DNA, which is absent from the normal cytoplasm, and only conditions such as senescence and mitochondrial stress can lead to its leakage and cause sterile inflammation. A growing body of research has shown that the cGAS-STING pathway is strongly associated with sterile inflammation. In this study, we reviewed the regulatory mechanisms and biological functions of the cGAS-STING pathway through its involvement in aseptic inflammation in liver disease, kidney disease, and cellular senescence.
Collapse
Affiliation(s)
- Ling Wang
- Department of Pharmacy, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Zhengwei Zhang
- Department of Pharmacy, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Haichao Zhang
- Department of Pharmacy, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Minmin Zhou
- Department of Pharmacy, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wenjiang Xia
- Department of Pharmacy, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hongmei You
- Department of Pharmacy, Hangzhou Women’s Hospital, Hangzhou, China
| |
Collapse
|
37
|
Lescoat A, Rimar D, Farge D. Systemic sclerosis, silica exposure and cellular therapies: The sand in the gears? Rev Med Interne 2024; 45:431-436. [PMID: 38395716 DOI: 10.1016/j.revmed.2024.02.003] [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: 11/14/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Systemic sclerosis (SSc) is a chronic orphan autoimmune disease with the highest mortality rate among rheumatic diseases. SSc-related interstitial-lung disease (ILD) remains among the leading causes of SSc-related mortality with still few therapeutic effective strategies. In patients with crystallin silica exposure, SSc is recognized as an occupational disease according to the French social security system (Table 25A of the general insurance regimen). Lympho-ablative or myeloablative immunosuppression followed by autologous hematopoietic stem-cell transplantation (aHSCT) is the only therapeutic approach with demonstrated efficacy, improved survival with disease modifying effects on SSc-fibrotic manifestations (skin disease and ILD) and quality of life. A documented past and/or present occupational silica exposure, with extensive exposure and/or silica-related ILD and/or with persistent silica content in the broncho-alveolar lavage fluid are contra-indications to aHSCT in SSc patients, due to the risk of silica-related malignancy or of SSc relapse. This article aims to discuss alternative options in SSc patients with a history of silica exposure, and how innovative cellular therapies (mesenchymal stromal cells, CAR cells) could represent new therapeutic options for these patients.
Collapse
Affiliation(s)
- A Lescoat
- Institut de recherche en santé, environnement et travail (Irset) - UMR_S 1085, CHU de Rennes, Inserm, EHESP, University of Rennes, Rennes, France; Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France.
| | - D Rimar
- Rheumatology Unit, Bnai-Zion, Medical Center, Technion Institute of Technology, Haifa, Israel
| | - D Farge
- Unité de médecine interne (UF04): CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, recherche clinique en hématologie, immunologie et transplantation, URP3518, hôpital St-Louis, AP-HP, université Paris Cité, IRSL, 75010 Paris, France; Department of Medicine, McGill University, H3A 1A1 Montreal, Canada
| |
Collapse
|
38
|
Wang Z, Zhang J, Wang T, Liu Z, Zhang W, Sun Y, Wu X, Shao H, Du Z. The value of single biomarkers in the diagnosis of silicosis: A meta-analysis. iScience 2024; 27:109948. [PMID: 38799583 PMCID: PMC11126947 DOI: 10.1016/j.isci.2024.109948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/09/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
This study aims to establish a scientific foundation for early detection and diagnosis of silicosis by conducting meta-analysis on the role of single biomarkers in independent diagnosis. The combined sensitivity (Sen), specificity (Spe), positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic score, and diagnostic odds ratio (DOR) were 0.84 (95% confidence interval (CI): 0.77-0.90), 0.83 (95% CI: 0.78-0.88), 5.08 (95% CI: 3.92-6.59), 0.19 (95% CI: 0.13-0.27), 3.31 (95% CI: 2.88-3.74) and 27.29 (95% CI: 17.77-41.91), respectively. The area under the curve (AUC) was 0.90 (95% CI: 0.88-0.93). The Fagan plot shows a positive posterior probability of 82% and a negative posterior probability of 15%. This study establishes an academic basis for the swift identification, mitigation, and control of silicosis through scientific approaches. The assessed biomarkers offer precision and dependability in silicosis diagnosis, opening novel paths for early detection and intervention, thereby mitigating the disease burden associated with silicosis.
