101
|
Inhibition of cGAS ameliorates acute lung injury triggered by zinc oxide nanoparticles. Toxicol Lett 2022; 373:62-75. [PMID: 36368621 DOI: 10.1016/j.toxlet.2022.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Zinc oxide nanoparticles (ZnONPs) have been widely used in various industrial and biomedical fields. Occupational or accidental inhalation exposure to ZnONPs might lead to acute lung injury (ALI). Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are critical for the initiation and expansion of inflammation and contribute to tissue injury; however, the role and mechanism of the cGAS-STING pathway in ALI-induced by ZnONPs are unclear. METHODS Male C57BL/6 J mice were intratracheally injected with ZnONPs (0.6 mg/kg) or mock. The mice were euthanized and the degree of lung injury was determined 3 days after the instillation of ZnONPs. The BEAS-2B cell line was used as a cell model to investigate the cytotoxicity of ZnONPs in vitro. RESULTS We found that ZnONPs inhalation induced ALI in mice, manifested by exacerbated lung pathological changes, mitochondrial damage, oxidative stress and inflammation. Interestingly, cGAS and STING were activated in the lung tissues of the mice and BEAS-2B lung epithelial cells treated with ZnONPs. More importantly, we illustrated that the cGAS inhibitor RU.521 inhibited the activation of the cGAS-STING pathway, further decreased oxidative stress and inflammation, and led to ameliorated lung injury in mice treated with ZnONPs. CONCLUSION This study demonstrated that ZnONPs trigger the activation of the cGAS-STING pathway, which plays an important role in ZnONPs-induced ALI. Inhibition of cGAS with RU.521 mitigates the oxidative stress induced by ZnONPs, suggesting that targeting the cGAS-STING pathway may be a feasible strategy to ameliorate the pulmonary injury caused by nanoparticles.
Collapse
|
102
|
de Moura Rodrigues D, Lacerda-Queiroz N, Couillin I, Riteau N. STING Targeting in Lung Diseases. Cells 2022; 11:3483. [PMID: 36359882 PMCID: PMC9657237 DOI: 10.3390/cells11213483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 01/30/2024] Open
Abstract
The cGAS-STING pathway displays important functions in the regulation of innate and adaptive immunity following the detection of microbial and host-derived DNA. Here, we briefly summarize biological functions of STING and review recent literature highlighting its important contribution in the context of respiratory diseases. Over the last years, tremendous progress has been made in our understanding of STING activation, which has favored the development of STING agonists or antagonists with potential therapeutic benefits. Antagonists might alleviate STING-associated chronic inflammation and autoimmunity. Furthermore, pharmacological activation of STING displays strong antiviral properties, as recently shown in the context of SARS-CoV-2 infection. STING agonists also elicit potent stimulatory activities when used as an adjuvant promoting antitumor responses and vaccines efficacy.
Collapse
Affiliation(s)
- Dorian de Moura Rodrigues
- Experimental and Molecular Immunology and Neurogenetics Laboratory, University of Orleans, Centre National de la Recherche Scientifique (CNRS), UMR7355, 45100 Orleans, France
| | | | - Isabelle Couillin
- Experimental and Molecular Immunology and Neurogenetics Laboratory, University of Orleans, Centre National de la Recherche Scientifique (CNRS), UMR7355, 45100 Orleans, France
| | - Nicolas Riteau
- Experimental and Molecular Immunology and Neurogenetics Laboratory, University of Orleans, Centre National de la Recherche Scientifique (CNRS), UMR7355, 45100 Orleans, France
| |
Collapse
|
103
|
Chen C, Xu P. Cellular functions of cGAS-STING signaling. Trends Cell Biol 2022:S0962-8924(22)00252-5. [DOI: 10.1016/j.tcb.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022]
|
104
|
Jeong MH, Han H, Lagares D, Im H. Recent Advances in Molecular Diagnosis of Pulmonary Fibrosis for Precision Medicine. ACS Pharmacol Transl Sci 2022; 5:520-538. [PMID: 35983278 PMCID: PMC9379941 DOI: 10.1021/acsptsci.2c00028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis is a serious, progressive lung disease characterized by scarring and stiffening lung tissues, affecting the respiratory system and leading to organ failure. It is a complex disease consisting of alveolar damage, chronic inflammation, and a varying degree of lung fibrosis. Significant challenges with pulmonary fibrosis include the lack of effective means to diagnose the disease at early stages, identify patients at higher risks of progress, and assess disease progression and treatment response. Precision medicine powered by accurate molecular profiling and phenotyping could significantly improve our understanding of the disease's heterogeneity, potential biomarkers for diagnosis and prognosis, and molecular targets for treatment development. This Review discusses various translational model systems, including organoids and lung-on-a-chip systems, biomarkers in single cells and extracellular vesicles, and functional pharmacodynamic markers. We also highlight emerging sensing technologies for molecular characterization of pulmonary fibrosis and biomarker detection.
Collapse
Affiliation(s)
- Mi Ho Jeong
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Hongwei Han
- Department
of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts
General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - David Lagares
- Department
of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts
General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hyungsoon Im
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| |
Collapse
|
105
|
Wang S, Wu Q, Chen T, Su R, Pan C, Qian J, Huang H, Yin S, Xie H, Zhou L, Zheng S. Blocking CD47 promotes antitumour immunity through CD103 + dendritic cell-NK cell axis in murine hepatocellular carcinoma model. J Hepatol 2022; 77:467-478. [PMID: 35367532 DOI: 10.1016/j.jhep.2022.03.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS The CD47-signal regulatory protein α (SIRPα) axis inhibits dendritic cell (DC) phagocytosis and contributes to immune evasion. However, the behaviour of DCs and the potential crosstalk between DCs and natural killer (NK) cells in the hepatocellular carcinoma (HCC) microenvironment after CD47 blockade remain unclear. METHODS The infiltration of CD103+ DCs and NK cells were analysed by immunohistochemistry and immunofluorescence in both human and murine HCC specimens. An orthotopic liver tumour model was used to evaluate the function of the CD103+ DC-NK cell axis after CD47 blockade in vivo in wild-type, Rag1-/-, Batf3-/-, and STING1-/- mice. Phagocytosis assays were performed in CD103+ DC and HCC cell lines. CD103+ DC-derived cytokines were analysed by chemokine array. Spleen-derived NK cells in C57BL/6J mice were used to evaluate cytotoxic functions in vitro. RESULTS Higher CD47 expression was associated with worse prognosis in patients with HCC. CD47 blockade enhanced antitumour efficacy by stimulating the CD103+ DC-NK cell axis. The hypoxic microenvironment promoted CD47 blockade-induced tumour DNA phagocytosis by CD103+ DCs. By releasing IL-12 and CXCL9, activated CD103+ DCs induced the recruitment of NK cells with upregulated expression of granzyme B, NKG2D, interferon-γ, and tumour necrosis factor-α and downregulated expression of NKG2A. The antitumour effects of CD47 blockade could be abolished by cyclic GMP-AMP synthase (cGAS)-STING pathway inhibition. CONCLUSIONS In addition to the classical DC-T cell axis, CD47 blockade significantly enhanced the ability of CD103+ DCs to take up tumour DNA, resulting in the stimulation of the cGAS-STING pathway, which promoted the infiltration and activation of NK cells in liver cancer. LAY SUMMARY Hypoxia (low oxygen levels) is prevalent in the hepatocellular carcinoma microenvironment and promotes the phagocytosis (ingestion and elimination) of tumour DNA by CD103+ dendritic cells (a type of immune cell). Blockade of the cell surface protein CD47 resulted in activation of CD103+ dendritic cells which led to the recruitment and activation of natural killer cells (a different immune cell). When activated, these cells exhibit an antitumour effect.
