151
|
Gong W, Lu L, Zhou Y, Liu J, Ma H, Fu L, Huang S, Zhang Y, Zhang A, Jia Z. The novel STING antagonist H151 ameliorates cisplatin-induced acute kidney injury and mitochondrial dysfunction. Am J Physiol Renal Physiol 2021; 320:F608-F616. [PMID: 33615891 DOI: 10.1152/ajprenal.00554.2020] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stimulator of interferon genes (STING) is an important adaptor in cytosolic DNA-sensing pathways. A recent study found that the deletion of STING ameliorated cisplatin-induced acute kidney injury (AKI), suggesting that STING could serve as a potential target for AKI therapy. Up to now, a series of small-molecule STING inhibitors/antagonists have been identified. However, none of the research was performed to explore the role of human STING inhibitors in AKI. Here, we investigated the effect of a newly generated covalent antagonist, H151, which targets both human and murine STING, in cisplatin-induced AKI. We found that H151 treatment significantly ameliorated cisplatin-induced kidney injury as shown by the improvement of renal function, kidney morphology, and renal inflammation. In addition, tubular cell apoptosis and increased renal tubular injury marker neutrophil gelatinase-associated lipocalin induced by cisplatin were also effectively attenuated in H151-treated mice. Moreover, the mitochondrial injury caused by cisplatin was also reversed as evidenced by improved mitochondrial morphology, restored mitochondrial DNA content, and reversed mitochondrial gene expression. Finally, we observed enhanced mitochondrial DNA levels in the plasma of patients receiving platinum-based chemotherapy compared with healthy controls, which could potentially activate STING signaling. Taken together, these findings suggested that H151 could be a potential therapeutic agent for treating AKI possibly through inhibiting STING-mediated inflammation and mitochondrial injury.NEW & NOTEWORTHY Although various stimulator of interferon genes (STING) inhibitors have been identified, no research was performed to investigate the role of human STING inhibitors in AKI. Here, we evaluated the effect of H151 targeting both human and murine STING on cisplatin-induced AKI and observed a protection against renal injury possibly through ameliorating inflammation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Wei Gong
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Lu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Zhou
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiaye Liu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Haoyang Ma
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lvhan Fu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
152
|
Cavagnero KJ, Badrani JH, Naji LH, Amadeo MB, Leng AS, Lacasa LD, Strohm AN, Renusch SR, Gasparian SS, Doherty TA. Cyclic-di-GMP Induces STING-Dependent ILC2 to ILC1 Shift During Innate Type 2 Lung Inflammation. Front Immunol 2021; 12:618807. [PMID: 33679760 PMCID: PMC7935536 DOI: 10.3389/fimmu.2021.618807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Type 2 inflammation is found in most forms of asthma, which may co-exist with recurrent viral infections, bacterial colonization, and host cell death. These processes drive the accumulation of intracellular cyclic-di-nucleotides such as cyclic-di-GMP (CDG). Group 2 innate lymphoid cells (ILC2s) are critical drivers of type 2 lung inflammation during fungal allergen exposure in mice; however, it is unclear how CDG regulates lung ILC responses during lung inflammation. Here, we show that intranasal CDG induced early airway type 1 interferon (IFN) production and dramatically suppressed CD127+ST2+ ILC2s and type 2 lung inflammation during Alternaria and IL-33 exposure. Further, CD127-ST2-Thy1.2+ lung ILCs, which showed a transcriptomic signature consistent with ILC1s, were expanded and activated by CDG combined with either Alternaria or IL-33. CDG-mediated suppression of type 2 inflammation occurred independent of IL-18R, IL-12, and STAT6 but required the stimulator of interferon genes (STING) and type 1 IFN signaling. Thus, CDG potently suppresses ILC2-driven lung inflammation and promotes ILC1 responses. These results suggest potential therapeutic modulation of STING to suppress type 2 inflammation and/or increase anti-viral responses during respiratory infections.
Collapse
Affiliation(s)
- Kellen J. Cavagnero
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Dermatology, University of California, San Diego, La Jolla, CA, United States
| | - Jana H. Badrani
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Luay H. Naji
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michael B. Amadeo
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Anthea S. Leng
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Lee Diego Lacasa
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Allyssa N. Strohm
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Samantha R. Renusch
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Suzanna S. Gasparian
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Taylor A. Doherty
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- Veterans Affairs San Diego Health Care System, La Jolla, CA, United States
| |
Collapse
|
153
|
Cao Z, Li S, Liu H, Li W, Sun Y, Li Z, Jia M, Su M. The UFSP2/UFMylation Pathway Is Involved in Silica-Induced Pulmonary Injury. DNA Cell Biol 2021; 40:589-594. [PMID: 33600261 DOI: 10.1089/dna.2020.6421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Silicosis is an irreversible occupational pulmonary disease that is characterized as progressed pulmonary fibrosis. In this study, we investigated the changes of UFSP2 and the related UFMylation in silica-induced pulmonary injury mice models. The experimental silicosis models were prepared by intratracheal injection of silica particles, and the lung samples were harvested at the first or the seventh day after treatment. We found that the UFSP2 expression in the 1-day models was comparable, whereas it was upregulated in the 7-day models. Consistently, the UFMylation in the lung tissues of the 7-day models was activated. In addition, we observed the CADM2, an adhesion molecule, was reported to associate with epithelial-mesenchymal transition, was upregulated in the lungs of 7-day models. In contrast, it remained comparable in the 1-day models. Our data indicated that the UFSP2/UFMylation pathway and the CADM2 might be involved in the silica-induced pulmonary injury.
Collapse
Affiliation(s)
- Zhenju Cao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Shanshan Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Hangqi Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Wendi Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Yi Sun
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Zichen Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Ming Su
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
154
|
Frémond ML, Crow YJ. STING-Mediated Lung Inflammation and Beyond. J Clin Immunol 2021; 41:501-514. [PMID: 33532887 DOI: 10.1007/s10875-021-00974-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Mendelian autoinflammatory diseases characterized by constitutive activation of the type I interferon pathway, the so-called type I interferonopathies, constitute a rapidly expanding group of inborn errors of immunity. Among the type I interferonopathies, STING-associated vasculopathy with onset in infancy (SAVI) and COPA syndrome were described in the last 6 years, both manifesting a major inflammatory lung component associated with significant morbidity and increased mortality. There is striking clinical and histopathological overlap between SAVI and COPA syndrome, although distinct features are also present. Of note, there is a remarkably high frequency of clinical non-penetrance among individuals harboring pathogenic COPA mutations. SAVI is caused by, principally heterozygous, gain-of-function mutations in STING1 (previously referred to as TMEM173) encoding STING, a key adaptor of the interferon signaling pathway induced by DNA. COPA syndrome results from heterozygous dominant-negative mutations in the coatomer protein subunit alpha, forming part of a complex involved in intracellular cargo protein transport between the Golgi and the endoplasmic reticulum (ER). Of importance, a role for COPA in regulating the trafficking of STING, an ER-resident protein which translocates to the Golgi during the process of its activation, was recently defined, thereby possibly explaining some aspects of the phenotypic overlap between SAVI and COPA syndrome. Here, we review the expanding phenotype of these diseases, highlighting common as well as specific features, and recent advances in our understanding of STING biology that have informed therapeutic decision-making in both conditions. Beyond these rare Mendelian disorders, DNA sensing through STING is likely relevant to the pathology of several diseases associated with lung inflammation, including systemic lupus erythematosus, dermatomyositis, environmental toxin exposure, and viral infection.
Collapse
Affiliation(s)
- Marie-Louise Frémond
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 boulevard du Montparnasse, F-75015, Paris, France. .,Paediatric Haematology-Immunology and Rheumatology Unit, AP-HP, Necker Hospital, F-75015, Paris, France.
| | - Yanick J Crow
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 boulevard du Montparnasse, F-75015, Paris, France.,Centre for Genomic and Experimental Medicine, Medical Research Council Institute of Genetics and Molecular Medicine, Edinburgh, UK
| |
Collapse
|
155
|
Orvain C, Lin YL, Jean-Louis F, Hocini H, Hersant B, Bennasser Y, Ortonne N, Hotz C, Wolkenstein P, Boniotto M, Tisserand P, Lefebvre C, Lelièvre JD, Benkirane M, Pasero P, Lévy Y, Hüe S. Hair follicle stem cell replication stress drives IFI16/STING-dependent inflammation in hidradenitis suppurativa. J Clin Invest 2021; 130:3777-3790. [PMID: 32240121 DOI: 10.1172/jci131180] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 03/31/2020] [Indexed: 12/28/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic, relapsing, inflammatory skin disease. HS appears to be a primary abnormality in the pilosebaceous-apocrine unit. In this work, we characterized hair follicle stem cells (HFSCs) isolated from HS patients and more precisely the outer root sheath cells (ORSCs). We showed that hair follicle cells from HS patients had an increased number of proliferating progenitor cells and lost quiescent stem cells. Remarkably, we also showed that the progression of replication forks was altered in ORSCs from hair follicles of HS patients, leading to activation of the ATR/CHK1 pathway. These alterations were associated with an increased number of micronuclei and with the presence of cytoplasmic ssDNA, leading to the activation of the IFI16/STING pathway and the production of type I IFNs. This mechanistic analysis of the etiology of HS in the HFSC compartment establishes a formal link between genetic predisposition and skin inflammation observed in HS.