Collapse
Affiliation(s)
- Zhuofeng Wang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Jiaqi Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Tian Wang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Zuodong Liu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Wanxin Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Yuxin Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Xi Wu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, P.R. China
| |
Collapse
|
39
|
Shi W, Xu G, Gao Y, Yang H, Liu T, Zhao J, Li H, Wei Z, Hou X, Chen Y, Wen J, Li C, Zhao J, Zhang P, Wang Z, Xiao X, Bai Z. Compound Danshen Dripping Pill effectively alleviates cGAS-STING-triggered diseases by disrupting STING-TBK1 interaction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155404. [PMID: 38507852 DOI: 10.1016/j.phymed.2024.155404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (IFN) genes (STING) pathway is critical in the innate immune system and can be mobilized by cytosolic DNA. The various inflammatory and autoimmune diseases progression is highly correlated with aberrant cGAS-STING pathway activation. While some cGAS-STING pathway inhibitor were identified, there are no drugs that can be applied to the clinic. Compound Danshen Dripping Pill (CDDP) has been successfully used in clinic around the world, but the most common application is limited to cardiovascular disease. Therefore, the purpose of the present investigation was to examine whether CDDP inhibits the cGAS-STING pathway and could be used as a therapeutic agent for multiple cGAS-STING-triggered diseases. METHODS BMDMs, THP1 cells or Trex1-/- BMDMs were stimulated with various cGAS-STING-agonists after pretreatment with CDDP to detect the function of CDDP on IFN-β and ISGs productionn. Next, we detect the influence on IRF3 and P65 nuclear translocation, STING oligomerization and STING-TBK1-IRF3 complex formation of CDDP. Additionally, the DMXAA-mediated activation mice model of cGAS-STING pathway was used to study the effects of CDDP. Trex1-/- mice model and HFD-mediated obesity model were established to clarify the efficacy of CDDP on inflammatory and autoimmune diseases. RESULTS CDDP efficacy suppressed the IRF3 phosphorylation or the generation of IFN-β, ISGs, IL-6 and TNF-α. Mechanistically, CDDP did not influence the STING oligomerization and IRF3-TBK1 and STING-IRF3 interaction, but remarkably eliminated the STING-TBK1 interaction, ultimately blocking the downstream responses. In addition, we also clarified that CDDP could suppress cGAS-STING pathway activation triggered by DMXAA, in vivo. Consistently, CDDP could alleviate multi-organ inflammatory responses in Trex1-/- mice model and attenuate the inflammatory disorders, incleding obesity-induced insulin resistance. CONCLUSION CDDP is a specifically cGAS-STING pathway inhibitor. Furthermore, we provide novel mechanism for CDDP and discovered a clinical agent for the therapy of cGAS-STING-triggered inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Wei Shi
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guang Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huijie Yang
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Liu
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jia Zhao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui Li
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ziying Wei
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaorong Hou
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuanyuan Chen
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jincai Wen
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chengwei Li
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jun Zhao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ping Zhang
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhongxia Wang
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China; National Key Laboratory of Kidney Diseases, China.
| | - Zhaofang Bai
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China; National Key Laboratory of Kidney Diseases, China.