Collapse
Affiliation(s)
- Shuai Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Qinchuan Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Tianchi Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Rong Su
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Caixu Pan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Hechen Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Shengyong Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, China; Key Laboratory of Organ Transplantation, Hangzhou, China.
| |
Collapse
|
106
|
Peng Z, Duan M, Tang Y, Wu J, Zhao K, Zhong Y, He Z, Meng J, Chen F, Xiao X, Wang H, Billiar TR, Lu B, Liang F. Impaired interferon-γ signaling promotes the development of silicosis. iScience 2022; 25:104647. [PMID: 35800765 PMCID: PMC9254453 DOI: 10.1016/j.isci.2022.104647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/18/2022] [Accepted: 06/15/2022] [Indexed: 12/27/2022] Open
Abstract
Silicosis is caused by inhalation of crystalline silica dust particles and known as one of the most serious occupational diseases worldwide. However, little is known about intrinsic factors leading to disease susceptibility. Single-cell sequencing of bronchoalveolar lavage fluid cells of mine workers with silicosis and their co-workers who did not develop silicosis revealed that the impaired interferon (IFN)-γ signaling in myeloid cells was strongly associated with the occurrence of silicosis. Global or myeloid cell-specific deletion of interferon γ receptor (IFN-γR) markedly enhanced the crystalline silica-induced pulmonary injury in wild-type but not in NLRP3 deficient mice. In vitro, IFN-γ priming of macrophages suppressed the crystalline silica-induced NLRP3 inflammasome activation partly by inducing the formation of spacious phagosomes with relatively reduced ratio of crystalline silica/phagosomal areas volumes to resistant crystalline silica-induced lysosomal membrane damage. Thus, these findings provide molecular insights into the intricate mechanisms underlying innate immunity-mediated host responses to environmental irritants.
Collapse
Affiliation(s)
- Zhouyangfan Peng
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Mingwu Duan
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kai Zhao
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Yanjun Zhong
- ICU Center, The Second Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Zhihui He
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Jie Meng
- Department of Respiratory Diseases and Critical Care Illness, The 3Road Xiangya Hospital, Central South University, Changsha 410000, P.R. China
- Hunan Key Laboratory of Organ Fibrosis, Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Fangping Chen
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Xianzhong Xiao
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P.R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Haichao Wang
- The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ben Lu
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P.R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| |
Collapse
|
107
|
Pulmonary Toxicity of Silica Linked to Its Micro- or Nanometric Particle Size and Crystal Structure: A Review. NANOMATERIALS 2022; 12:nano12142392. [PMID: 35889616 PMCID: PMC9318389 DOI: 10.3390/nano12142392] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023]
Abstract
Silicon dioxide (SiO2) is a mineral compound present in the Earth’s crust in two mineral forms: crystalline and amorphous. Based on epidemiological and/or biological evidence, the pulmonary effects of crystalline silica are considered well understood, with the development of silicosis, emphysema, chronic bronchitis, or chronic obstructive pulmonary disease. The structure and capacity to trigger oxidative stress are recognized as relevant determinants in crystalline silica’s toxicity. In contrast, natural amorphous silica was long considered nontoxic, and was often used as a negative control in experimental studies. However, as manufactured amorphous silica nanoparticles (or nanosilica or SiNP) are becoming widely used in industrial applications, these paradigms must now be reconsidered at the nanoscale (<100 nm). Indeed, recent experimental studies appear to point towards significant toxicity of manufactured amorphous silica nanoparticles similar to that of micrometric crystalline silica. In this article, we present an extensive review of the nontumoral pulmonary effects of silica based on in vitro and in vivo experimental studies. The findings of this review are presented both for micro- and nanoscale particles, but also based on the crystalline structure of the silica particles.
Collapse
|
108
|
Lee D, Huntoon K, Kang M, Lu Y, Gallup T, Jiang W, Kim BYS. Harnessing cGAS‐STING Pathway for Cancer Immunotherapy: From Bench to Clinic. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Kristin Huntoon
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Yifei Lu
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Thomas Gallup
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y S Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
109
|
Leinardi R, Longo Sanchez-Calero C, Huaux F. Think Beyond Particle Cytotoxicity: When Self-Cellular Components Released After Immunogenic Cell Death Explain Chronic Disease Development. FRONTIERS IN TOXICOLOGY 2022; 4:887228. [PMID: 35846433 PMCID: PMC9284505 DOI: 10.3389/ftox.2022.887228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
The prolonged perturbation of the immune system following the release of a plethora of self-molecules (known as damage-associated molecular patterns, DAMPs) by stressed or dying cells triggers acute and chronic pathological responses. DAMPs are commonly released after plasma membrane damage or complete rupture due to immunogenic cell death (ICD), upon numerous stressors including infectious and toxic agents. The set of DAMPs released after ICD include mature proinflammatory cytokines and alarmins, but also polymeric macromolecules. These self-intracellular components are recognized by injured and healthy surrounding cells via innate receptors, and induce upregulation of stress-response mechanisms, including inflammation. In this review, by overstepping the simple toxicological evaluation, we apply ICD and DAMP concepts to silica cytotoxicity, providing new insights on the mechanisms driving the progress and/or the exacerbation of certain SiO2–related pathologies. Finally, by proposing self-DNA as new crucial DAMP, we aim to pave the way for the development of innovative and easy-to-perform predictive tests to better identify the hazard of fine and ultrafine silica particles. Importantly, such mechanisms could be extended to nano/micro plastics and diesel particles, providing strategic advice and reports on their health issues.
Collapse
|
110
|
Albez FS, Araz Ö, Yılmazel Uçar E, Alper F, Karaman A, Sağlam L, Akgün M. Long-term follow-up of young denim sandblasters in Turkey. Occup Med (Lond) 2022; 72:403-410. [PMID: 35762845 DOI: 10.1093/occmed/kqac043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Denim sandblasting-induced silicosis is a recently identified occupational disease. AIMS In this study, we aimed to evaluate pulmonary and radiological changes in the long-term follow-up of former denim sandblasters. METHODS Ninety former denim sandblasters were followed from 2007 to 2018. Chest X-rays were evaluated according to the International Labour Organization (ILO) classification. Baseline and final data were compared. Silicosis prevalence, radiological progression and pulmonary dysfunction were evaluated. RESULTS All of the sandblasters were men. Their mean age was 34 ± 5 years, mean follow-up time was 9 ± 2 years (mean time since initial exposure: 17 ± 2 years) and mean duration of exposure was 34 ± 25 months. Rates of radiological progression and decline in pulmonary during follow-up were 63% and 39%, respectively. During follow-up, all patients were diagnosed with silicosis. All workers who were ILO category 0 at baseline (n = 26, 29%) progressed to higher categories. The number of patients in Category 2 doubled and the number of patients in Category 3 increased by 2.5-fold. Eleven patients developed new large opacities and the number of patients with category C opacity increased from 4 to 13. Exposure time was an independent determinant of radiological progression (OR: 1.0, P = 0.036) and decline in pulmonary function (OR: 1.3, P = 0.019). CONCLUSION The prevalence of silicosis in denim sandblasters increases steadily even after exposure is discontinued. Radiological progression was observed in a higher proportion of workers than a decline in lung function. Duration of exposure was the major determinant of disease progression in our study.