Collapse
Affiliation(s)
- Cindy Orvain
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Yea-Lih Lin
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Francette Jean-Louis
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Hakim Hocini
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Barbara Hersant
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Yamina Bennasser
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Nicolas Ortonne
- Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'anatomopathologie
| | - Claire Hotz
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service de Dermatologie
| | - Pierre Wolkenstein
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service de Dermatologie
| | - Michele Boniotto
- INSERM U955, Equipe 16, Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Pascaline Tisserand
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Cécile Lefebvre
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Jean-Daniel Lelièvre
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Clinique, and
| | - Monsef Benkirane
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Philippe Pasero
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Yves Lévy
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Clinique, and
| | - Sophie Hüe
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Biologique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
156
|
Savigny F, Schricke C, Lacerda-Queiroz N, Meda M, Nascimento M, Huot-Marchand S, Da Gama Monteiro F, Ryffel B, Gombault A, Le Bert M, Couillin I, Riteau N. Protective Role of the Nucleic Acid Sensor STING in Pulmonary Fibrosis. Front Immunol 2021; 11:588799. [PMID: 33488589 PMCID: PMC7820752 DOI: 10.3389/fimmu.2020.588799] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and severe type of interstitial lung disease for which current treatments display limited efficacy. IPF is largely driven by host-derived danger signals released upon recurrent local tissue damage. Here we explored the roles of self-DNA and stimulator of interferon genes (STING), a protein belonging to an intracellular DNA sensing pathway that leads to type I and/or type III interferon (IFN) production upon activation. Using a mouse model of IPF, we report that STING deficiency leads to exacerbated pulmonary fibrosis with increased collagen deposition in the lungs and excessive remodeling factors expression. We further show that STING-mediated protection does not rely on type I IFN signaling nor on IL-17A or TGF-β modulation but is associated with dysregulated neutrophils. Together, our data support an unprecedented immunoregulatory function of STING in lung fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Isabelle Couillin
- Experimental and Molecular Immunology and Neurogenetics Laboratory (INEM), CNRS Orleans (UMR7355) and University of Orleans, Orleans, France
| | - Nicolas Riteau
- Experimental and Molecular Immunology and Neurogenetics Laboratory (INEM), CNRS Orleans (UMR7355) and University of Orleans, Orleans, France
| |
Collapse
|
157
|
Huang R, Bai C, Liu X, Zhou Y, Hu S, Li D, Xiang J, Chen J, Zhou P. The p53/RMRP/miR122 signaling loop promotes epithelial-mesenchymal transition during the development of silica-induced lung fibrosis by activating the notch pathway. CHEMOSPHERE 2021; 263:128133. [PMID: 33297121 DOI: 10.1016/j.chemosphere.2020.128133] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 07/13/2020] [Accepted: 08/24/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Understanding the roles of long noncoding RNAs (lncRNAs) in EMT would help with establishing novel avenues for further uncovering the mechanisms of lung fibrosis and identifying preventative and therapeutic targets. This study aimed to identify silica-induced specific lncRNAs and investigate the feedback loop regulation among their upstream and downstream genes. METHODS AND MATERIALS A microarray assay, quantitative real-time polymerase chain reaction and Western blot analysis dual-luciferase reporter gene activity and chromatin immunoprecipitation assays were used. Moreover, a silica-induced lung fibrosis mouse model was used to verify the roles of the lncRNAs. RESULTS Following silica exposure, both RNA component of mitochondrial RNA processing endoribonuclease (RMRP) and p53 were significantly upregulated during the EMT. The upregulation of p53 upon silica exposure activated RMRP expression, which promoted the EMT. When RMRP is overexpressed, additional RMRP acts as a sponge to bind to miR122, thus decreasing miR122 levels. Using microarrays, miR122 was identified as a potential upstream regulator of p53. This relationship was also verified using the dual-luciferase reporter gene. Hence, decreased miR122 levels result in an increase in p53 activity. More importantly, RMRP promotes the transcription of Notch 1, which, in turn, results in Notch pathway activation. We show that the p53/RMRP/miR122 pathway creates a positive feedback loop that promotes EMT progress by activating the Notch signaling pathway. CONCLUSION Our data indicated that p53/RMRP/miR122 feedback loop might contribute to the EMT development by activating Notch pathway, which provides new sight into understanding of the complex network regulating silica-induced lung fibrosis.
Collapse
Affiliation(s)
- Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| | - Chenjun Bai
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, 100850, China.
| | - Xiaodan Liu
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, 100850, China.
| | - Yao Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| | - Sai Hu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| | - Decheng Li
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| | - Jing Xiang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, 410078, China.
| | - Jihua Chen
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, 410078, Changsha, 63455553, China.
| | - Pingkun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, 100850, China; Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory, School of Public Health, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
158
|
Abstract
The cGAS-STING signalling pathway has emerged as a key mediator of inflammation in the settings of infection, cellular stress and tissue damage. Underlying this broad involvement of the cGAS-STING pathway is its capacity to sense and regulate the cellular response towards microbial and host-derived DNAs, which serve as ubiquitous danger-associated molecules. Insights into the structural and molecular biology of the cGAS-STING pathway have enabled the development of selective small-molecule inhibitors with the potential to target the cGAS-STING axis in a number of inflammatory diseases in humans. Here, we outline the principal elements of the cGAS-STING signalling cascade and discuss the general mechanisms underlying the association of cGAS-STING activity with various autoinflammatory, autoimmune and degenerative diseases. Finally, we outline the chemical nature of recently developed cGAS and STING antagonists and summarize their potential clinical applications.
Collapse
|
159
|
Bryant AJ, Pham A, Gogoi H, Mitchell CR, Pais F, Jin L. The Third Man: DNA sensing as espionage in pulmonary vascular health and disease. Pulm Circ 2021; 11:2045894021996574. [PMID: 33738095 PMCID: PMC7934053 DOI: 10.1177/2045894021996574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
For as long as nucleic acids have been utilized to vertically and horizontally transfer genetic material, living organisms have had to develop methods of recognizing cytosolic DNA as either pathogenic (microbial invasion) or physiologic (mitosis and cellular proliferation). Derangement in key signaling molecules involved in these pathways of DNA sensing result in a family of diseases labeled interferonopathies. An interferonopathy, characterized by constitutive expression of type I interferons, ultimately manifests as severe autoimmune disease at a young age. Afflicted patients present with a constellation of immune-mediated conditions, including primary lung manifestations such as pulmonary fibrosis and pulmonary hypertension. The latter condition is especially interesting in light of the known role that DNA damage plays in a variety of types of inherited and induced pulmonary hypertension, with free DNA detection elevated in the circulation of affected individuals. While little is known regarding the role of cytosolic DNA sensing in development of pulmonary vascular disease, exciting new research in the related fields of immunology and oncology potentially sheds light on future areas of fruitful exploration. As such, the goal of this review is to summarize the state of the field of nucleic acid sensing, extrapolating common shared pathways that parallel our knowledge of pulmonary hypertension, in a molecular and cell-specific manner. Principles of DNA sensing related to known pulmonary injury inducing stimuli are also evaluated, in addition to potential therapeutic targets. Finally, future directions in pulmonary hypertension research and treatments will be briefly discussed.
Collapse
Affiliation(s)
- Andrew J. Bryant
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Ann Pham
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Himanshu Gogoi
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Carly R. Mitchell
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Faye Pais
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Lei Jin
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| |
Collapse
|
160
|
Karataş M, Büyükşekerci M, Gündüzöz M, Özakıncı G, Öziş TN, Gök G, Neşelioğlu S, Erel Ö. Alteration of thiol disulfide homeostasis and ischemia-modified albumin levels as indicators of oxidative status in patients with silicosis. Toxicol Ind Health 2020; 37:38-46. [PMID: 33305688 DOI: 10.1177/0748233720977987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to evaluate the oxidative status in patients with silicosis by detecting dynamic thiol disulfide homeostasis (TDH), ischemia-modified albumin level (IMA) catalase (CAT) activity, and the correlation of these markers with pulmonary function tests. Male ceramic workers with silicosis (n = 91) and healthy individuals (n = 47) were recruited for the study. Radiographic abnormalities of pneumoconiosis were classified into three profusion categories (categories 1, 2, and 3), and patients with silicosis, those with category 1, were defined as group 1 and those with category 2 or 3 were defined as group 2. Plasma levels of native thiol (NT), total thiol (TT), disulfide (Ds), IMA, and CAT activities were determined. Pulmonary function tests of groups were compared. NT, TT, and NT/TT ratios were significantly lower in groups 1 and 2 than the control group (p < 0.05). These did not differ between patients with silicosis (groups 1 and 2) and control group (p = 0.421). Ds/NT and Ds/TT ratios were significantly higher in group 2 than the control group (p < 0.05). NT, TT, and Ds did not differ significantly between groups 1 and 2. The oxidant biomarker IMA was higher (p < 0.001), and the antioxidant parameters albumin and CAT were lower in groups 1 and 2 (p < 0.001) compared with the control group. The mean FEV1act, FVCact, forced expiratory volume in 1 second/forced vital capacity (%), and value of 25-75 percent maximum expiratory flow were significantly lower in groups 1 and 2 than control group. We have used a novel colorimetric method to assess TDH in patients with silicosis. Alteration of plasma thiol/disulfide homeostasis and IMA levels might be novel indicators of oxidative stress in silicosis.