| |
Collapse
|
40
|
Ednacot EMQ, Nabhani A, Dinh DM, Morehouse BR. Pharmacological potential of cyclic nucleotide signaling in immunity. Pharmacol Ther 2024; 258:108653. [PMID: 38679204 DOI: 10.1016/j.pharmthera.2024.108653] [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: 01/07/2024] [Revised: 03/16/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Cyclic nucleotides are important signaling molecules that play many critical physiological roles including controlling cell fate and development, regulation of metabolic processes, and responding to changes in the environment. Cyclic nucleotides are also pivotal regulators in immune signaling, orchestrating intricate processes that maintain homeostasis and defend against pathogenic threats. This review provides a comprehensive examination of the pharmacological potential of cyclic nucleotide signaling pathways within the realm of immunity. Beginning with an overview of the fundamental roles of cAMP and cGMP as ubiquitous second messengers, this review delves into the complexities of their involvement in immune responses. Special attention is given to the challenges associated with modulating these signaling pathways for therapeutic purposes, emphasizing the necessity for achieving cell-type specificity to avert unintended consequences. A major focus of the review is on the recent paradigm-shifting discoveries regarding specialized cyclic nucleotide signals in the innate immune system, notably the cGAS-STING pathway. The significance of cyclic dinucleotides, exemplified by 2'3'-cGAMP, in controlling immune responses against pathogens and cancer, is explored. The evolutionarily conserved nature of cyclic dinucleotides as antiviral agents, spanning across diverse organisms, underscores their potential as targets for innovative immunotherapies. Findings from the last several years have revealed a striking diversity of novel bacterial cyclic nucleotide second messengers which are involved in antiviral responses. Knowledge of the existence and precise identity of these molecules coupled with accurate descriptions of their associated immune defense pathways will be essential to the future development of novel antibacterial therapeutic strategies. The insights presented herein may help researchers navigate the evolving landscape of immunopharmacology as it pertains to cyclic nucleotides and point toward new avenues or lines of thinking about development of therapeutics against the pathways they regulate.
Collapse
Affiliation(s)
- Eirene Marie Q Ednacot
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Ali Nabhani
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - David M Dinh
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Benjamin R Morehouse
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA; Center for Virus Research, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
41
|
Wang Y, He X, Wang H, Hu W, Sun L. Qingfei xieding prescription ameliorates mitochondrial DNA-initiated inflammation in bleomycin-induced pulmonary fibrosis through activating autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117820. [PMID: 38286157 DOI: 10.1016/j.jep.2024.117820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingfei Xieding prescription was gradually refined and produced by Hangzhou Red Cross Hospital. The raw material includes Ephedra sinica Stapf, Morus alba L., Bombyx Batryticatus, Gypsum Fibrosum, Prunus armeniaca L. var. ansu Maxim., Houttuynia cordata Thunb. , Pueraria edulis Pamp. Paeonia L., Scutellaria baicalensis Georgi and Anemarrhena asphodeloides Bge. It is effective in clinical adjuvant treatment of patients with pulmonary diseases. AIM OF THE STUDY To explore the efficacy and underlying mechanism of Qingfei Xieding (QF) in the treatment of bleomycin-induced mouse model. MATERIALS AND METHODS TGF-β induced fibrotic phenotype in vitro. Bleomycin injection induced lung tissue fibrosis mouse model in vivo. Flow cytometry was used to detect apoptosis, cellular ROS and lipid oxidation. Mitochondria substructure was observed by transmission electron microscopy. Autophagolysosome and nuclear entry of P65 were monitored by immunofluorescence. Quantitative real-time PCR was performed to detect the transcription of genes associated with mtDNA-cGAS-STING pathway and subsequent inflammatory signaling activation. RESULTS TGF-β induced the expression of α-SMA and Collagen I, inhibited cell viability in lung epithelial MLE-12 cells that was reversed by QF-containing serum. TGF-β-mediated downregulation in autophagy, upregulation in lipid oxidation and ROS contents, and mitochondrial damage were rescued by QF-containing serum treatment, but CQ exposure, an autophagy inhibitor, prevented the protective role of QF. In addition to that, the decreased autophagolysosome in TGF-β-exposed MLE-12 cells was reversed by QF and restored to low level in the combination treatment of QF and CQ. Mechanistically, QF-containing serum treatment significantly inhibited mtDNA-cGAS-STING pathway and subsequent inflammatory signaling in TGF-β-challenged cells, which were abolished by CQ-mediated autophagy inhibition. In bleomycin-induced mouse model, QF ameliorated pulmonary fibrosis, reduced mortality, re-activated autophagy in lung tissues and restrained mtDNA-cGAS-STING inflammation pathway. However, the protective effects of QF in bleomycin-induced model mice were also abrogated by CQ. CONCLUSION QF alleviated bleomycin-induced pulmonary fibrosis by activating autophagy, inhibiting mtDNA-cGAS-STING pathway-mediated inflammation. This research recognizes the protection role of QF on bleomycin-induced mouse model, and offers evidence for the potentiality of QF in clinical application for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Yunguang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, PR China.