Collapse
Affiliation(s)
- F S Albez
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| | - Ö Araz
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| | - E Yılmazel Uçar
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| | - F Alper
- Department of Radiology, Atatürk University School of Medicine, Erzurum, Turkey
| | - A Karaman
- Department of Radiology, Atatürk University School of Medicine, Erzurum, Turkey
| | - L Sağlam
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| | - M Akgün
- Department of Pulmonary Diseases, Atatürk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
111
|
Yang M, Ma YX, Zhi Y, Wang HB, Zhao L, Wang PS, Niu JT. Inhibitors of IFN gene stimulators (STING) improve intestinal ischemia-reperfusion-induced acute lung injury by activating AMPK signaling. Eur J Med Res 2022; 27:79. [PMID: 35642042 PMCID: PMC9153160 DOI: 10.1186/s40001-022-00703-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) caused by intestinal ischemia-reperfusion is a life-threatening disease. Interferon gene stimulator (STING) is a cytoplasmic DNA sensor that participates in the initiation of the inflammatory response. This study aims to establish whether C-176 (STING inhibitor) improves ALI under intestinal ischemia-reperfusion conditions. METHODS To induce ALI, 72 male C57BL/6 mice were subjected to intestinal ischemia for 60 min and reperfusion for 3 h. Through intraperitoneal injection, C-176, a selective STING inhibitor, was injected 30 min before surgical treatment; meanwhile, compound C, an antagonist of adenosine monophosphate-activated protein kinase (AMPK), was administered 30 min after surgery. Based on immunofluorescence and Western blot assays, post-ALI assessments included lung water content (TLW), bronchoalveolar lavage fluid (BALF) protein, H&E staining, Masson staining, pulmonary pyroptosis [Gasdermin-D (GSDMD), cleaved caspase-1], and apoptosis (TUNEL, cleaved caspase-3). RESULTS C-176 administration significantly attenuated intestinal ischemia-reperfusion-mediated ALI; this effect was reflected by exacerbated TLW and BALF protein, aggravated lung injury score, elevated degree of pulmonary fibrosis, increased TUNEL- and GSDMD-positive cells, and upregulated phospho-AMPK, cleaved caspase-1, cleaved caspase-3 and IFNβ mRNA expression. Moreover, C-176 increased phospho-AMPK under ALI conditions. Nonetheless, compound C partially reversed these beneficial effects. CONCLUSION C-176, a selective STING inhibitor, improves intestinal ischemia-reperfusion-mediated ALI, and its underlying mechanism may be associated with AMPK signal activation.
Collapse
Affiliation(s)
- Mei Yang
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China.
| | - Yu-Xia Ma
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| | - Ying Zhi
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| | - Hai-Bin Wang
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| | - Li Zhao
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| | - Peng-Sheng Wang
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| | - Jie-Ting Niu
- Department of Gerontology, Cangzhou Central Hospital, Hebei Medical University, No. 16, Xinhua West Road, Cangzhou, China
| |
Collapse
|
112
|
Singh RS, Vidhyasagar V, Yang S, Arna AB, Yadav M, Aggarwal A, Aguilera AN, Shinriki S, Bhanumathy KK, Pandey K, Xu A, Rapin N, Bosch M, DeCoteau J, Xiang J, Vizeacoumar FJ, Zhou Y, Misra V, Matsui H, Ross SR, Wu Y. DDX41 is required for cGAS-STING activation against DNA virus infection. Cell Rep 2022; 39:110856. [PMID: 35613581 PMCID: PMC9205463 DOI: 10.1016/j.celrep.2022.110856] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 12/27/2022] Open
Abstract
Upon binding double-stranded DNA (dsDNA), cyclic GMP-AMP synthase (cGAS) is activated and initiates the cGAS-stimulator of IFN genes (STING)-type I interferon pathway. DEAD-box helicase 41 (DDX41) is a DEAD-box helicase, and mutations in DDX41 cause myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML). Here, we show that DDX41-knockout (KO) cells have reduced type I interferon production after DNA virus infection. Unexpectedly, activations of cGAS and STING are affected in DDX41 KO cells, suggesting that DDX41 functions upstream of cGAS. The recombinant DDX41 protein exhibits ATP-dependent DNA-unwinding activity and ATP-independent strand-annealing activity. The MDS/AML-derived mutant R525H has reduced unwinding activity but retains normal strand-annealing activity and stimulates greater cGAS dinucleotide-synthesis activity than wild-type DDX41. Overexpression of R525H in either DDX41-deficient or -proficient cells results in higher type I interferon production. Our results have led to the hypothesis that DDX41 utilizes its unwinding and annealing activities to regulate the homeostasis of dsDNA and single-stranded DNA (ssDNA), which, in turn, regulates cGAS-STING activation. cGAS is activated by dsDNA. Singh et al. find DDX41 regulates cGAS activation through unwinding and annealing activities on dsDNA and ssDNA, respectively, and MDS/AML patient mutant R525H causes overactivation of innate immune response due to its unbalanced activities. This DDX41-cGAS-STING pathway may be related to molecular pathogenesis of MDS/AML.
Collapse
Affiliation(s)
- Ravi Shankar Singh
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | | | - Shizhuo Yang
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Ananna Bhadra Arna
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Manisha Yadav
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Aanchal Aggarwal
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Alexya N Aguilera
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Satoru Shinriki
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Kannupriya Pandey
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Aizhang Xu
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - Noreen Rapin
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Mark Bosch
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - John DeCoteau
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jim Xiang
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - Franco J Vizeacoumar
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yan Zhou
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Vikram Misra
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Susan R Ross
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yuliang Wu
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada.
| |
Collapse
|
113
|
Thalidomide Alleviates Pulmonary Fibrosis Induced by Silica in Mice by Inhibiting ER Stress and the TLR4-NF-κB Pathway. Int J Mol Sci 2022; 23:ijms23105656. [PMID: 35628464 PMCID: PMC9144898 DOI: 10.3390/ijms23105656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Silicosis is the most prevalent occupational disease in China. It is a form of pulmonary fibrosis caused by the inhalation of silicon particles. As there is no cure for the potentially lethal and progressive condition, the treatment of silicotic fibrosis is an important and difficult problem to address. Thalidomide, a drug with anti-inflammatory and immunoregulatory properties, has been reported to have lung-protective effects. The purpose of this study was to observe the therapeutic effect of thalidomide on silicotic mice and to determine the protective mechanism. By using silicotic mice models and MH-S cells, we found the expression of endoplasmic reticulum stress (ER stress) and Toll-like receptor 4 (TLR4)-nuclear factor kappa-B (NF-κB) pathway as well as inflammation-related factors were upregulated in the macrophages of silicotic mice. The same indexes were detected in silica-stimulated MH-S cells, and the results were consistent with those in vivo. That is, silica activated ER stress and the TLR4-NF-κB pathway as well as the inflammatory response in vitro. Treating both silicotic mice and silica-stimulated MH-S cells with thalidomide inhibited ER stress and the TLR4-NF-κB pathway as well as the inflammatory response. The present study demonstrates thalidomide as a potential therapeutic agent against silicosis.
Collapse
|
114
|
Radiation-induced non-targeted effect of immunity provoked by mitochondrial DNA damage triggered cGAS/ AIM2 pathways. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
115
|
Early Identification, Accurate Diagnosis, and Treatment of Silicosis. Can Respir J 2022; 2022:3769134. [PMID: 35509892 PMCID: PMC9061058 DOI: 10.1155/2022/3769134] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Silicosis is a global problem, and it has brought about great burdens to society and patients' families. The etiology of silicosis is clear, preventable, and controllable, but the onset is hidden and the duration is long. Thus, it is difficult to diagnose it early and treat it effectively, leaving workers unaware of the consequences of dust exposure. As such, a lack of details in the work history and a slow progression of lung disease contribute to the deterioration of patients until silicosis has advanced to fibrosis. These issues are the key factors impeding the diagnosis and the treatment of silicosis. This article reviews the literature on the early identification, diagnosis, and treatment of silicosis as well as analyzes the difficulties in the diagnosis and the treatment of silicosis and discusses its direction of future development.