Collapse
Affiliation(s)
- Mevlüt Karataş
- Department of Chest Diseases, Occupational and Environmental Diseases Hospital, Ankara, Turkey
| | - Murat Büyükşekerci
- Department of Pharmacology, Occupational and Environmental Diseases Hospital, Ankara, Turkey
| | - Meşide Gündüzöz
- Department of Family Medicine, Occupational and Environmental Diseases Hospital, Ankara, Turkey
| | - Gökhan Özakıncı
- Department of Public Health, Occupational and Environmental Diseases Hospital, Ankara, Turkey
| | - Türkan Nadir Öziş
- Department of Chest Diseases, Occupational and Environmental Diseases Hospital, Ankara, Turkey.,Department of Biochemistry, 442146Yıldırım Beyazıt University, Ankara, Turkey
| | - Gamze Gök
- Department of Biochemistry, 442146Yıldırım Beyazıt University, Ankara, Turkey
| | - Salim Neşelioğlu
- Department of Biochemistry, 442146Yıldırım Beyazıt University, Ankara, Turkey
| | - Özcan Erel
- Department of Biochemistry, 442146Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
161
|
Lescoat A, Ballerie A, Lecureur V, Belhomme N, Cazalets C, Jouneau S, Paris C, Menéndez-Navarro A, Rosental PA, Jégo P, Cavalin C. The neglected association of crystalline silica exposure and systemic sclerosis. Rheumatology (Oxford) 2020; 59:3587-3588. [PMID: 33020830 DOI: 10.1093/rheumatology/keaa638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Alain Lescoat
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France.,Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Alice Ballerie
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France.,Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Valérie Lecureur
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Nicolas Belhomme
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Claire Cazalets
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Stéphane Jouneau
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France.,Department of Respiratory Diseases, Rennes University Hospital, Rennes, France
| | - Christophe Paris
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France.,INSERM, EHESP, Institut de Recherche en Santé, Department of Occupational Medicine, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | | | | | - Patrick Jégo
- INSERM, EHESP, Institut de Recherche en Santé, Université de Rennes, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Catherine Cavalin
- Interdisciplinary Research Institute for the Social Sciences, National Centre for Scientific Research-National Institute for Agricultural Research, France
| |
Collapse
|
162
|
Togbe D, Benmerzoug S, Jacobs M, Ryffel B, Quesniaux VFJ. Silica-related diseases in the modern world: A role for self-DNA sensing in lung inflammatory diseases. Allergy 2020; 75:3009-3010. [PMID: 33155702 DOI: 10.1111/all.14463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/28/2022]
Affiliation(s)
| | - Sulayman Benmerzoug
- Experimental and Molecular Immunology and Neurogenetics UMR7355 CNRS and University of Orleans Lausanne France
- Department of Urology Urology Research Unit CHUV Lausanne Switzerland
| | - Muazzam Jacobs
- Institute of Infectious Disease and Molecular Medicine University of Cape Town Cape Town South Africa
| | - Bernhard Ryffel
- Artimmune SAS Orleans‐Cedex 2 France
- Institute of Infectious Disease and Molecular Medicine University of Cape Town Cape Town South Africa
| | - Valerie F. J. Quesniaux
- Experimental and Molecular Immunology and Neurogenetics UMR7355 CNRS and University of Orleans Lausanne France
| |
Collapse
|
163
|
Chambers DC, Hoy RF. Reply to correspondence: Silica-related diseases in the modern world: A role for self-DNA sensing in lung inflammatory. Allergy 2020; 75:3011. [PMID: 33155703 DOI: 10.1111/all.14367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Daniel C. Chambers
- The Prince Charles Hospital Brisbane and The University of Queensland Brisbane Qld Australia
| | - Ryan Francis Hoy
- Department of Epidemiology and PreventiveMedicine School of Public Health Monash University Melbourne Vic Australia
| |
Collapse
|
164
|
Rajasinghe LD, Chauhan PS, Wierenga KA, Evered AO, Harris SN, Bates MA, Gavrilin MA, Pestka JJ. Omega-3 Docosahexaenoic Acid (DHA) Impedes Silica-Induced Macrophage Corpse Accumulation by Attenuating Cell Death and Potentiating Efferocytosis. Front Immunol 2020; 11:2179. [PMID: 33123123 PMCID: PMC7573148 DOI: 10.3389/fimmu.2020.02179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
Airway exposure of lupus-prone NZBWF1 mice to crystalline silica (cSiO2), a known trigger of human autoimmune disease, elicits sterile inflammation and alveolar macrophage death in the lung that, in turn, induces early autoimmune onset and accelerates lupus progression to fatal glomerulonephritis. Dietary supplementation with docosahexaenoic acid (DHA), a marine ω-3 polyunsaturated fatty acid (PUFA), markedly ameliorates cSiO2-triggered pulmonary, systemic, and renal manifestations of lupus. Here, we tested the hypothesis that DHA influences both cSiO2-induced death and efferocytotic clearance of resultant cell corpses using three murine macrophage models: (i) primary alveolar macrophages (AM) isolated from NZBWF1 mice; (ii) self-renewing AM-like Max Planck Institute (MPI) cells isolated from fetuses of C57BL/6 mice, and (iii) RAW 264.7 murine macrophages, a virus-transformed cell line derived from BALB/c mice stably transfected with the inflammasome adaptor protein ASC (RAW-ASC). Incubation with cSiO2 at 25 and 50 μg/ml for 6 h was found to dose-dependently induce cell death (p < 0.05) in all three models as determined by both acridine orange/propidium iodide staining and release of lactate dehydrogenase into cell culture supernatant. Pre-incubation with DHA at a physiologically relevant concentration (25 μM) significantly reduced cSiO2-induced death (p < 0.05) in all three models. Cell death induction by cSiO2 alone and its suppression by DHA were primarily associated with caspase-3/7 activation, suggestive of apoptosis, in AM, MPI, and RAW-ASC cells. Fluorescence microscopy revealed that all three macrophage models were similarly capable of efferocytosing RAW-ASC target cell corpses. Furthermore, MPI effector cells could likewise engulf RAW-ASC target cell corpses elicited by treatment with staurosporine (apoptosis), LPS, and nigericin (pyroptosis), or cSiO2. Pre-incubation of RAW-ASC target cells with 25 μM DHA prior to death induced by these agents significantly enhanced their efferocytosis (p < 0.05) by MPI effector cells. In contrast, pre-incubating MPI effector cells with DHA did not affect engulfment of RAW-ASC target cells pre-incubated with vehicle. Taken together, these findings indicate that DHA at a physiologically relevant concentration was capable of attenuating macrophage death and could potentiate efferocytosis, with the net effect of reducing accumulation of cell corpses capable of eliciting autoimmunity.
Collapse
Affiliation(s)
- Lichchavi D Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S Chauhan
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Kathryn A Wierenga
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Augustus O Evered
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Shamya N Harris
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Melissa A Bates
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Mikhail A Gavrilin
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH, United States
| | - James J Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
165
|
Unno H, Arae K, Matsuda A, Ikutani M, Tamari M, Motomura K, Toyama S, Suto H, Okumura K, Matsuda A, Morita H, Sudo K, Saito H, Matsumoto K, Nakae S. Critical role of IL-33, but not IL-25 or TSLP, in silica crystal-mediated exacerbation of allergic airway eosinophilia. Biochem Biophys Res Commun 2020; 533:493-500. [PMID: 32977946 DOI: 10.1016/j.bbrc.2020.09.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 11/17/2022]
Abstract
Silica crystals (silica), which are a major mineral component of volcanic ash and desert dust, contribute to the pathogenesis of pulmonary disorders such as asthma and fibrosis. Although administration of silica or sand dust to rodents exacerbates development of ovalbumin-induced or house dust mite-induced asthma-like airway inflammation, the detailed mechanisms remain unclear. Here, using murine models, we found that silica can induce IL-33 expression in pulmonary epithelial cells. IL-33, but not IL-25 or TSLP, and type 2 cytokines such as IL-5 and IL-13 were critically involved in silica's exacerbation of OVA-induced airway eosinophilia in mice. Innate lymphoid cells (ILCs), but not T, B or NKT cells, were also involved in the setting. Moreover, a scavenger receptor that recognized silica was important for silica's exacerbating effect. These observations suggest that IL-33 induced in epithelial cells by silica activates ILCs to produce IL-5 and/or IL-13, contributing to silica's exacerbation of OVA-induced airway eosinophilia in mice. Our findings provide new insight into the underlying mechanisms of exacerbation of pulmonary disorders such as asthma following inhalation of silica-containing materials such as volcanic ash and desert dust.
Collapse
Affiliation(s)
- Hirotoshi Unno
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, 105-8461, Japan; Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Ken Arae
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan; Department of Immunology, Faculty of Health Sciences, Kyorin University, Tokyo, 181-8612, Japan
| | - Akira Matsuda
- Laboratory of Ocular Atopic Diseases, Department of Ophthalmology, Juntendo University School, Tokyo, 113-8412, Japan
| | - Masashi Ikutani
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan
| | - Masato Tamari
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, 105-8461, Japan; Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Sumika Toyama
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hajime Suto
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo, 113-8412, Japan
| | - Ko Okumura
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo, 113-8412, Japan
| | - Akio Matsuda
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Katsuko Sudo
- Animal Research Center, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan; Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama, 332-0012, Japan.
| |
Collapse
|
166
|
Teng F, Li M, Yu J. Radiation recall pneumonitis induced by PD-1/PD-L1 blockades: mechanisms and therapeutic implications. BMC Med 2020; 18:275. [PMID: 32943072 PMCID: PMC7499987 DOI: 10.1186/s12916-020-01718-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The synergistic effect of radiotherapy (RT) in combination with immunotherapy has been shown in several clinical trials and case reports. The overlapping pulmonary toxicity induced by thoracic RT and programmed death 1/programmed death ligand-1 (PD-1/PD-L1) blockades is an important issue of clinical investigation in combination treatment. Thus far, the underlying mechanism of this toxicity remains largely unknown. MAIN TEXT In this review, we discuss the unique pattern of radiation recall pneumonitis (RRP) induced by PD-1 blockade. The clinical presentation is different from common radiation pneumonitis (RP) or RRP induced by cytotoxic drugs. The immune checkpoint inhibitors may evoke an inflammatory reaction in patients' previously irradiated fields, with infiltrating lymphocytes and potential involvement of related cytokines. All RRP patients have showed durable response to anti-PD-1/PD-L1. RRP is manageable; however, interruption of checkpoint blockades is necessary and immunosuppressive treatment should be started immediately. Further analyses of the predictive factors, including RT dosimetric parameters, tumor-infiltrating lymphocytes (TILs), and PD-L1 expression, are needed given the wide use of immune checkpoint inhibitors and high mortality from lung toxicity with the combination treatment. CONCLUSION Immune checkpoint inhibitors may evoke an RRP in the patients' previously irradiated fields. Interactions between immune checkpoint inhibitors and radiotherapy should be studied further.
Collapse
Affiliation(s)
- Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, China
| | - Min Li
- Department of Surgery, Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, China.
| |
Collapse
|
167
|
Piperno GM, Naseem A, Silvestrelli G, Amadio R, Caronni N, Cervantes-Luevano KE, Liv N, Klumperman J, Colliva A, Ali H, Graziano F, Benaroch P, Haecker H, Hanna RN, Benvenuti F. Wiskott-Aldrich syndrome protein restricts cGAS/STING activation by dsDNA immune complexes. JCI Insight 2020; 5:132857. [PMID: 32721945 PMCID: PMC7526445 DOI: 10.1172/jci.insight.132857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 07/16/2020] [Indexed: 01/06/2023] Open
Abstract
Dysregulated sensing of self-nucleic acid is a leading cause of autoimmunity in multifactorial and monogenic diseases. Mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in immune cells, cause autoimmune manifestations and increased production of type I IFNs by innate cells. Here we show that immune complexes of self-DNA and autoantibodies (DNA-ICs) contribute to elevated IFN levels via activation of the cGAS/STING pathway of cytosolic sensing. Mechanistically, lack of endosomal F-actin nucleation by WASp caused a delay in endolysosomal maturation and prolonged the transit time of ingested DNA-ICs. Stalling in maturation-defective organelles facilitated leakage of DNA-ICs into the cytosol, promoting activation of the TBK1/STING pathway. Genetic deletion of STING and STING and cGAS chemical inhibitors abolished IFN production and rescued systemic activation of IFN-stimulated genes in vivo. These data unveil the contribution of cytosolic self-nucleic acid sensing in WAS and underscore the importance of WASp-mediated endosomal actin remodeling in preventing innate activation.