| | - Xinxin He
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, PR China.
| | - Huijie Wang
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, PR China.
| | - Wei Hu
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| | - Lifang Sun
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, PR China; Department of Tuberculosis, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
42
|
Wang Y, Huang X, Luo G, Xu Y, Deng X, Lin Y, Wang Z, Zhou S, Wang S, Chen H, Tao T, He L, Yang L, Yang L, Chen Y, Jin Z, He C, Han Z, Zhang X. The aging lung: microenvironment, mechanisms, and diseases. Front Immunol 2024; 15:1383503. [PMID: 38756780 PMCID: PMC11096524 DOI: 10.3389/fimmu.2024.1383503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
With the development of global social economy and the deepening of the aging population, diseases related to aging have received increasing attention. The pathogenesis of many respiratory diseases remains unclear, and lung aging is an independent risk factor for respiratory diseases. The aging mechanism of the lung may be involved in the occurrence and development of respiratory diseases. Aging-induced immune, oxidative stress, inflammation, and telomere changes can directly induce and promote the occurrence and development of lung aging. Meanwhile, the occurrence of lung aging also further aggravates the immune stress and inflammatory response of respiratory diseases; the two mutually affect each other and promote the development of respiratory diseases. Explaining the mechanism and treatment direction of these respiratory diseases from the perspective of lung aging will be a new idea and research field. This review summarizes the changes in pulmonary microenvironment, metabolic mechanisms, and the progression of respiratory diseases associated with aging.
Collapse
Affiliation(s)
- Yanmei Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Xuewen Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guofeng Luo
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunying Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiqian Deng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Haoran Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Tao
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Lei He
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Luchuan Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Li Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zi Jin
- Department of Anesthesiology and Pain Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Chengshi He
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Zhang
- Department of Emergency Medicine Center, Sichuan Province People’s Hospital University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
43
|
Peng Y, Yang Y, Li Y, Shi T, Xu N, Liu R, Luan Y, Yao Y, Yin C. Mitochondrial (mt)DNA-cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling promotes pyroptosis of macrophages via interferon regulatory factor (IRF)7/IRF3 activation to aggravate lung injury during severe acute pancreatitis. Cell Mol Biol Lett 2024; 29:61. [PMID: 38671352 PMCID: PMC11055249 DOI: 10.1186/s11658-024-00575-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Macrophage proinflammatory activation contributes to the pathology of severe acute pancreatitis (SAP) and, simultaneously, macrophage functional changes, and increased pyroptosis/necrosis can further exacerbate the cellular immune suppression during the process of SAP, where cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) plays an important role. However, the function and mechanism of cGAS-STING in SAP-induced lung injury (LI) remains unknown. METHODS Lipopolysaccharide (LPS) was combined with caerulein-induced SAP in wild type, cGAS -/- and sting -/- mice. Primary macrophages were extracted via bronchoalveolar lavage and peritoneal lavage. Ana-1 cells were pretreated with LPS and stimulated with nigericin sodium salt to induce pyroptosis in vitro. RESULTS SAP triggered NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation-mediated pyroptosis of alveolar and peritoneal macrophages in mouse model. Knockout of cGAS/STING could ameliorate NLRP3 activation and macrophage pyroptosis. In addition, mitochondrial (mt)DNA released from damaged mitochondria further induced macrophage STING activation in a cGAS- and dose-dependent manner. Upregulated STING signal can promote NLRP3 inflammasome-mediated macrophage pyroptosis and increase serum interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α levels and, thus, exacerbate SAP-associated LI (SAP-ALI). Downstream molecules of STING, IRF7, and IRF3 connect the mtDNA-cGAS-STING axis and the NLRP3-pyroptosis axis. CONCLUSIONS Negative regulation of any molecule in the mtDNA-cGAS-STING-IRF7/IRF3 pathway can affect the activation of NLRP3 inflammasomes, thereby reducing macrophage pyroptosis and improving SAP-ALI in mouse model.