Collapse
|
116
|
Lv N, Zhao Y, Liu X, Ye L, Liang Z, Kang Y, Dong Y, Wang W, Kolliputi N, Shi L. Dysfunctional telomeres through mitostress-induced cGAS/STING activation to aggravate immune senescence and viral pneumonia. Aging Cell 2022; 21:e13594. [PMID: 35313074 PMCID: PMC9009109 DOI: 10.1111/acel.13594] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/28/2022] [Accepted: 03/05/2022] [Indexed: 12/16/2022] Open
Abstract
Disproportionately high incidence and mortality of respiratory infection such as influenza A virus (IAV) and SARS-CoV-2 have been evidenced in the elderly, but the role and the mechanism of age-associated immune deregulation in disease exacerbation are not well defined. Using a late generation of mice deficient in telomerase RNA (Terc-/- ), we herein demonstrated that aged mice were exquisitely susceptible to respiratory viral infection, with excessive inflammation and increased mortality. Furthermore, we identified the cGAS/STING pathway, which was essentially induced by the leaked mitochondrial DNA, as a biologically relevant mechanism contributing to exaggerated inflammation in Terc-/- mice following viral infection. Innate immune cells, mainly, macrophages with shortened telomeres, exhibited hallmarks of cellular senescence, mitochondrial distress, and aberrant activation of STING and NLRP3 inflammasome pathways, which predisposed mice to severe viral pneumonia during commonly mild infections. Application of STING inhibitor and, more importantly, senolytic agent, reduced the burden of stressed macrophages, improved mitochondrial integrity, and suppressed STING activation, thereby conferring the protection for Terc-/- mice against respiratory infection. Together, the findings expand our understanding of innate immune senescence and reveal the potential of the senolytics as a promising treatment to alleviate the symptom of viral pneumonia, particularly for the older population.
Collapse
Affiliation(s)
- Nianyin Lv
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yufang Zhao
- Department of Basic Medicine Jiangxi Medical College Nanchang Jiangxi China
| | - Xiaoyi Liu
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Department of Pharmacology and Physiology University of Rochester School of Medicine and Dentistry Rochester New York USA
| | - Lusha Ye
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Zihao Liang
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yanhua Kang
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yeping Dong
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou Zhejiang China
| | - Wei Wang
- Department of Clinical Laboratory the Tongde Hospital Affiliated to Zhejiang TCM University Hangzhou Zhejiang China
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology Department of Internal Medicine Morsani College of Medicine University of South Florida Tampa Florida USA
| | - Liyun Shi
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou Zhejiang China
| |
Collapse
|
117
|
STING Agonists/Antagonists: Their Potential as Therapeutics and Future Developments. Cells 2022; 11:cells11071159. [PMID: 35406723 PMCID: PMC8998017 DOI: 10.3390/cells11071159] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/28/2022] [Accepted: 03/25/2022] [Indexed: 01/07/2023] Open
Abstract
The cGAS STING pathway has received much attention in recent years, and it has been recognized as an important component of the innate immune response. Since the discovery of STING and that of cGAS, many observations based on preclinical models suggest that the faulty regulation of this pathway is involved in many type I IFN autoinflammatory disorders. Evidence has been accumulating that cGAS/STING might play an important role in pathologies beyond classical immune diseases, as in, for example, cardiac failure. Human genetic mutations that result in the activation of STING or that affect the activity of cGAS have been demonstrated as the drivers of rare interferonopathies affecting young children and young adults. Nevertheless, no data is available in the clinics demonstrating the therapeutic benefit in modulating the cGAS/STING pathway. This is due to the lack of STING/cGAS-specific low molecular weight modulators that would be qualified for clinical exploration. The early hopes to learn from STING agonists, which have reached the clinics in recent years for selected oncology indications, have not yet materialized since the initial trials are progressing very slowly. In addition, transforming STING agonists into potent selective antagonists has turned out to be more challenging than expected. Nevertheless, there has been progress in identifying novel low molecular weight compounds, in some cases with unexpected mode of action, that might soon move to clinical trials. This study gives an overview of some of the potential indications that might profit from modulation of the cGAS/STING pathway and a short overview of the efforts in identifying STING modulators (agonists and antagonists) suitable for clinical research and describing their potential as a "drug".
Collapse
|
118
|
STING agonist diABZI induces PANoptosis and DNA mediated acute respiratory distress syndrome (ARDS). Cell Death Dis 2022; 13:269. [PMID: 35338116 PMCID: PMC8953969 DOI: 10.1038/s41419-022-04664-5] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
Stimulator of interferon genes (STING) contributes to immune responses against tumors and may control viral infection including SARS-CoV-2 infection. However, activation of the STING pathway by airway silica or smoke exposure leads to cell death, self-dsDNA release, and STING/type I IFN dependent acute lung inflammation/ARDS. The inflammatory response induced by a synthetic non-nucleotide-based diABZI STING agonist, in comparison to the natural cyclic dinucleotide cGAMP, is unknown. A low dose of diABZI (1 µg by endotracheal route for 3 consecutive days) triggered an acute neutrophilic inflammation, disruption of the respiratory barrier, DNA release with NET formation, PANoptosis cell death, and inflammatory cytokines with type I IFN dependent acute lung inflammation. Downstream upregulation of DNA sensors including cGAS, DDX41, IFI204, as well as NLRP3 and AIM2 inflammasomes, suggested a secondary inflammatory response to dsDNA as a danger signal. DNase I treatment, inhibition of NET formation together with an investigation in gene-deficient mice highlighted extracellular DNA and TLR9, but not cGAS, as central to diABZI-induced neutrophilic response. Therefore, activation of acute cell death with DNA release may lead to ARDS which may be modeled by diABZI. These results show that airway targeting by STING activator as a therapeutic strategy for infection may enhance lung inflammation with severe ARDS. STING agonist diABZI induces neutrophilic lung inflammation and PANoptosis A, Airway STING priming induce a neutrophilic lung inflammation with epithelial barrier damage, double-stranded DNA release in the bronchoalvelolar space, cell death, NETosis and type I interferon release. B, 1. The diamidobenzimidazole (diABZI), a STING agonist is internalized into the cytoplasm through unknown receptor and induce the activation and dimerization of STING followed by TBK1/IRF3 phosporylation leading to type I IFN response. STING activation also leads to NF-kB activation and the production of pro-inflammatory cytokines TNFα and IL-6. 2. The activation of TNFR1 and IFNAR1 signaling pathway results in ZBP1 and RIPK3/ASC/CASP8 activation leading to MLKL phosphorylation and necroptosis induction. 3. This can also leads to Caspase-3 cleavage and apoptosis induction. 4. Self-dsDNA or mtDNA sensing by NLRP3 or AIM2 induces inflammsome formation leading to Gasdermin D cleavage enabling Gasdermin D pore formation and the release mature IL-1β and pyroptosis. NLRP3 inflammasome formation can be enhanced by the ZBP1/RIPK3/CASP8 complex. 5. A second signal of STING activation with diABZI induces cell death and the release of self-DNA which is sensed by cGAS and form 2′3′-cGAMP leading to STING hyper activation, the amplification of TBK1/IRF3 and NF-kB pathway and the subsequent production of IFN-I and inflammatory TNFα and IL-6. This also leads to IFI204 and DDX41 upregulation thus, amplifying the inflammatory loop. The upregulation of apoptosis, pyroptosis and necroptosis is indicative of STING-dependent PANoptosis. ![]()
Collapse
|
119
|
Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev 2022; 122:5977-6039. [PMID: 35107989 PMCID: PMC8994686 DOI: 10.1021/acs.chemrev.1c00750] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The stimulator of interferon genes (STING) cellular signaling pathway is a promising target for cancer immunotherapy. Activation of the intracellular STING protein triggers the production of a multifaceted array of immunostimulatory molecules, which, in the proper context, can drive dendritic cell maturation, antitumor macrophage polarization, T cell priming and activation, natural killer cell activation, vascular reprogramming, and/or cancer cell death, resulting in immune-mediated tumor elimination and generation of antitumor immune memory. Accordingly, there is a significant amount of ongoing preclinical and clinical research toward further understanding the role of the STING pathway in cancer immune surveillance as well as the development of modulators of the pathway as a strategy to stimulate antitumor immunity. Yet, the efficacy of STING pathway agonists is limited by many drug delivery and pharmacological challenges. Depending on the class of STING agonist and the desired administration route, these may include poor drug stability, immunocellular toxicity, immune-related adverse events, limited tumor or lymph node targeting and/or retention, low cellular uptake and intracellular delivery, and a complex dependence on the magnitude and kinetics of STING signaling. This review provides a concise summary of the STING pathway, highlighting recent biological developments, immunological consequences, and implications for drug delivery. This review also offers a critical analysis of an expanding arsenal of chemical strategies that are being employed to enhance the efficacy, safety, and/or clinical utility of STING pathway agonists and lastly draws attention to several opportunities for therapeutic advancements.