Collapse
Affiliation(s)
| | - Asma Naseem
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Giulia Silvestrelli
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Roberto Amadio
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nicoletta Caronni
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | | | - Nalan Liv
- Section Cell Biology, Center for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Andrea Colliva
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Hashim Ali
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Francesca Graziano
- Institute Curie Laboratoire Immunité et Cancer - INSERM U932 Transport Intracellulaire et Immunité, Paris, France
| | - Philippe Benaroch
- Institute Curie Laboratoire Immunité et Cancer - INSERM U932 Transport Intracellulaire et Immunité, Paris, France
| | - Hans Haecker
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richard N Hanna
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Federica Benvenuti
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
168
|
Wiegman CH, Li F, Ryffel B, Togbe D, Chung KF. Oxidative Stress in Ozone-Induced Chronic Lung Inflammation and Emphysema: A Facet of Chronic Obstructive Pulmonary Disease. Front Immunol 2020; 11:1957. [PMID: 32983127 PMCID: PMC7492639 DOI: 10.3389/fimmu.2020.01957] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD) caused by cigarette smoke and characterized by chronic inflammation, alveolar destruction (emphysema) and bronchiolar obstruction. Ozone is a gaseous constituent of urban air pollution resulting from photochemical interaction of air pollutants such as nitrogen oxide and organic compounds. While acute exposure to ozone induces airway hyperreactivity and neutrophilic inflammation, chronic ozone exposure in mice causes activation of oxidative pathways resulting in cell death and a chronic bronchial inflammation with emphysema, mimicking cigarette smoke-induced COPD. Therefore, the chronic exposure to ozone has become a model for studying COPD. We review recent data on mechanisms of ozone induced lung disease focusing on pathways causing chronic respiratory epithelial cell injury, cell death, alveolar destruction, and tissue remodeling associated with the development of chronic inflammation and AHR. The initial oxidant insult may result from direct effects on the integrity of membranes and organelles of exposed epithelial cells in the airways causing a stress response with the release of mitochondrial reactive oxygen species (ROS), DNA, and proteases. Mitochondrial ROS and mitochondrial DNA activate NLRP3 inflammasome and the DNA sensors cGAS and STING accelerating cell death pathways including caspases with inflammation enhancing alveolar septa destruction, remodeling, and fibrosis. Inhibitors of mitochondrial ROS, NLRP3 inflammasome, DNA sensor, cell death pathways, and IL-1 represent novel therapeutic targets for chronic airways diseases underlined by oxidative stress.
Collapse
Affiliation(s)
- Coen H. Wiegman
- Section of Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Feng Li
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Dieudonnée Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
- ArtImmune SAS, Orléans, France
| | - Kian Fan Chung
- Section of Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
169
|
Ma R, Ortiz Serrano TP, Davis J, Prigge AD, Ridge KM. The cGAS-STING pathway: The role of self-DNA sensing in inflammatory lung disease. FASEB J 2020; 34:13156-13170. [PMID: 32860267 PMCID: PMC8121456 DOI: 10.1096/fj.202001607r] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
The presence of DNA in the cytosol is usually a sign of microbial infections, which alerts the host innate immune system to mount a defense response. Cyclic GMP-AMP synthase (cGAS) is a critical cytosolic DNA sensor that elicits robust innate immune responses through the production of the second messenger, cyclic GMP-AMP (cGAMP), which binds and activates stimulator of interferon genes (STING). However, cGAS binds to DNA irrespective of DNA sequence, therefore, self-DNA leaked from the nucleus or mitochondria can also serve as a cGAS ligand to activate this pathway and trigger extensive inflammatory responses. Dysregulation of the cGAS-STING pathway is responsible for a broad array of inflammatory and autoimmune diseases. Recently, evidence has shown that self-DNA release and cGAS-STING pathway over-activation can drive lung disease, making this pathway a promising therapeutic target for inflammatory lung disease. Here, we review recent advances on the cGAS-STING pathway governing self-DNA sensing, highlighting its role in pulmonary disease.
Collapse
Affiliation(s)
- Ruihua Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tatiana P Ortiz Serrano
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Davis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew D Prigge
- Division of Critical Care Medicine, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
170
|
Liu S, Chen D, Li X, Guan M, Zhou Y, Li L, Jia W, Zhou C, Shu C, Wang C, Bai C. Fullerene nanoparticles: a promising candidate for the alleviation of silicosis-associated pulmonary inflammation. NANOSCALE 2020; 12:17470-17479. [PMID: 32808001 DOI: 10.1039/d0nr04401f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Chronic exposure to crystalline silica causes the development of silicosis, which is one of the most important occupational diseases worldwide. In the early stage of silicosis, inhaled silica crystals initiate oxidative stress, a cycle of persistent inflammation and lung injury. And it is crucial to prevent the deteriorative progression in the onset of the disease. Herein, we present a promising candidate for the treatment of crystalline silica-induced pulmonary inflammation, using a silicosis mouse model caused by intratracheal instillation based on local administration of β-alanine and hydroxyl functionalized C70 fullerene nanoparticles (FNs). The results demonstrate that FNs could significantly alleviate inflammatory cells infiltration, lower the secretion of pro-inflammatory cytokines, and reduce the destruction of lung architecture stimulated by crystalline silica. Further investigations reveal that FNs could effectively inhibit the activation of NLRP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome, and thus prevent the secretion of mature IL-1β and neutrophil influx, deriving from the superior ROS scavenging capability. Importantly, FNs could not cause any obvious toxicity after pulmonary administration.
Collapse
Affiliation(s)
- Shuai Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daiqin Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mirong Guan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wang Jia
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Shu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunli Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
171
|
Wu R, Högberg J, Adner M, Ramos-Ramírez P, Stenius U, Zheng H. Crystalline silica particles cause rapid NLRP3-dependent mitochondrial depolarization and DNA damage in airway epithelial cells. Part Fibre Toxicol 2020; 17:39. [PMID: 32778128 PMCID: PMC7418441 DOI: 10.1186/s12989-020-00370-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Respirable crystalline silica causes lung carcinomas and many thousand future cancer cases are expected in e.g. Europe. Critical questions are how silica causes genotoxicity in the respiratory epithelium and if new cases can be avoided by lowered permissible exposure levels. In this study we investigate early DNA damaging effects of low doses of silica particles in respiratory epithelial cells in vitro and in vivo in an effort to understand low-dose carcinogenic effects of silica particles. RESULTS We find DNA damage accumulation already after 5-10 min exposure to low doses (5 μg/cm2) of silica particles (Min-U-Sil 5) in vitro. DNA damage was documented as increased levels of γH2AX, pCHK2, by Comet assay, AIM2 induction, and by increased DNA repair (non-homologous end joining) signaling. The DNA damage response (DDR) was not related to increased ROS levels, but to a NLRP3-dependent mitochondrial depolarization. Particles in contact with the plasma membrane elicited a Ser198 phosphorylation of NLRP3, co-localization of NLRP3 to mitochondria and depolarization. FCCP, a mitochondrial uncoupler, as well as overexpressed NLRP3 mimicked the silica-induced depolarization and the DNA damage response. A single inhalation of 25 μg silica particles gave a similar rapid DDR in mouse lung. Biomarkers (CC10 and GPRC5A) indicated an involvement of respiratory epithelial cells. CONCLUSIONS Our findings demonstrate a novel mode of action (MOA) for silica-induced DNA damage and mutagenic double strand breaks in airway epithelial cells. This MOA seems independent of particle uptake and of an involvement of macrophages. Our study might help defining models for estimating exposure levels without DNA damaging effects.
Collapse
Affiliation(s)
- Rongrong Wu
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden
| | - Johan Högberg
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden
| | - Mikael Adner
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden
| | - Patricia Ramos-Ramírez
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden
| | - Ulla Stenius
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden
| | - Huiyuan Zheng
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-17177, Stockholm, Sweden.
| |
Collapse
|
172
|
Self DNA perpetuates IPF lung fibroblast senescence in a cGAS-dependent manner. Clin Sci (Lond) 2020; 134:889-905. [PMID: 32219338 DOI: 10.1042/cs20191160] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
Senescence and mitochondrial stress are mutually reinforcing age-related processes that contribute to idiopathic pulmonary fibrosis (IPF); a lethal disease that manifests primarily in the elderly. Whilst evidence is accumulating that GMP-AMP synthase (cGAS) is crucial in perpetuating senescence by binding damaged DNA released into the cytosol, its role in IPF is not known. The present study examines the contributions of cGAS and self DNA to the senescence of lung fibroblasts from IPF patients (IPF-LFs) and age-matched controls (Ctrl-LFs). cGAS immunoreactivity was observed in regions of fibrosis associated with fibroblasts in lung tissue of IPF patients. Pharmacological inhibition of cGAS or its knockdown by silencing RNA (siRNA) diminished the escalation of IPF-LF senescence in culture over 7 days as measured by decreased p21 and p16 expression, histone 2AXγ phosphorylation and/or IL-6 production (P < 0.05, n = 5-8). The targeting of cGAS also attenuated etoposide-induced senescence in Ctrl-LFs (P < 0.05, n = 5-8). Levels of mitochondrial DNA (mDNA) detected by qPCR in the cytosol and medium of IPF-LFs or senescence-induced Ctrl-LFs were higher than Ctrl-LFs at baseline (P < 0.05, n = 5-7). The addition of DNAse I (100 U/ml) deaccelerated IPF-LF senescence (P < 0.05, n = 5), whereas ectopic mDNA or the induction of endogenous mDNA release augmented Ctrl-LF senescence in a cGAS-dependent manner (P < 0.05, n = 5). In conclusion, we provide evidence that cGAS reinforces lung fibroblast senescence involving damaged self DNA. The targeting of cGAS to supress senescent-like responses may have potential important therapeutic implications in the treatment of IPF.
Collapse
|
173
|
Segueni N, Jacobs M, Ryffel B. Innate type 1 immune response, but not IL-17 cells control tuberculosis infection. Biomed J 2020; 44:165-171. [PMID: 32798210 PMCID: PMC8178558 DOI: 10.1016/j.bj.2020.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/16/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023] Open
Abstract
The role of the innate immune response and host resistance to Mycobacterium tuberculosis infection (TB) is reviewed. Based on our data and the abundant literature, an early type 1 immune response is critical for infection control, while ILC3 and Th17 cells seem to be dispensable. Indeed, in M. tuberculosis infected mice, transcriptomic levels of Il17, Il17ra, Il22 and Il23a were not significantly modified as compared to controls, suggesting a limited role of IL-17 and IL-22 pathways in TB infection control. Neutralization of IL-17A or IL-17F did not affect infection control either. Ongoing clinical studies with IL-17 neutralizing antibodies show high efficacy in patients with psoriasis without increased incidence of TB infection or reactivation. Therefore, both experimental studies in mice and clinical trials in human patients suggest no risk of TB infection or reactivation by therapeutic IL-17 antibodies, unlike by TNF.