Collapse
Affiliation(s)
- Yiqiu Peng
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yuxi Yang
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yingying Li
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Tingjuan Shi
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Ning Xu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Ruixia Liu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yingyi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China.
| | - Yongming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.
| | - Chenghong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China.
| |
Collapse
|
44
|
Gao KM, Chiang K, Jiang Z, Korkmaz FT, Janardhan HP, Trivedi CM, Quinton LJ, Gingras S, Fitzgerald KA, Marshak-Rothstein A. Endothelial cell expression of a STING gain-of-function mutation initiates pulmonary lymphocytic infiltration. Cell Rep 2024; 43:114114. [PMID: 38625791 PMCID: PMC11108094 DOI: 10.1016/j.celrep.2024.114114] [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: 08/19/2023] [Revised: 03/13/2024] [Accepted: 03/29/2024] [Indexed: 04/18/2024] Open
Abstract
Patients afflicted with Stimulator of interferon gene (STING) gain-of-function mutations frequently present with debilitating interstitial lung disease (ILD) that is recapitulated in mice expressing the STINGV154M mutation (VM). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in initiating ILD. To identify STING-expressing non-hematopoietic cell types required for the development of ILD, we use a conditional knockin (CKI) model and direct expression of the VM allele to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted VM expression results in enhanced recruitment of immune cells to the lung associated with elevated chemokine expression and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of STING-associated vasculopathy with onset in infancy (SAVI) patients or patients afflicted with other ILD-related disorders.
Collapse
Affiliation(s)
- Kevin MingJie Gao
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kristy Chiang
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Zhaozhao Jiang
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Filiz T Korkmaz
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Harish P Janardhan
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Lee J Quinton
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Katherine A Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| | - Ann Marshak-Rothstein
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
45
|
Xuan L, Wang Y, Qu C, Yi W, Yang J, Pan H, Zhang J, Chen C, Bai C, Zhou PK, Huang R. Exposure to polystyrene nanoplastics induces abnormal activation of innate immunity via the cGAS-STING pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116255. [PMID: 38552388 DOI: 10.1016/j.ecoenv.2024.116255] [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: 11/01/2023] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/12/2024]
Abstract
Endogenous immune defenses provide an intrinsic barrier against external entity invasion. Microplastics in the environment, especially those at the nanoscale (nanoplastics or NPs), may pose latent health risks through direct exposure. While links between nanoplastics and inflammatory processes have been established, detailed insights into how they may perturb the innate immune mechanisms remain uncharted. Employing murine and macrophage (RAW264.7) cellular models subjected to polystyrene nanoplastics (PS-NPs), our investigative approach encompassed an array of techniques: Cell Counting Kit-8 assays, flow cytometric analysis, acridine orange/ethidium bromide (AO/EB) fluorescence staining, cell transfection, cell cycle scrutiny, genetic manipulation, messenger RNA expression profiling via quantitative real-time PCR, and protein expression evaluation through western blotting. The results showed that PS-NPs caused RAW264.7 cell apoptosis, leading to cell cycle arrest, and activated the cGAS-STING pathway. This resulted in NF-κB signaling activation and increased pro-inflammatory mediator expression. Importantly, PS-NPs-induced activation of NF-κB and its downstream inflammatory cascade were markedly diminished after the silencing of the STING gene. Our findings highlight the critical role of the cGAS-STING pathway in the immunotoxic effects induced by PS-NPs. We outline a new mechanism whereby nanoplastics may trigger dysregulated innate immune and inflammatory responses via the cGAS/STING pathway.
Collapse
Affiliation(s)
- Lihui Xuan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Yin Wang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China
| | - Wensen Yi
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China
| | - Jingjing Yang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China
| | - Huiji Pan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Jing Zhang
- Clinical Medical Oncology, Xiangya Medical College, Central South University, China.
| | - Cuimei Chen
- School of Public Health, Xiang Nan University, Chenzhou, Hunan 423000, China.