Collapse
Affiliation(s)
- Kyle M Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - Taylor L Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
| |
Collapse
|
120
|
Zhao X, Zhou L, Kou Y, Kou J. Activated neutrophils in the initiation and progression of COVID-19: hyperinflammation and immunothrombosis in COVID-19. Am J Transl Res 2022; 14:1454-1468. [PMID: 35422922 PMCID: PMC8991139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic respiratory disease caused by a novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-COV-2). COVID-19 is typically associated with fever and influenza-like symptoms in its early stages. Severe cases progress to acute respiratory distress syndrome/acute lung injury (ARDS/ALI), multiple organ damage, and even death. Until now, there has been a lack of specific and definitive treatment for COVID-19, which further challenges the situation. Previous clinical and laboratory data showed that neutrophils were significantly decreased in patients who died from COVID-19 in the early stages of disease; when patients were admitted to the hospital the number of neutrophils increased dramatically from 7 to 14 days after admission, which is correlated to myocardial and liver injury, thromboembolic complications, and poor prognosis. Autopsy findings revealed abundant neutrophil infiltration in the pulmonary capillaries and exudation into the alveolar cavity. Therefore, we speculate that neutrophils may play an important role in the initiation and progression of COVID-19. In this review, the relationship among the dynamic changes in neutrophils, cytokine storms, and the release of neutrophil extracellular traps (NETs) with the progression of COVID-19 was elucidated in detail. With a better understanding of the pathogenic mechanisms this can lead to improved clinical applications which are identified and discussed in this review.
Collapse
Affiliation(s)
- Xinyi Zhao
- Department of Cardiology of The Second Hospital, Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Lijin Zhou
- Department of Cardiology of The Second Hospital, Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Yan Kou
- Department of Cardiology of The Second Hospital, Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Junjie Kou
- Department of Cardiology of The Second Hospital, Harbin Medical University Harbin 150001, Heilongjiang, China
| |
Collapse
|
121
|
Shi W, Hao J, Wu Y, Liu C, Shimizu K, Li R, Zhang C. Protective effects of heterophyllin B against bleomycin-induced pulmonary fibrosis in mice via AMPK activation. Eur J Pharmacol 2022; 921:174825. [PMID: 35283110 DOI: 10.1016/j.ejphar.2022.174825] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/28/2022]
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disease with unknown etiology. In the present study, we evaluated the anti-fibrotic effects of heterophyllin B, a natural product from Radix Pseudostellariae having anti-inflammatory and tyrosinase inhibitory activities. In bleomycin (BLM)-induced PF mouse model, heterophyllin B treatments (5 or 20 mg/kg/d) significantly attenuated BLM-induced alveolar cavity collapse, inflammatory cell infiltration, alveolar wall thickening and collagen deposition. When compared to model group, heterophyllin B treatments also increased adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation levels by 359% (P < 0.001) and reduced the expression of stimulator of interferon genes (STING) by 73% (P < 0.001). Furthermore, co-administration of heterophyllin B with AMPK inhibitor dorsomorphin (Compound C) significantly blocked the improvement effects of heterophyllin B on BLM-damaged lung tissue, and also increased the protein expression of STING which was inhibited by heterophyllin B in fibrotic lungs (P < 0.001). It is known that alveolar epithelia and lung fibroblasts exert prominent roles in the fibrosis progression. In the present study we found that, in vitro, heterophyllin B significantly inhibited alveolar epithelial mesenchymal transition (EMT) and lung fibroblast transdifferentiation. We also found that the inhibition of heterophyllin B on lung fibroblast transdifferentiation and STING expression was reversed by Compound C. To summarize, heterophyllin B exhibited protective effects on BLM-induced lung fibrosis potentially by inhibiting TGF-Smad2/3 signalings and AMPK-mediated STING signalings.
Collapse
Affiliation(s)
- Wen Shi
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiatong Hao
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Yanliang Wu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Chang Liu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Kuniyoshi Shimizu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China; Department of Forest and Forest Products Sciences, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | - Renshi Li
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
122
|
Yang Y, Huang Y, Zeng Z. Advances in cGAS-STING Signaling Pathway and Diseases. Front Cell Dev Biol 2022; 10:800393. [PMID: 35186921 PMCID: PMC8851069 DOI: 10.3389/fcell.2022.800393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Pathogens can produce conserved pathogen-associated molecular patterns (PAMPs) after invading the body, which can be specifically recognized by host pattern recognition receptors (PRRs). In recent years, it has been found that cytoplasmic DNA receptors recognize exogenous DNA inducing activation of interferon 1 (IFN1), which is a rapid advance in various research areas. The cyclic GMP–AMP synthase (cGAS) stimulator of interferon gene (STING) signaling pathway is a critical natural immune pathway in cells. Early studies revealed that it plays a crucial regulatory role in pathogen infection and tumor, and it is associated with various human autoimmune diseases. Recently studies have found that activation of cGAS-STING signaling pathway is related to different organ injuries. The present review elaborates on the regulation of the cGAS-STING signaling pathway and its role in various diseases, aiming to provide a theoretical basis for immunotherapy targeting this pathway.
Collapse
|
123
|
Whalen W, Buyukozkan M, Moore B, Moon JS, Dela Cruz CS, Martinez FJ, Choi AMK, Krumsiek J, Stout-Delgado H, Cho SJ. Association of circulating cell-free double-stranded DNA and metabolic derangements in idiopathic pulmonary fibrosis. Thorax 2022; 77:186-190. [PMID: 34521729 PMCID: PMC8758529 DOI: 10.1136/thoraxjnl-2021-217315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/26/2021] [Indexed: 01/05/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with unclear aetiology and poorly understood pathophysiology. Although plasma levels of circulating cell-free DNA (ccf-DNA) and metabolomic changes have been reported in IPF, the associations between ccf-DNA levels and metabolic derangements in lung fibrosis are unclear. Here, we demonstrate that ccf-double-stranded DNA (dsDNA) is increased in patients with IPF with rapid progression of disease compared with slow progressors and healthy controls and that ccf-dsDNA associates with amino acid metabolism, energy metabolism and lipid metabolism pathways in patients with IPF.