Collapse
Affiliation(s)
- Noria Segueni
- Molecular and Experimental Immunology and Neurogenetics, UMR 7355, INEM, CNRS-University of Orleans, Orleans, France
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, South Africa; National Health Laboratory Service, Johannesburg, South Africa; Immunology of Infectious Disease Research Unit, University of Cape Town, South Africa
| | - Bernhard Ryffel
- Molecular and Experimental Immunology and Neurogenetics, UMR 7355, INEM, CNRS-University of Orleans, Orleans, France.
| |
Collapse
|
174
|
Fluoropyrimidine Modulation of the Anti-Tumor Immune Response-Prospects for Improved Colorectal Cancer Treatment. Cancers (Basel) 2020; 12:cancers12061641. [PMID: 32575843 PMCID: PMC7352193 DOI: 10.3390/cancers12061641] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy modulates the anti-tumor immune response and outcomes depend on the balance of favorable and unfavorable effects of drugs on anti-tumor immunity. 5-Florouracil (5-FU) is widely used in adjuvant chemotherapy regimens to treat colorectal cancer (CRC) and provides a survival benefit. However, survival remains poor for CRC patients with advanced and metastatic disease and immune checkpoint blockade therapy benefits only a sub-set of CRC patients. Here we discuss the effects of 5-FU-based chemotherapy regimens to the anti-tumor immune response. We consider how different aspects of 5-FU's multi-factorial mechanism differentially affect malignant and immune cell populations. We summarize recent studies with polymeric fluoropyrimidines (e.g., F10, CF10) that enhance DNA-directed effects and discuss how such approaches may be used to enhance the anti-tumor immune response and improve outcomes.
Collapse
|
175
|
Peng Y, Zhuang J, Ying G, Zeng H, Zhou H, Cao Y, Chen H, Xu C, Fu X, Xu H, Li J, Cao S, Chen J, Gu C, Yan F, Chen G. Stimulator of IFN genes mediates neuroinflammatory injury by suppressing AMPK signal in experimental subarachnoid hemorrhage. J Neuroinflammation 2020; 17:165. [PMID: 32450897 PMCID: PMC7247752 DOI: 10.1186/s12974-020-01830-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neuroinflammation is closely associated with the poor prognosis in subarachnoid hemorrhage (SAH) patients. This study was aimed to determine the role of stimulator of IFN genes (STING), an essential regulator to innate immunity, in the context of SAH. METHODS A total of 344 male C57BL/6 J mice were subjected to endovascular perforation to develop a model of SAH. Selective STING antagonist C-176 and STING agonist CMA were administered at 30 min or 1 h post-modeling separately. To investigate the underlying mechanism, the AMPK inhibitor compound C was administered intracerebroventricularly at 30 min before surgery. Post-SAH assessments included SAH grade, neurological test, brain water content, western blotting, RT-PCR, and immunofluorescence. Oxygenated hemoglobin was introduced into BV2 cells to establish a SAH model in vitro. RESULTS STING was mainly distributed in microglia, and microglial STING expression was significantly increased after SAH. Administration of C-176 substantially attenuated SAH-induced brain edema and neuronal injury. More importantly, C-176 significantly alleviated both short-term and persistent neurological dysfunction after SAH. Meanwhile, STING agonist CMA remarkably exacerbated neuronal injury and deteriorated neurological impairments. Mechanically, STING activation aggravated neuroinflammation via promoting microglial activation and polarizing into M1 phenotype, evidenced by microglial morphological changes, as well as the increased level of microglial M1 markers including IL-1β, iNOS, IL-6, TNF-α, MCP-1, and NLRP3 inflammasome, while C-176 conferred a robust anti-inflammatory effect. However, all the mentioned beneficial effects of C-176 including alleviated neuroinflammation, attenuated neuronal injury and the improved neurological function were reversed by AMPK inhibitor compound C. Meanwhile, the critical role of AMPK signal in C-176 mediated anti-inflammatory effect was also confirmed in vitro. CONCLUSION Microglial STING yielded neuroinflammation after SAH, while pharmacologic inhibition of STING could attenuate SAH-induced inflammatory injury at least partly by activating AMPK signal. These data supported the notion that STING might be a potential therapeutic target for SAH.
Collapse
Affiliation(s)
- Yucong Peng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Guangyu Ying
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Hanhai Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Hang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Huaijun Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Chaoran Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Hangzhe Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Jianru Li
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Shenglong Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Jingyin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Chi Gu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China.
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310000, China.
| |
Collapse
|
176
|
Hu D, Cui YX, Wu MY, Li L, Su LN, Lian Z, Chen H. Cytosolic DNA sensor cGAS plays an essential pathogenetic role in pressure overload-induced heart failure. Am J Physiol Heart Circ Physiol 2020; 318:H1525-H1537. [PMID: 32383996 DOI: 10.1152/ajpheart.00097.2020] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Growing evidence shows that activation of inflammation in the heart provokes left ventricular (LV) remodeling and dysfunction in humans and experimental animals with heart failure (HF). Moreover, recent studies found that cyclic GMP-AMP synthase (cGAS), serving as a cytosolic DNA sensor, was essential for activating innate immunity against infection and cellular damage by initiating the STING-IRFs-type I IFN signaling cascade, which played important roles in regulating the inflammatory response. However, the pathophysiological role of cGAS in pressure overload-induced HF is unclear. Wild-type C57BL/6J mice and cGAS inhibition mice were subjected to transverse aortic constriction (TAC) to induce HF or sham operation. Inhibition of cGAS in the murine heart was performed using adeno-associated virus 9 (AAV9). Alterations of the cGAS/STING pathway were examined by qPCR and Western blotting. Cardiac remodeling was assessed by echocardiography as well as histological and molecular phenotyping. Compared with sham-operated mice, the cGAS/STING pathway was activated in LV tissues in TAC mice. Whereas TAC mice exhibited significant pathological cardiac remodeling and LV dysfunction, inhibition of cGAS improved early survival rates after TAC, preserved LV contractile function, and blunted pathological remodeling, including cardiac hypertrophy, fibrosis, and apoptosis. Furthermore, downregulation of cGAS diminished early inflammatory cell infiltration and inflammatory cytokine expression in response to TAC. These results demonstrated that cGAS played an essential pathogenetic role in pressure overload-induced HF to promote pathological cardiac remodeling and dysfunction. Our results suggest that inhibition of cGAS may be a novel therapeutic approach for HF.NEW & NOTEWORTHY In this study, we first revealed a novel role of cGAS in the regulation of pathological cardiac remodeling and dysfunction upon pressure overload. We found that the cGAS/STING pathway was activated during pressure overload. Moreover, we also demonstrated that inhibition of the cGAS/STING pathway alleviated pathological cardiac remodeling and downregulated the early inflammatory response during pressure overload-induced HF. Together, these findings will provide a new therapeutic target for HF.
Collapse
Affiliation(s)
- Dan Hu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Yu-Xia Cui
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Man-Yan Wu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Long Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Li-Na Su
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Zheng Lian
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| |
Collapse
|
177
|
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a novel coronavirus that has caused a worldwide pandemic of the human respiratory illness COVID-19, resulting in a severe threat to public health and safety. Analysis of the genetic tree suggests that SARS-CoV-2 belongs to the same Betacoronavirus group as severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). Although the route for viral transmission remains a mystery, SARS-CoV-2 may have originated in an animal reservoir, likely that of bat. The clinical features of COVID-19, such as fever, cough, shortness of breath, and fatigue, are similar to those of many acute respiratory infections. There is currently no specific treatment for COVID-19, but antiviral therapy combined with supportive care is the main strategy. Here, we summarize recent progress in understanding the epidemiological, virological, and clinical characteristics of COVID-19 and discuss potential targets with existing drugs for the treatment of this emerging zoonotic disease.
Collapse
Affiliation(s)
- Daolin Tang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Paul Comish
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
178
|
Wan D, Jiang W, Hao J. Research Advances in How the cGAS-STING Pathway Controls the Cellular Inflammatory Response. Front Immunol 2020; 11:615. [PMID: 32411126 PMCID: PMC7198750 DOI: 10.3389/fimmu.2020.00615] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
Double-stranded DNA (dsDNA) sensor cyclic-GMP-AMP synthase (cGAS) along with the downstream stimulator of interferon genes (STING) acting as essential immune-surveillance mediators have become hot topics of research. The intrinsic function of the cGAS-STING pathway facilitates type-I interferon (IFN) inflammatory signaling responses and other cellular processes such as autophagy, cell survival, senescence. cGAS-STING pathway interplays with other innate immune pathways, by which it participates in regulating infection, inflammatory disease, and cancer. The therapeutic approaches targeting this pathway show promise for future translation into clinical applications. Here, we present a review of the important previous works and recent advances regarding the cGAS-STING pathway, and provide a comprehensive understanding of the modulatory pattern of the cGAS-STING pathway under multifarious pathologic states.
Collapse
Affiliation(s)
- Dongshan Wan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
179
|
Decreased Soluble Receptor of Advanced Glycation End Product Levels Correlated with Inflammation in Silicosis. Mediators Inflamm 2020; 2020:2683753. [PMID: 32351319 PMCID: PMC7178542 DOI: 10.1155/2020/2683753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Silicosis is a devastating disease caused by inhalation of silica dust that leads to inflammatory cascade and then scarring of the lung tissue. Increasing evidences indicate that soluble receptor for advanced glycation end products (sRAGE) is involved in inflammatory diseases. However, no data on the possible relationship between sRAGE and inflammation of silicosis are available. In this study, serum from subjects with silicosis (n = 59) or from healthy controls (HC, n = 14) was analyzed for the secretion of sRAGE, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), and oxidized low-density lipoprotein (ox-LDL). The associations between sRAGE and cytokines and ox-LDL and lung function were assessed by Pearson's correlation analyses. Mean levels of serum sRAGE were lower in silicosis than those in controls (p < 0.05). The subjects who had a longer term of occupational exposure had higher levels of sRAGE (p < 0.05). The secretion of TNF-α, IL-1β, IL-6, TGF-β1, and ox-LDL was significantly higher in the silicosis group than that in the HC group (p < 0.05). Furthermore, the levels of sRAGE were negatively correlated with TNF-α, IL-6, IL-1β, and ox-LDL. There is no correlation between sRAGE and TGF-β1 and lung function. The optimal point of sRAGE for differentiating silicosis from healthy controls was 14250.02 pg/ml by ROC curve analysis. A decrease in serum sRAGE and its association with inflammatory response might suggest a role for sRAGE in the pathogenesis of silicosis.