| | - Chenjun Bai
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| |
Collapse
|
46
|
Morse J, Wang D, Mei S, Whitham D, Hladun C, Darie CC, Sintim HO, Wang M, Leung K. Chloride Homeostasis Regulates cGAS-STING Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588475. [PMID: 38645072 PMCID: PMC11030317 DOI: 10.1101/2024.04.08.588475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The cGAS-STING signaling pathway has emerged as a key mediator of inflammation. However, the roles of chloride homeostasis on this pathway are unclear. Here, we uncovered a correlation between chloride homeostasis and cGAS-STING signaling. We found that dysregulation of chloride homeostasis attenuates cGAS-STING signaling in a lysosome-independent manner. Treating immune cells with chloride channel inhibitors attenuated 2'3'-cGAMP production by cGAS and also suppressed STING polymerization, leading to reduced cytokine production. We also demonstrate that non-selective chloride channel blockers can suppress the NPC1 deficiency-induced, hyper-activated STING signaling in skin fibroblasts derived from Niemann Pick disease type C (NPC) patients. Our findings reveal that chloride homeostasis majorly affects cGAS-STING pathway and suggest a provocative strategy to dampen STING-mediated inflammation via targeting chloride channels.
Collapse
Affiliation(s)
- Jared Morse
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danna Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Serena Mei
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danielle Whitham
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Colby Hladun
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Costel C. Darie
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Modi Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - KaHo Leung
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| |
Collapse
|
47
|
Li Q, Wu P, Du Q, Hanif U, Hu H, Li K. cGAS-STING, an important signaling pathway in diseases and their therapy. MedComm (Beijing) 2024; 5:e511. [PMID: 38525112 PMCID: PMC10960729 DOI: 10.1002/mco2.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Since cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway was discovered in 2013, great progress has been made to elucidate the origin, function, and regulating mechanism of cGAS-STING signaling pathway in the past decade. Meanwhile, the triggering and transduction mechanisms have been continuously illuminated. cGAS-STING plays a key role in human diseases, particularly DNA-triggered inflammatory diseases, making it a potentially effective therapeutic target for inflammation-related diseases. Here, we aim to summarize the ancient origin of the cGAS-STING defense mechanism, as well as the triggers, transduction, and regulating mechanisms of the cGAS-STING. We will also focus on the important roles of cGAS-STING signal under pathological conditions, such as infections, cancers, autoimmune diseases, neurological diseases, and visceral inflammations, and review the progress in drug development targeting cGAS-STING signaling pathway. The main directions and potential obstacles in the regulating mechanism research and therapeutic drug development of the cGAS-STING signaling pathway for inflammatory diseases and cancers will be discussed. These research advancements expand our understanding of cGAS-STING, provide a theoretical basis for further exploration of the roles of cGAS-STING in diseases, and open up new strategies for targeting cGAS-STING as a promising therapeutic intervention in multiple diseases.
Collapse
Affiliation(s)
- Qijie Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ping Wu
- Department of Occupational DiseasesThe Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital)ChengduSichuanChina
| | - Qiujing Du
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ullah Hanif
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Hongbo Hu
- Center for Immunology and HematologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ka Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| |
Collapse
|
48
|
Liu TT, Sun HF, Han YX, Zhan Y, Jiang JD. The role of inflammation in silicosis. Front Pharmacol 2024; 15:1362509. [PMID: 38515835 PMCID: PMC10955140 DOI: 10.3389/fphar.2024.1362509] [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: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Silicosis is a chronic illness marked by diffuse fibrosis in lung tissue resulting from continuous exposure to SiO2-rich dust in the workplace. The onset and progression of silicosis is a complicated and poorly understood pathological process involving numerous cells and molecules. However, silicosis poses a severe threat to public health in developing countries, where it is the most prevalent occupational disease. There is convincing evidence supporting that innate and adaptive immune cells, as well as their cytokines, play a significant role in the development of silicosis. In this review, we describe the roles of immune cells and cytokines in silicosis, and summarize current knowledge on several important inflammatory signaling pathways associated with the disease, aiming to provide novel targets and strategies for the treatment of silicosis-related inflammation.