Collapse
Affiliation(s)
- William Whalen
- Department of Medicine, Pulmonary and Critical Care, Weill Cornell Medicine, New York, New York, USA
| | - Mustafa Buyukozkan
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Bethany Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Chungcheongnam-do, The Republic of Korea
| | - Charles S Dela Cruz
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Pulmonary Critical Care, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Fernando J Martinez
- Department of Medicine, Pulmonary and Critical Care, Weill Cornell Medicine, New York, New York, USA
| | - Augustine M K Choi
- Department of Medicine, Pulmonary and Critical Care, Weill Cornell Medicine, New York, New York, USA
| | - Jan Krumsiek
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Heather Stout-Delgado
- Department of Medicine, Pulmonary and Critical Care, Weill Cornell Medicine, New York, New York, USA
| | - Soo Jung Cho
- Department of Medicine, Pulmonary and Critical Care, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
124
|
David C, Frémond ML. Lung Inflammation in STING-Associated Vasculopathy with Onset in Infancy (SAVI). Cells 2022; 11:318. [PMID: 35159128 PMCID: PMC8834229 DOI: 10.3390/cells11030318] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/15/2022] Open
Abstract
STING-associated vasculopathy with onset in infancy (SAVI) is a type I interferonopathy caused by gain-of-function mutations in STING1 encoding stimulator of interferon genes (STING) protein. SAVI is characterized by severe inflammatory lung disease, a feature not observed in previously described type I interferonopathies i.e., Mendelian autoinflammatory disorders defined by constitutive activation of the type I interferon (IFN) pathway. Molecular defects in nucleic acid metabolism or sensing are central to the pathophysiology of these diseases, with such defects occurring at any step of the tightly regulated pathway of type I IFN production and signaling (e.g., exonuclease loss of function, RNA-DNA hybrid accumulation, constitutive activation of adaptor proteins such as STING). Among over 30 genotypes, SAVI and COPA syndrome, whose pathophysiology was recently linked to a constitutive activation of STING signaling, are the only type I interferonopathies presenting with predominant lung involvement. Lung disease is the leading cause of morbidity and mortality in these two disorders which do not respond to conventional immunosuppressive therapies and only partially to JAK1/2 inhibitors. In human silicosis, STING-dependent sensing of self-DNA following cell death triggered by silica exposure has been found to drive lung inflammation in mice and human models. These recent findings support a key role for STING and nucleic acid sensing in the homeostasis of intrinsic pulmonary inflammation. However, mechanisms by which monogenic defects in the STING pathway lead to pulmonary damages are not yet fully elucidated, and an improved understanding of such mechanisms is fundamental to improved future patient management. Here, we review the recent insights into the pathophysiology of SAVI and outline our current understanding of self-nucleic acid-mediated lung inflammation in humans.
Collapse
Affiliation(s)
- Clémence David
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Marie-Louise Frémond
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
- Paediatric Immunology-Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP.Centre-Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| |
Collapse
|
125
|
Jaiswal AK, Yadav J, Makhija S, Mazumder S, Mitra AK, Suryawanshi A, Sandey M, Mishra A. Irg1/itaconate metabolic pathway is a crucial determinant of dendritic cells immune-priming function and contributes to resolute allergen-induced airway inflammation. Mucosal Immunol 2022; 15:301-313. [PMID: 34671116 PMCID: PMC8866123 DOI: 10.1038/s41385-021-00462-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Itaconate is produced from the mitochondrial TCA cycle enzyme aconitase decarboxylase (encoded by immune responsive gene1; Irg1) that exerts immunomodulatory function in myeloid cells. However, the role of the Irg1/itaconate pathway in dendritic cells (DC)-mediated airway inflammation and adaptive immunity to inhaled allergens, which are the primary antigen-presenting cells in allergic asthma, remains largely unknown. House dust mite (HDM)-challenged Irg1-/- mice displayed increases in eosinophilic airway inflammation, mucous cell metaplasia, and Th2 cytokine production with a mechanism involving impaired mite antigen presentations by DC. Adoptive transfer of HDM-pulsed DC from Irg1-deficient mice into naïve WT mice induced a similar phenotype of elevated type 2 airway inflammation and allergic sensitization. Untargeted metabolite analysis of HDM-pulsed DC revealed itaconate as one of the most abundant polar metabolites that potentially suppress mitochondrial oxidative damage. Furthermore, the immunomodulatory effect of itaconate was translated in vivo, where intranasal administration of 4-octyl itaconate 4-OI following antigen priming attenuated the manifestations of HDM-induced airway disease and Th2 immune response. Taken together, these data demonstrated for the first time a direct regulatory role of the Irg1/itaconate pathway in DC for the development of type 2 airway inflammation and suggest a possible therapeutic target in modulating allergic asthma.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Jyoti Yadav
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Sangeet Makhija
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Suman Mazumder
- grid.252546.20000 0001 2297 8753Department of Drug Discovery and Development, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, AL USA
| | - Amit Kumar Mitra
- grid.252546.20000 0001 2297 8753Department of Drug Discovery and Development, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, AL USA
| | - Amol Suryawanshi
- grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Maninder Sandey
- grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Amarjit Mishra
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| |
Collapse
|
126
|
Zhang Z, Zhou J, Verma V, Liu X, Wu M, Yu J, Chen D. Crossed Pathways for Radiation-Induced and Immunotherapy-Related Lung Injury. Front Immunol 2021; 12:774807. [PMID: 34925345 PMCID: PMC8672113 DOI: 10.3389/fimmu.2021.774807] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Radiation-induced lung injury (RILI) is a form of radiation damage to normal lung tissue caused by radiotherapy (RT) for thoracic cancers, which is most commonly comprised of radiation pneumonitis (RP) and radiation pulmonary fibrosis (RPF). Moreover, with the widespread utilization of immunotherapies such as immune checkpoint inhibitors as first- and second-line treatments for various cancers, the incidence of immunotherapy-related lung injury (IRLI), a severe immune-related adverse event (irAE), has rapidly increased. To date, we know relatively little about the underlying mechanisms and signaling pathways of these complications. A better understanding of the signaling pathways may facilitate the prevention of lung injury and exploration of potential therapeutic targets. Therefore, this review provides an overview of the signaling pathways of RILI and IRLI and focuses on their crosstalk in diverse signaling pathways as well as on possible mechanisms of adverse events resulting from combined radiotherapy and immunotherapy. Furthermore, this review proposes potential therapeutic targets and avenues of further research based on signaling pathways. Many new studies on pyroptosis have renewed appreciation for the value and importance of pyroptosis in lung injury. Therefore, the authors posit that pyroptosis may be the common downstream pathway of RILI and IRLI; discussion is also conducted regarding further perspectives on pyroptosis as a crucial signaling pathway in lung injury treatment.
Collapse
Affiliation(s)
- Zengfu Zhang
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jialin Zhou
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Vivek Verma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xu Liu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Meng Wu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Dawei Chen
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
127
|
Bao T, Liu J, Leng J, Cai L. The cGAS-STING pathway: more than fighting against viruses and cancer. Cell Biosci 2021; 11:209. [PMID: 34906241 PMCID: PMC8670263 DOI: 10.1186/s13578-021-00724-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/02/2021] [Indexed: 01/07/2023] Open
Abstract
In the classic Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway, downstream signals can control the production of type I interferon and nuclear factor kappa-light-chain-enhancer of activated B cells to promote the activation of pro-inflammatory molecules, which are mainly induced during antiviral responses. However, with progress in this area of research, studies focused on autoimmune diseases and chronic inflammatory conditions that may be relevant to cGAS-STING pathways have been conducted. This review mainly highlights the functions of the cGAS-STING pathway in chronic inflammatory diseases. Importantly, the cGAS-STING pathway has a major impact on lipid metabolism. Different research groups have confirmed that the cGAS-STING pathway plays an important role in the chronic inflammatory status in various organs. However, this pathway has not been studied in depth in diabetes and diabetes-related complications. Current research on the cGAS-STING pathway has shown that the targeted therapy of diseases that may be caused by inflammation via the cGAS-STING pathway has promising outcomes.