Collapse
|
180
|
Hu Q, Wu J, Ren Y, Wu X, Gao L, Wang G, Gu G, Ren H, Hong Z, Slade DA, Ren J. Degree of STING activation is associated with disease outcomes. Gut 2020; 69:792-794. [PMID: 30996043 DOI: 10.1136/gutjnl-2019-318597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/01/2019] [Accepted: 04/06/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Qiongyuan Hu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanhan Ren
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lin Gao
- Pancreatic Center, Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Gefei Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guosheng Gu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huajian Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiwu Hong
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Dominic A Slade
- Department of Surgery, Salford Royal NHS Foundation Trust, Salford, UK
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
181
|
Kim SJ, Cheresh P, Jablonski RP, Rachek L, Yeldandi A, Piseaux-Aillon R, Ciesielski MJ, Ridge K, Gottardi C, Lam AP, Pardo A, Selman M, Natarajan V, Kamp DW. Mitochondrial 8-oxoguanine DNA glycosylase mitigates alveolar epithelial cell PINK1 deficiency, mitochondrial DNA damage, apoptosis, and lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1084-L1096. [PMID: 32209025 DOI: 10.1152/ajplung.00069.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alveolar epithelial cell (AEC) apoptosis, arising from mitochondrial dysfunction and mitophagy defects, is important in mediating idiopathic pulmonary fibrosis (IPF). Our group established a role for the mitochondrial (mt) DNA base excision repair enzyme, 8-oxoguanine-DNA glycosylase 1 (mtOGG1), in preventing oxidant-induced AEC mtDNA damage and apoptosis and showed that OGG1-deficient mice have increased lung fibrosis. Herein, we determined whether mice overexpressing the mtOGG1 transgene (mtOgg1tg) are protected against lung fibrosis and whether AEC mtOGG1 preservation of mtDNA integrity mitigates phosphatase and tensin homolog-induced putative kinase 1 (PINK1) deficiency and apoptosis. Compared with wild type (WT), mtOgg1tg mice have diminished asbestos- and bleomycin-induced pulmonary fibrosis that was accompanied by reduced lung and AEC mtDNA damage and apoptosis. Asbestos and H2O2 promote the MLE-12 cell PINK1 deficiency, as assessed by reductions in the expression of PINK1 mRNA and mitochondrial protein expression. Compared with WT, Pink1-knockout (Pink1-KO) mice are more susceptible to asbestos-induced lung fibrosis and have increased lung and alveolar type II (AT2) cell mtDNA damage and apoptosis. AT2 cells from Pink1-KO mice and PINK1-silenced (siRNA) MLE-12 cells have increased mtDNA damage that is augmented by oxidative stress. Interestingly, mtOGG1 overexpression attenuates oxidant-induced MLE-12 cell mtDNA damage and apoptosis despite PINK1 silencing. mtDNA damage is increased in the lungs of patients with IPF as compared with controls. Collectively, these findings suggest that mtOGG1 maintenance of AEC mtDNA is crucial for preventing PINK1 deficiency that promotes apoptosis and lung fibrosis. Given the key role of AEC apoptosis in pulmonary fibrosis, strategies aimed at preserving AT2 cell mtDNA integrity may be an innovative target.
Collapse
Affiliation(s)
- Seok-Jo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Paul Cheresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Renea P Jablonski
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Lyudmila Rachek
- Department of Cell Biology and Neuroscience, University of South Alabama College of Medicine, Mobile, Alabama
| | - Anjana Yeldandi
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Raul Piseaux-Aillon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Mark J Ciesielski
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Karen Ridge
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Cara Gottardi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Anna P Lam
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | - David W Kamp
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
182
|
Zhang Y, Chen W, Wang Y. STING is an essential regulator of heart inflammation and fibrosis in mice with pathological cardiac hypertrophy via endoplasmic reticulum (ER) stress. Biomed Pharmacother 2020; 125:110022. [PMID: 32106379 DOI: 10.1016/j.biopha.2020.110022] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Pathological cardiac hypertrophy is characterized by myocyte enlargement and cardiac dysfunction. However, the pathogenesis for this disease is still poorly understood. Stimulator of interferon genes (STING) could meditate inflammation and immune response in various kinds of diseases. In this work, we demonstrated that STING was critical for pressure overload-induced cardiac hypertrophy. Results showed that STING expression was up-regulated in human and mouse hypertrophic hearts. STING knockout attenuated cardiac hypertrophy induced by aortic banding (AB). The effects of STING deficiency on the improvement of cardiac hypertrophy and dysfunction were associated with the restrained macrophage infiltration, inflammatory response and fibrosis. Moreover, ER stress was detected in hearts of AB-operated mice, as evidenced by the increased expression of phospho-protein kinase RNA-like endoplasmic reticulum kinase (PERK), phospho-eukaryotic initiation factor 2 alpha (eIF2α) and phospho-inositol-requiring kinase (IRE)-1α. Importantly, these proteins were restrained in mice with STING knockout after AB surgery. What's more, angiotensin II (Ang II)-induced STING could be accelerated by ER stress activator, while being markedly abolished by the ER stress inhibitor. We then found that whether co-treated with or without transforming growth factor-beta 1 (TGF-β1), cardiac fibroblasts cultured in the conditional medium (CM) from Ang II-incubated cardiomyocytes with STING knockdown exhibited significantly reduced fibrosis, as displayed by the clearly down-regulated expression of α-SMA, Collagen type I (Col I) and Collagen type III (Col III). Therefore, we defined STING as an important signal contributing to cardiac hypertrophy closely associated with ER stress.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, China
| | - Wenzhong Chen
- Department of Cardiovascular Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Yan Wang
- Department of General Medicine, The Second Affiliated Hospital of Shenzhen University(People's Hospital of Shenzhen Baoan District), Shenzhen City, 518101, China.
| |
Collapse
|
183
|
Fang L, Sun Q, Roth M. Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma. Int J Mol Sci 2020; 21:ijms21030757. [PMID: 31979396 PMCID: PMC7037330 DOI: 10.3390/ijms21030757] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Asthma increases worldwide without any definite reason and patient numbers double every 10 years. Drugs used for asthma therapy relax the muscles and reduce inflammation, but none of them inhibited airway wall remodeling in clinical studies. Airway wall remodeling can either be induced through pro-inflammatory cytokines released by immune cells, or direct binding of IgE to smooth muscle cells, or non-immunological stimuli. Increasing evidence suggests that airway wall remodeling is initiated early in life by epigenetic events that lead to cell type specific pathologies, and modulate the interaction between epithelial and sub-epithelial cells. Animal models are only available for remodeling in allergic asthma, but none for non-allergic asthma. In human asthma, the mechanisms leading to airway wall remodeling are not well understood. In order to improve the understanding of this asthma pathology, the definition of “remodeling” needs to be better specified as it summarizes a wide range of tissue structural changes. Second, it needs to be assessed if specific remodeling patterns occur in specific asthma pheno- or endo-types. Third, the interaction of the immune cells with tissue forming cells needs to be assessed in both directions; e.g., do immune cells always stimulate tissue cells or are inflamed tissue cells calling immune cells to the rescue? This review aims to provide an overview on immunologic and non-immunologic mechanisms controlling airway wall remodeling in asthma.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
| | - Qinzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Michael Roth
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-2337
| |
Collapse
|
184
|
Mariotte A, De Cauwer A, Po C, Abou-Faycal C, Pichot A, Paul N, Aouadi I, Carapito R, Frisch B, Macquin C, Chatelus E, Sibilia J, Armspach JP, Bahram S, Georgel P. A mouse model of MSU-induced acute inflammation in vivo suggests imiquimod-dependent targeting of Il-1β as relevant therapy for gout patients. Am J Cancer Res 2020; 10:2158-2171. [PMID: 32104502 PMCID: PMC7019178 DOI: 10.7150/thno.40650] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: The role of Monosodium Urate (MSU) crystals in gout pathophysiology is well described, as is the major impact of IL-1β in the inflammatory reaction that constitutes the hallmark of the disease. However, despite the discovery of the NLRP3 inflammasome and its role as a Pattern Recognition Receptor linking the detection of a danger signal (MSU) to IL-1β secretion in vitro, the precise mechanisms leading to joint inflammation in gout patients are still poorly understood. Methods: Acute urate crystal inflammation was obtained by subcutaneous injections of MSU crystals in mice. Symptoms were followed by scoring, cytokine quantification by ELISA and western blot, gene expression by RT-qPCR and RNAseq; Magnetic Resonance Imaging was also used to assess inflammation. Results: We provide an extensive clinical, biological and molecular characterization of an acute uratic inflammation mouse model which accurately mimics human gout. We report the efficacy of topical imiquimod treatment and its impact on Interferon-dependent down modulation of Il-1β gene expression in this experimental model. Conclusion: Our work reveals several key features of MSU-dependent inflammation and identifies novel therapeutic opportunities for gout patients.
Collapse
|
185
|
Khan MF, Wang H. Environmental Exposures and Autoimmune Diseases: Contribution of Gut Microbiome. Front Immunol 2020; 10:3094. [PMID: 31998327 PMCID: PMC6970196 DOI: 10.3389/fimmu.2019.03094] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Environmental agents have been gaining more attention in recent years for their role in the pathogenesis of autoimmune diseases (ADs). Increasing evidence has linked environmental exposures, including trichloroethene (TCE), silica, mercury, pristane, pesticides, and smoking to higher risk for ADs. However, potential mechanisms by which these environmental agents contribute to the disease pathogenesis remains largely unknown. Dysbiosis of the gut microbiome is another important environmental factor that has been linked to the onset of different ADs. Altered microbiota composition is associated with impaired intestinal barrier function and dysregulation of mucosal immune system, but it is unclear if gut dysbiosis is a causal factor or an outcome of ADs. In this review article, we first describe the recent epidemiological and mechanistic evidences linking environmental/occupational exposures with various ADs (especially SLE). Secondly, we discuss how changes in the gut microbiome composition (dysbiosis) could contribute to the disease pathogenesis, especially in response to exposure to environmental chemicals.
Collapse
Affiliation(s)
- M. Firoze Khan
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | | |
Collapse
|
186
|
Qu H, Li L, Wang TL, Seckin T, Segars J, Shih IM. Epithelial Cells in Endometriosis and Adenomyosis Upregulate STING Expression. Reprod Sci 2020; 27:1276-1284. [PMID: 32046461 DOI: 10.1007/s43032-019-00127-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/09/2019] [Indexed: 01/10/2023]
Abstract
In response to cytosolic DNA, stimulator of interferon gene (STING) initiates and orchestrates host's innate immunity by inducing type I interferon. Since endometriosis is a chronic inflammatory disorder, we sought to determine whether STING pathway is activated in ectopic endometrium in comparison to eutopic endometrium. Immunohistochemistry was employed in evaluating the expression levels of STING in normal endometrium, endometriosis, and adenomyosis. The density of CD45+ intraepithelial lymphocytes was correlated with STING expression levels. A total of 39 cases of endometriosis and/or adenomyosis with normal endometrium were analyzed. Among them, 32 had adenomyosis, 26 had endometriosis, and 19 have both lesions. STING protein expression is mainly evident in the cytoplasm of epithelial cells but much less in stromal cells. Based on H-score, we found that the STING expression levels were significantly higher in the epithelial cells of adenomyosis and endometriosis than in eutopic endometrium (132.7 ± 12.20, 119.6 ± 12.57 vs. 19.74 ± 5.96, p < 0.0001). There was no significant difference in STING expression level between endometriosis and adenomyosis. More intraepithelial lymphocytes were detected in endometriosis and adenomyosis lesions than endometrium (5.60 ± 0.70%, 4.95 ± 0.54% vs. 1.25 ± 0.12%, p < 0.0001). A positive correlation between STING expression and intraepithelial lymphocytic infiltrate was observed (p < 0.0001). In summary, STING was upregulated in the epithelium of ectopic endometrium as compared to eutopic endometrium. Its expression levels correlate with the degree of intraepithelial lymphocyte infiltration, suggesting a role in promoting chronic inflammation of ectopic endometrium.