Collapse
Affiliation(s)
| | | | | | - Yun Zhan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
49
|
Xiong Q, Tian X, Xu C, Ma B, Li W, Xia Y, Liu W, Sun B, Ru Q, Shu X. Mediation of PM2.5-induced cytotoxicity: the role of P2X7 receptor in NR8383 cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:1602-1614. [PMID: 37394938 DOI: 10.1080/09603123.2023.2230920] [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: 02/16/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
Ambient fine particulate matter (PM2.5) is a threat to public health. The P2 X 7purinergic receptor (P2X7R) is a modulator that responds to inflammation. Yet the role of P2X7R in the mediation of PM2.5-induced pulmonary cytotoxicity is rarely investigated. In this study, the expression of P2X7R and its effect on cell viability, oxidative damage, apoptosis, mitochondrial dysfunction and underlying mechanism following PM2.5 treatment in rat alveolar macrophages (NR8383) were analyzed. The outcome indicated that PM2.5 exposure significantly increased the expression of P2X7R, while P2X7R antagonist oATP markedly alleviate the production of reactive oxygen species (ROS), Nitrite Oxidation (NO), mitochondrial membrane potential, apoptosis rate, and release of inflammatory cytokines. In contrast, P2X7 agonist BzATP showed opposite effect in PM2.5-treated NR8383 cells. Therefore, these results demonstrated that P2X7R participated in PM2.5-induced pulmonary toxicity, while the blockade of P2X7R is a promising therapeutic approach of treating PM2.5-induced lung diseases.
Collapse
Affiliation(s)
- Qi Xiong
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Xiang Tian
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Congyue Xu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Baomiao Ma
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Wenshuang Li
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Yiyuan Xia
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Wei Liu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Binlian Sun
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| | - Qin Ru
- School of Health and Physical Education, Jianghan University, Wuhan City, Hubei Province, China
| | - Xiji Shu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan City, Hubei Province, China
| |
Collapse
|
50
|
Morin L, Lecureur V, Lescoat A. Results from omic approaches in rat or mouse models exposed to inhaled crystalline silica: a systematic review. Part Fibre Toxicol 2024; 21:10. [PMID: 38429797 PMCID: PMC10905840 DOI: 10.1186/s12989-024-00573-x] [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: 11/23/2022] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Crystalline silica (cSiO2) is a mineral found in rocks; workers from the construction or denim industries are particularly exposed to cSiO2 through inhalation. cSiO2 inhalation increases the risk of silicosis and systemic autoimmune diseases. Inhaled cSiO2 microparticles can reach the alveoli where they induce inflammation, cell death, auto-immunity and fibrosis but the specific molecular pathways involved in these cSiO2 effects remain unclear. This systematic review aims to provide a comprehensive state of the art on omic approaches and exposure models used to study the effects of inhaled cSiO2 in mice and rats and to highlight key results from omic data in rodents also validated in human. METHODS The protocol of systematic review follows PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Eligible articles were identified in PubMed, Embase and Web of Science. The search strategy included original articles published after 1990 and written in English which included mouse or rat models exposed to cSiO2 and utilized omic approaches to identify pathways modulated by cSiO2. Data were extracted and quality assessment was based on the SYRCLE's Risk of Bias tool for animal studies. RESULTS Rats and male rodents were the more used models while female rodents and autoimmune prone models were less studied. Exposure of animals were both acute and chronic and the timing of outcome measurement through omics approaches were homogeneously distributed. Transcriptomic techniques were more commonly performed while proteomic, metabolomic and single-cell omic methods were less utilized. Immunity and inflammation were the main domains modified by cSiO2 exposure in lungs of mice and rats. Less than 20% of the results obtained in rodents were finally verified in humans. CONCLUSION Omic technics offer new insights on the effects of cSiO2 exposure in mice and rats although the majority of data still need to be validated in humans. Autoimmune prone model should be better characterised and systemic effects of cSiO2 need to be further studied to better understand cSiO2-induced autoimmunity. Single-cell omics should be performed to inform on pathological processes induced by cSiO2 exposure.
Collapse
Affiliation(s)
- Laura Morin
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France
| | - Valérie Lecureur
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France.
| | - Alain Lescoat
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en sante, environnement et travail), UMR_S 1085, 35000, Rennes, France
- Department of Internal Medicine, Rennes University Hospital, 35000, Rennes, France
| |
Collapse
|