Collapse
Affiliation(s)
- Terigen Bao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
- Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA
- Departments of Pharmacology and Toxicology, The University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
128
|
Wu Q, Jiao B, Gui W, Zhang Q, Wang F, Han L. Long non-coding RNA SNHG1 promotes fibroblast-to-myofibroblast transition during the development of pulmonary fibrosis induced by silica particles exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112938. [PMID: 34741930 DOI: 10.1016/j.ecoenv.2021.112938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 06/13/2023]
Abstract
Inhaling silica dust in the environment can cause progressive pulmonary fibrosis, then silicosis. Silicosis is the most harmful occupational disease in the world, so the study of the mechanism is of great significance for the prevention and treatment of silicosis. Long non-coding RNAs (lncRNAs) are important players in the pathological process of fibrotic diseases. However, the function of specific lncRNA in regulating pulmonary fibrosis remains elusive. In this study, a mouse model of pulmonary fibrosis via intratracheal instillation of silica particles was established, and the differential expression of lnc-SNHG1 and miR-326 in lung tissues and TGF-β1-treated fibroblasts was detected by the qRT-PCR method. Short interfering RNA (siRNA) and plasmid were designed for knockdown or overexpression of lnc-SNHG1 in fibroblasts. MiRNA simulant was designed for overexpression of miR-326 in vivo and in vitro. Dual-luciferase reporter system, immunofluorescence, western blot, wound healing and transwell assay were performed to investigate the function and the underlying mechanisms of lnc-SNHG1. As a result, we found that lnc-SNHG1 was highly expressed in fibrotic lung tissues of mice and TGF-β1-treated fibroblasts. Moreover, the high expression of lnc-SNHG1 facilitated the migration and invasion of fibroblasts and the secretion of fibrotic molecules, while the low expression of lnc-SNHG1 exerted the opposite effects. Further mechanism studies showed that miR-326 was the potential target of lnc-SNHG1, and there is a negative correlation between the expression levels of lnc-SNHG1 and miR-326. Combined with mitigating fibrotic effects of miR-326 in a mouse model of silica particles exposure, we revealed that lnc-SNHG1 significantly sponged miR-326 and facilitated the expression of SP1, thus accelerating fibroblast-to-myofibroblast transition and synergistically promoting the development of pulmonary fibrosis. Our study uncovered a key mechanism by which lnc-SNHG1 regulated pulmonary fibrosis through miR-326/SP1 axis, and lnc-SNHG1 is a potential target for the prevention and treatment of silicosis.
Collapse
Affiliation(s)
- Qiuyun Wu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou 221004, China.
| | - Biyang Jiao
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenwen Gui
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Qianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Feng Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Lei Han
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| |
Collapse
|
129
|
Schuliga M, Kanwal A, Read J, Blokland KEC, Burgess JK, Prêle CM, Mutsaers SE, Grainge C, Thomson C, James A, Bartlett NW, Knight DA. A cGAS-dependent response links DNA damage and senescence in alveolar epithelial cells: a potential drug target in IPF. Am J Physiol Lung Cell Mol Physiol 2021; 321:L859-L871. [PMID: 34524912 DOI: 10.1152/ajplung.00574.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Alveolar epithelial cell (AEC) senescence is implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Mitochondrial dysfunction including release of mitochondrial DNA (mtDNA) is a feature of senescence, which led us to investigate the role of the DNA-sensing guanine monophosphate-adenine monophosphate (GMP-AMP) synthase (cGAS) in IPF, with a focus on AEC senescence. cGAS expression in fibrotic tissue from lungs of patients with IPF was detected within cells immunoreactive for epithelial cell adhesion molecule (EpCAM) and p21, epithelial and senescence markers, respectively. Submerged primary cultures of AECs isolated from lung tissue of patients with IPF (IPF-AECs, n = 5) exhibited higher baseline senescence than AECs from control donors (Ctrl-AECs, n = 5-7), as assessed by increased nuclear histone 2AXγ phosphorylation, p21 mRNA, and expression of senescence-associated secretory phenotype (SASP) cytokines. Pharmacological cGAS inhibition using RU.521 diminished IPF-AEC senescence in culture and attenuated induction of Ctrl-AEC senescence following etoposide-induced DNA damage. Short interfering RNA (siRNA) knockdown of cGAS also attenuated etoposide-induced senescence of the AEC line, A549. Higher levels of mtDNA were detected in the cytosol and culture supernatants of primary IPF- and etoposide-treated Ctrl-AECs when compared with Ctrl-AECs at baseline. Furthermore, ectopic mtDNA augmented cGAS-dependent senescence of Ctrl-AECs, whereas DNAse I treatment diminished IPF-AEC senescence. This study provides evidence that a self-DNA-driven, cGAS-dependent response augments AEC senescence, identifying cGAS as a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Amama Kanwal
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Kaj E C Blokland
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,Department of Pathology and Medical Biology, Groningen Research Institute of Asthma and COPD and KOLFF Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Department of Pathology and Medical Biology, Groningen Research Institute of Asthma and COPD and KOLFF Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cecilia M Prêle
- Institute for Respiratory Health, University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Christopher Grainge
- Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia.,John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Claire Thomson
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia.,St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Allen James
- John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
130
|
Abstract
Innate immunity is regulated by a broad set of evolutionary conserved receptors to finely probe the local environment and maintain host integrity. Besides pathogen recognition through conserved motifs, several of these receptors also sense aberrant or misplaced self-molecules as a sign of perturbed homeostasis. Among them, self-nucleic acid sensing by the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway alerts on the presence of both exogenous and endogenous DNA in the cytoplasm. We review recent literature demonstrating that self-nucleic acid detection through the STING pathway is central to numerous processes, from cell physiology to sterile injury, auto-immunity and cancer. We address the role of STING in autoimmune diseases linked to dysfunctional DNAse or related to mutations in DNA sensing pathways. We expose the role of the cGAS/STING pathway in inflammatory diseases, neurodegenerative conditions and cancer. Connections between STING in various cell processes including autophagy and cell death are developed. Finally, we review proposed mechanisms to explain the sources of cytoplasmic DNA.
Collapse
Affiliation(s)
| | - Nicolas Riteau
- Experimental and Molecular Immunology and Neurogenetics Laboratory (INEM), Centre National de la Recherche Scientifique (CNRS), UMR7355 and University of Orleans, Orleans, France
| |
Collapse
|
131
|
Rauwolf S, Bag S, Rouqueiro R, Schwaminger SP, Dias-Cabral AC, Berensmeier S, Wenzel W. Insights on Alanine and Arginine Binding to Silica with Atomic Resolution. J Phys Chem Lett 2021; 12:9384-9390. [PMID: 34551250 DOI: 10.1021/acs.jpclett.1c02398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Interactions of biomolecules with inorganic oxide surfaces such as silica in aqueous solutions are of profound interest in various research fields, including chemistry, biotechnology, and medicine. While there is a general understanding of the dominating electrostatic interactions, the binding mechanism is still not fully understood. Here, chromatographic zonal elution and flow microcalorimetry experiments were combined with molecular dynamic simulations to describe the interaction of different capped amino acids with the silica surface. We demonstrate that ion pairing is the dominant electrostatic interaction. Surprisingly, the interaction strength is more dependent on the repulsive carboxy group than on the attracting amino group. These findings are essential for conducting experimental and simulative studies on amino acids when transferring the results to biomolecule-surface interactions.