Collapse
Affiliation(s)
- Hong Qu
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lihong Li
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tamer Seckin
- Lenox Hill Hospital and Zucker School of Medicine at Hofstra/Northwell , New York, USA
| | - James Segars
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
187
|
Luo W, Wang Y, Zhang L, Ren P, Zhang C, Li Y, Azares AR, Zhang M, Guo J, Ghaghada KB, Starosolski ZA, Rajapakshe K, Coarfa C, Li Y, Chen R, Fujiwara K, Abe JI, Coselli JS, Milewicz DM, LeMaire SA, Shen YH. Critical Role of Cytosolic DNA and Its Sensing Adaptor STING in Aortic Degeneration, Dissection, and Rupture. Circulation 2019; 141:42-66. [PMID: 31887080 DOI: 10.1161/circulationaha.119.041460] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Sporadic aortic aneurysm and dissection (AAD), caused by progressive aortic smooth muscle cell (SMC) loss and extracellular matrix degradation, is a highly lethal condition. Identifying mechanisms that drive aortic degeneration is a crucial step in developing an effective pharmacologic treatment to prevent disease progression. Recent evidence has indicated that cytosolic DNA and abnormal activation of the cytosolic DNA sensing adaptor STING (stimulator of interferon genes) play a critical role in vascular inflammation and destruction. Here, we examined the involvement of this mechanism in aortic degeneration and sporadic AAD formation. METHODS The presence of cytosolic DNA in aortic cells and activation of the STING pathway were examined in aortic tissues from patients with sporadic ascending thoracic AAD. The role of STING in AAD development was evaluated in Sting-deficient (Stinggt/gt) mice in a sporadic AAD model induced by challenging mice with a combination of a high-fat diet and angiotensin II. We also examined the direct effects of STING on SMC death and macrophage activation in vitro. RESULTS In human sporadic AAD tissues, we observed the presence of cytosolic DNA in SMCs and macrophages and significant activation of the STING pathway. In the sporadic AAD model, Stinggt/gt mice showed significant reductions in challenge-induced aortic enlargement, dissection, and rupture in both the thoracic and abdominal aortic regions. Single-cell transcriptome analysis revealed that aortic challenge in wild-type mice induced the DNA damage response, the inflammatory response, dedifferentiation and cell death in SMCs, and matrix metalloproteinase expression in macrophages. These changes were attenuated in challenged Stinggt/gt mice. Mechanistically, nuclear and mitochondrial DNA damage in SMCs and the subsequent leak of DNA to the cytosol activated STING signaling, which induced cell death through apoptosis and necroptosis. In addition, DNA from damaged SMCs was engulfed by macrophages in which it activated STING and its target interferon regulatory factor 3, which directly induced matrix metalloproteinase-9 expression. We also found that pharmacologically inhibiting STING activation partially prevented AAD development. CONCLUSIONS Our findings indicate that the presence of cytosolic DNA and subsequent activation of cytosolic DNA sensing adaptor STING signaling represent a key mechanism in aortic degeneration and that targeting STING may prevent sporadic AAD development.
Collapse
Affiliation(s)
- Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Alon R Azares
- Molecular Cardiology Research Lab (A.R.A.), Texas Heart Institute, Houston
| | - Michelle Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| | - Jiao Guo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, Houston (K.B.G., Z.A.S.)
| | | | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology (K.R., C.C.), Baylor College of Medicine, Houston, TX
| | - Cristian Coarfa
- Dan L. Duncan Cancer Center (C.C.), Baylor College of Medicine, Houston, TX
| | - Yumei Li
- Human Genome Sequencing Center (Yumei Li, R.C.), Baylor College of Medicine, Houston, TX
| | - Rui Chen
- Department of Biochemistry and Molecular Biology (R.C.), Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics (R.C.), Baylor College of Medicine, Houston, TX
- Human Genome Sequencing Center (Yumei Li, R.C.), Baylor College of Medicine, Houston, TX
| | - Keigi Fujiwara
- Department of Biostatistics and Division of Internal Medicine, Department of Cardiology Research, The University of Texas MD Anderson Cancer Center, Houston (K.F., J.A.)
| | - Jun-Ichi Abe
- Department of Biostatistics and Division of Internal Medicine, Department of Cardiology Research, The University of Texas MD Anderson Cancer Center, Houston (K.F., J.A.)
| | - Joseph S Coselli
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M.)
| | - Scott A LeMaire
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Ying H Shen
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| |
Collapse
|
188
|
Benninghoff AD, Bates MA, Chauhan PS, Wierenga KA, Gilley KN, Holian A, Harkema JR, Pestka JJ. Docosahexaenoic Acid Consumption Impedes Early Interferon- and Chemokine-Related Gene Expression While Suppressing Silica-Triggered Flaring of Murine Lupus. Front Immunol 2019; 10:2851. [PMID: 31921124 PMCID: PMC6923248 DOI: 10.3389/fimmu.2019.02851] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 11/20/2019] [Indexed: 12/18/2022] Open
Abstract
Exposure of lupus-prone female NZBWF1 mice to respirable crystalline silica (cSiO2), a known human autoimmune trigger, initiates loss of tolerance, rapid progression of autoimmunity, and early onset of glomerulonephritis. We have previously demonstrated that dietary supplementation with the ω-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) suppresses autoimmune pathogenesis and nephritis in this unique model of lupus flaring. In this report, we utilized tissues from prior studies to test the hypothesis that DHA consumption interferes with upregulation of critical genes associated with cSiO2-triggered murine lupus. A NanoString nCounter platform targeting 770 immune-related genes was used to assess the effects cSiO2 on mRNA signatures over time in female NZBWF1 mice consuming control (CON) diets compared to mice fed diets containing DHA at an amount calorically equivalent to human consumption of 2 g per day (DHA low) or 5 g per day (DHA high). Experimental groups of mice were sacrificed: (1) 1 d after a single intranasal instillation of 1 mg cSiO2 or vehicle, (2) 1 d after four weekly single instillations of vehicle or 1 mg cSiO2, and (3) 1, 5, 9, and 13 weeks after four weekly single instillations of vehicle or 1 mg cSiO2. Genes associated with inflammation as well as innate and adaptive immunity were markedly upregulated in lungs of CON-fed mice 1 d after four weekly cSiO2 doses but were significantly suppressed in mice fed DHA high diets. Importantly, mRNA signatures in lungs of cSiO2-treated CON-fed mice over 13 weeks reflected progressive amplification of interferon (IFN)- and chemokine-related gene pathways. While these responses in the DHA low group were suppressed primarily at week 5, significant downregulation was observed at weeks 1, 5, 9, and 13 in mice fed the DHA high diet. At week 13, cSiO2 treatment of CON-fed mice affected 214 genes in kidney tissue associated with inflammation, innate/adaptive immunity, IFN, chemokines, and antigen processing, mostly by upregulation; however, feeding DHA dose-dependently suppressed these responses. Taken together, dietary DHA intake in lupus-prone mice impeded cSiO2-triggered mRNA signatures known to be involved in ectopic lymphoid tissue neogenesis, systemic autoimmunity, and glomerulonephritis.
Collapse
Affiliation(s)
- Abby D. Benninghoff
- Department of Animal, Dairy and Veterinary Sciences and The School of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Melissa A. Bates
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S. Chauhan
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kathryn A. Wierenga
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Kristen N. Gilley
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Andrij Holian
- Department of Biomedical and Pharmaceutical Sciences, Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States
| | - Jack R. Harkema
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
189
|
Abstract
Two environmental factors, crystalline silica (cSiO2), a toxic airborne particle encountered occupationally, and docosahexaenoic acid (DHA), a dietary omega-3 highly unsaturated fatty acid (HUFA), have the potential to influence the development of systemic lupus erythematosus (lupus). Using the NZBWF1 mouse, which spontaneously develops lupus, we found that intranasal exposure to cSiO2 significantly decreases latency and promotes rapid progression of the disease. Specifically, cSiO2 induces the development of ectopic lymphoid structures (ELS) containing germinal centers in the lungs that yield vigorous and diverse autoantibody responses locally and systemically. Transcriptomic analysis revealed that cSiO2 promotes a robust type I interferon gene signature that likely precipitates ELS neogenesis. Intriguingly, dietary supplementation with human-relevant doses of DHA impedes cSiO2-induced gene expression, ELS neogenesis, autoantibody elevation, and glomerulonephritis in this lupus-prone mouse model. Together, our findings point to the feasibility of enhancing tissue omega-3 HUFAs as a personalized nutritional intervention to impede onset and progression of environment-triggered autoimmune disease.
Collapse
Affiliation(s)
- Kathryn A Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Jack R Harkema
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - James J Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
190
|
Maekawa H, Inagi R. Pathophysiological Role of Organelle Stress/Crosstalk in AKI-to-CKD Transition. Semin Nephrol 2019; 39:581-588. [DOI: 10.1016/j.semnephrol.2019.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
191
|
Self-DNA release and STING-dependent sensing drives inflammation to cigarette smoke in mice. Sci Rep 2019; 9:14848. [PMID: 31619733 PMCID: PMC6795997 DOI: 10.1038/s41598-019-51427-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/30/2019] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoke exposure is a leading cause of chronic obstructive pulmonary disease (COPD), a major health issue characterized by airway inflammation with fibrosis and emphysema. Here we demonstrate that acute exposure to cigarette smoke causes respiratory barrier damage with the release of self-dsDNA in mice. This triggers the DNA sensor cGAS (cyclic GMP-AMP synthase) and stimulator of interferon genes (STING), driving type I interferon (IFN I) dependent lung inflammation, which are attenuated in cGAS, STING or type I interferon receptor (IFNAR) deficient mice. Therefore, we demonstrate a critical role of self-dsDNA release and of the cGAS-STING-type I interferon pathway upon cigarette smoke-induced damage, which may lead to therapeutic targets in COPD.
Collapse
|
192
|
Akgün M. Reporting an Index Case: "Mighty Oaks from Little Acorns Grow". Turk Thorac J 2019; 20:267-268. [PMID: 31584391 DOI: 10.5152/turkthoracj.2019.191909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 11/22/2022]
Affiliation(s)
- Metin Akgün
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
193
|
Sokolowska M, Quesniaux VFJ, Akdis CA, Chung KF, Ryffel B, Togbe D. Acute Respiratory Barrier Disruption by Ozone Exposure in Mice. Front Immunol 2019; 10:2169. [PMID: 31608051 PMCID: PMC6758598 DOI: 10.3389/fimmu.2019.02169] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022] Open
Abstract
Ozone exposure causes irritation, airway hyperreactivity (AHR), inflammation of the airways, and destruction of alveoli (emphysema), the gas exchange area of the lung in human and mice. This review focuses on the acute disruption of the respiratory epithelial barrier in mice. A single high dose ozone exposure (1 ppm for 1 h) causes first a break of the bronchiolar epithelium within 2 h with leak of serum proteins in the broncho-alveolar space, disruption of epithelial tight junctions and cell death, which is followed at 6 h by ROS activation, AHR, myeloid cell recruitment, and remodeling. High ROS levels activate a novel PGAM5 phosphatase dependent cell-death pathway, called oxeiptosis. Bronchiolar cell wall damage and inflammation upon a single ozone exposure are reversible. However, chronic ozone exposure leads to progressive and irreversible loss of alveolar epithelial cells and alveoli with reduced gas exchange space known as emphysema. It is further associated with chronic inflammation and fibrosis of the lung, resembling other environmental pollutants and cigarette smoke in pathogenesis of asthma, and chronic obstructive pulmonary disease (COPD). Here, we review recent data on the mechanisms of ozone induced injury on the different cell types and pathways with a focus on the role of the IL-1 family cytokines and the related IL-33. The relation of chronic ozone exposure induced lung disease with asthma and COPD and the fact that ozone exacerbates asthma and COPD is emphasized.
Collapse
Affiliation(s)
- Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Valerie F J Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Kian Fan Chung
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Dieudonnée Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France.,ArtImmune SAS, Artinem, Orléans, France
| |
Collapse
|
194
|
Diminished stimulator of interferon genes production with cigarette smoke-exposure contributes to weakened anti-adenovirus vectors response and destruction of lung in chronic obstructive pulmonary disease model. Exp Cell Res 2019; 384:111545. [PMID: 31470016 DOI: 10.1016/j.yexcr.2019.111545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 12/14/2022]
Abstract
Cigarette smoke (CS) is the primary risk factor for chronic obstructive pulmonary disease (COPD) and dampens antiviral response, which increases viral infections and leads to COPD acute exacerbation (AECOPD). Adenovirus, a nonenveloped DNA virus, is linked with AECOPD, whose DNAs trigger innate immune response via interacting with pattern recognition receptors (PRRs). Stimulator of interferon genes (STING), as a cytosolic DNA sensor, participates in adenovirus-induced interferon β (IFNβ)-dependent antiviral response. STING is involved in various pulmonary diseases, but role of STING in pathogenesis of AECOPD is not well documented. In the present study, we explored relationship between STING and AECOPD induced by recombinant adenovirus vectors (rAdVs) and CS in wild type (WT) and STING-/- mice; and also characterized the inhibition of STING- IFNβ pathway in pulmonary epithelium exposed to cigarette smoke extract (CSE). We found that CS or CSE exposure alone dramatically inhibited STING expression, but not significantly effected IFNβ production. Moreover, CS or CSE-exposed significantly suppressed activation of STING-IFNβ pathway induced by rAdVs and suppressed clearance of rAdVs DNA. Inflammation, fibrosis and emphysema of lung tissues were exaggerated when treated with CS plus rAdVs, which further deteriorate in absences of STING. In A549 cells with knockdown of STING, we also observed enhancing apoptosis related to emphysema, especially CSE and adenovirus vectors in combination. Therefore, STING may play a protective role in preventing the progress of COPD.
Collapse
|
195
|
M10 peptide attenuates silica-induced pulmonary fibrosis by inhibiting Smad2 phosphorylation. Toxicol Appl Pharmacol 2019; 376:46-57. [DOI: 10.1016/j.taap.2019.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/18/2019] [Accepted: 05/20/2019] [Indexed: 01/06/2023]
|
196
|
Benmerzoug S, Ryffel B, Togbe D, Quesniaux VF. Self-DNA Sensing in Lung Inflammatory Diseases. Trends Immunol 2019; 40:719-734. [DOI: 10.1016/j.it.2019.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
|
197
|
Gonzalez-Quintial R, Mayeux JM, Kono DH, Theofilopoulos AN, Pollard KM, Baccala R. Silica exposure and chronic virus infection synergistically promote lupus-like systemic autoimmunity in mice with low genetic predisposition. Clin Immunol 2019; 205:75-82. [PMID: 31175964 DOI: 10.1016/j.clim.2019.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
Considerable evidence indicates that autoimmune disease expression depends on both genetic and environmental factors. Among potential environmental triggers, occupational airway exposure to crystalline silica and virus infections have been linked to lupus and other autoimmune diseases in both humans and mouse models. Here, we hypothesized that combined silica and virus exposures synergize and induce autoimmune manifestations more effectively than single exposure to either of these factors, particularly in individuals with low genetic predisposition. Accordingly, infection with the model murine pathogen lymphocytic choriomenigitis virus (LCMV) in early life, followed by airway exposure to crystalline silica in adult life, induced lupus-like autoantibodies to several nuclear self-antigens including chromatin, RNP and Sm, concurrent with kidney lesions, in non-autoimmune C57BL/6 (B6) mice. In contrast, given individually, LCMV or silica were largely ineffectual in this strain. These results support a multihit model of autoimmunity, where exposure to different environmental factors acting on distinct immunostimulatory pathways complements limited genetic predisposition and increases the risk of autoimmunity above a critical threshold.
Collapse
Affiliation(s)
| | - Jessica M Mayeux
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Dwight H Kono
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Kenneth M Pollard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Baccala
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
198
|
Zhong S, Li W, Bai Y, Wu B, Wang X, Jiang S, Zhao Y, Ren J, Li H, Jin R. Computational study on new natural compound agonists of stimulator of interferon genes (STING). PLoS One 2019; 14:e0216678. [PMID: 31120925 PMCID: PMC6532845 DOI: 10.1371/journal.pone.0216678] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/26/2019] [Indexed: 12/03/2022] Open
Abstract
Objective This study aimed to screen lead compounds and medication candidates from drug library (ZINC database) which has potential agonist effect targeting STING protein. Methods and materials A series of computer-aided virtual screening techniques were utilized to identify potential agonists of STING. Structure-based screening using Libdock was carried out followed by ADME (absorption, distribution, metabolism, excretion) and toxicity prediction. Molecular docking was performed to demonstrate the binding affinity and mechanism between ligands and STING dimers. Molecular dynamic simulation was utilized to evaluate the stability of ligand-receptor complex. Finally, animal experiment was conducted to validate the effectiveness of selected compounds. Results Three novel natural compounds 1,2,3 (ZINC000015149223, ZINC000011616633 and ZINC000001577210, respectively) from the ZINC15 database were found binding to STING with more favorable interaction energy. Also, they were predicted with less ames mutagenicity, rodent carcinogenicity, non-developmental toxic potential and tolerant with cytochrome P450 2D6 (CYP2D6). The ligand chemical structure analysis showed the three compounds were inborn axisymmetric, such chemical structures account for combining and activating process of STING protein dimers. The dynamic simulation analysis demonstrated that ZINC000015149223-, ZINC000011616633- and ZINC000001577210-STING dimer complex had more favorable potential energy compared with amidobenzimidazole (ABZI) and they can exist in natural environments stably. Animal experiments also demonstrated that these three compounds could suppress tumor growth. Conclusion This study demonstrates that ZINC000015149223, ZINC000011616633 and ZINC000001577210 are potential agonists targeting STING protein. These compounds are safe drug candidates and have a great significance in STING agonists development.
Collapse
Affiliation(s)
- Sheng Zhong
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, the United States of America
| | - Weihang Li
- Clinical College, Jilin University, Changchun, China
| | - Yang Bai
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Bo Wu
- Department of Orthopedics, the First Hospital of Jilin University, Changchun, China
| | - Xinhui Wang
- Department of Oncology, the First Hospital of Jilin University, Changchun, China
| | | | - Yingjing Zhao
- Clinical College, Jilin University, Changchun, China
| | - Jiaxin Ren
- Clinical College, Jilin University, Changchun, China
| | - Hui Li
- Clinical College, Jilin University, Changchun, China
| | - Rihua Jin
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
199
|
Sterile Lung Inflammation Induced by Silica Exacerbates Mycobacterium tuberculosis Infection via STING-Dependent Type 2 Immunity. Cell Rep 2019; 27:2649-2664.e5. [DOI: 10.1016/j.celrep.2019.04.110] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/17/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
|
200
|
Kumar V. A STING to inflammation and autoimmunity. J Leukoc Biol 2019; 106:171-185. [PMID: 30990921 DOI: 10.1002/jlb.4mir1018-397rr] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022] Open
Abstract
Various intracellular pattern recognition receptors (PRRs) recognize cytosolic pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Cyclic GMP-AMP synthase (cGAS), a cytosolic PRR, recognizes cytosolic nucleic acids including dsDNAs. The recognition of dsDNA by cGAS generates cyclic GMP-AMP (GAMP). The cGAMP is then recognized by STING generating type 1 IFNs and NF-κB-mediated generation of pro-inflammatory cytokines and molecules. Thus, cGAS-STING signaling mediated recognition of cytosolic dsDNA causing the induction of type 1 IFNs plays a crucial role in innate immunity against cytosolic pathogens, PAMPs, and DAMPs. The overactivation of this system may lead to the development of autoinflammation and autoimmune diseases. The article opens with the introduction of different PRRs involved in the intracellular recognition of dsDNA and gives a brief introduction of cGAS-STING signaling. The second section briefly describes cGAS as intracellular PRR required to recognize intracellular nucleic acids (dsDNA and CDNs) and the formation of cGAMP. The cGAMP acts as a second messenger to activate STING- and TANK-binding kinase 1-mediated generation of type 1 IFNs and the activation of NF-κB. The third section of the article describes the role of cGAS-STING signaling in the induction of autoinflammation and various autoimmune diseases. The subsequent fourth section describes both chemical compounds developed and the endogenous negative regulators of cGAS-STING signaling required for its regulation. Therapeutic targeting of cGAS-STING signaling could offer new ways to treat inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|