Collapse
Affiliation(s)
- Stefan Rauwolf
- Department Mechanical Engineering, Bioseparation Engineering Group, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching, Germany
| | - Saientan Bag
- Institute for Nanotechnology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Rodrigo Rouqueiro
- Department of Chemistry, CICS-UBI Health Science Research Center, University Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Sebastian Patrick Schwaminger
- Department Mechanical Engineering, Bioseparation Engineering Group, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching, Germany
| | - Ana Cristina Dias-Cabral
- Department of Chemistry, CICS-UBI Health Science Research Center, University Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Sonja Berensmeier
- Department Mechanical Engineering, Bioseparation Engineering Group, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching, Germany
| | - Wolfgang Wenzel
- Institute for Nanotechnology, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
132
|
Zhang Y, Huang S, Tan S, Chen M, Yang S, Chen S. 3-methyadenine inhibits lipopolysaccharides-induced pulmonary inflammation at the early stage of silicosis via blocking NF-κB signaling pathway. Toxicol Ind Health 2021; 37:662-673. [PMID: 34565256 DOI: 10.1177/07482337211039426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Occupational exposure to silica dust is related to pulmonary inflammation and silicosis. Lipopolysaccharides (LPSs) could aggravate apoptosis in alveolar macrophages (AMs) of human silicosis through autophagy, yet how the reduction of autophagy attenuated LPS-induced lung injury and the related mechanisms need to be investigated. In the study, we aim to understand the role of 3-methyladenine (3-MA), an inhibitor of autophagy, in LPS-mediated inflammatory responses and fibrosis. We collected AMs from observers/silicosis patients. The results showed that LPS induced NF-κB-related pulmonary inflammation in observers and silicosis patients, as confirmed by an increase in the expression of IL-1β, IL-6, TNF-α, and p65, which could be inhibited by 3-MA treatment. In mice models, at the early stage (7d) of silicosis, but not the late (28d) stage, blocking autophagy reversed the increased levels of IL-1β, IL-6, TNF-α, and p65 caused by LPS. Mechanism study revealed that LPS triggered the expression of LC3 II, p62, and cleaved caspase-3 at the early stage exposed to silica, which could be restored by 3-MA, while there was no difference in the expression of LAMP1 either at the early or late stage of silicosis in different groups. Similarly, 3-MA treatment did not prevent fibrosis characterized by destroyed alveoli, collagen deposition, and increased expression of α-SMA and Col-1 induced by LPS at the late stage of silicosis. The results suggested that 3-MA has a role in the protection of lung injury at the early stage of silicosis and provided an experimental basis for preventive strategies of pulmonary inflammation and silicosis.
Collapse
Affiliation(s)
- Yujing Zhang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shuai Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shiyi Tan
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Mingke Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shang Yang
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| | - Shi Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, 12568Hunan Normal University, Changsha, Hunan Province, China
| |
Collapse
|
133
|
Wang N, Wang X, Li Y, Shen H, Liu Z, Ma Z, Li Q, Zhao M. The STING-IRF3 pathway contributes to paraquat-induced acute lung injury. Toxicol Mech Methods 2021; 32:145-157. [PMID: 34455893 DOI: 10.1080/15376516.2021.1974133] [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: 10/20/2022]
Abstract
Immune and inflammatory responses play significant roles in paraquat (PQ)-induced acute lung injury (ALI), but the specific mechanisms remain unclear. Our study aimed to investigate the action of STING-IRF3 signaling on PQ-induced ALI in mice. Following PQ administration, samples were collected at 2, 12, 24, and 48 h for in vivo studies, and 24 h for in vitro studies. Following PQ administration (30 mg/kg, i.p.), injury to mouse lungs was evaluated by H&E staining and wet/dry ratios, and lung oxidative damage was evaluated by MDA and SOD assays. The mRNA levels of Sting, Irf3, and Ifnβ were detected by RT-PCR, the expression of STING and IRF3 were assessed by western blotting and IHC/IF, and the secretion of IFNβ was detected by ELISA. In vivo, PQ administration induced pathological changes and increased wet/dry ratios in lungs after 48 h. Sting, Irf3, and Ifnβ mRNA levels in lung tissues, STING and pIRF3 protein levels in lung tissues, and IFNβ secretion in serum, were upregulated by PQ in a time-dependent manner. PQ administration promoted IRF3 nuclear translocation in lung tissues after 48 h. The above changes were all attenuated by dexamethasone treatment (5 mg/kg, i.p., qd). In vitro, PQ induced STING and IRF3 translocation. Irf3 or Sting silencing decreased the mRNA levels and supernatant secretion of IFNβ in PQ-treated RAW264.7 mouse macrophages. Sting silencing also inhibited the protein and mRNA levels of IRF3 in vitro. Our study suggests that STING-IRF3 signaling contributes to PQ-induced ALI, providing new information for future treatment strategies.
Collapse
Affiliation(s)
- Na Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Occupational Disease and Occupational Health Prevention and Control Institute, Liaoning Center for Disease Control and Prevention, Shenyang, Liaoning, China
| | - Xiaofeng Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuhua Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haitao Shen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhenning Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongliang Ma
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Min Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
134
|
Seo SU, Jeong JH, Baek BS, Choi JM, Choi YS, Ko HJ, Kweon MN. Bleomycin-Induced Lung Injury Increases Resistance to Influenza Virus Infection in a Type I Interferon-Dependent Manner. Front Immunol 2021; 12:697162. [PMID: 34484196 PMCID: PMC8416411 DOI: 10.3389/fimmu.2021.697162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
Acute lung injury (ALI) results in acute respiratory disease that causes fatal respiratory diseases; however, little is known about the incidence of influenza infection in ALI. Using a ALI-mouse model, we investigated the pro-inflammatory cytokine response to ALI and influenza infection. Mice treated with bleomycin (BLM), which induces ALI, were more resistant to influenza virus infection and exhibited higher levels of type I interferon (IFN-I) transcription during the early infection period than that in PBS-treated control mice. BLM-treated mice also exhibited a lower viral burden, reduced pro-inflammatory cytokine production, and neutrophil levels. In contrast, BLM-treated IFN-I receptor 1 (IFNAR1)-knockout mice failed to show this attenuated phenotype, indicating that IFN-I is key to the antiviral response in ALI-induced mice. The STING/TBK1/IRF3 pathway was found to be involved in IFN-I production and the establishment of an antiviral environment in the lung. The depletion of plasmacytoid dendritic cells (pDCs) reduced the effect of BLM treatment against influenza virus infection, suggesting that pDCs are the major source of IFN-I and are crucial for defense against viral infection in BLM-induced lung injury. Overall, this study showed that BLM-mediated ALI in mice induced the release of double-stranded DNA, which in turn potentiated IFN-I-dependent pulmonary viral resistance by activating the STING/TBK1/IRF3 pathway in association with pDCs.
Collapse
Affiliation(s)
- Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae-Hyeon Jeong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Bum-Seo Baek
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, South Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, South Korea
| |
Collapse
|
135
|
Han B, Wang X, Wu P, Jiang H, Yang Q, Li S, Li J, Zhang Z. Pulmonary inflammatory and fibrogenic response induced by graphitized multi-walled carbon nanotube involved in cGAS-STING signaling pathway. JOURNAL OF HAZARDOUS MATERIALS 2021; 417:125984. [PMID: 34020360 DOI: 10.1016/j.jhazmat.2021.125984] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 04/09/2021] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
Graphitized multi-walled carbon nanotubes (GMWCNTs) are a new type of nanomaterial. Recently, their production and application in biological medicine have grown rapidly. However, GMWCNTs may cause adverse health effects, including the common occupational disease of pulmonary fibrosis. Pulmonary fibrosis is a serious progressive disease that often leads to lung failure, high mortality, and disability, and there is no effective therapy currently available. Therefore, identifying new biomarkers of the disease is important to better understand the disease mechanisms and explore new therapeutic strategies. In this study, 40 μg of GMWCNTs was used to treat mice in vivo by pharyngeal aspiration, and different genes were screened by transcriptome sequencing. Activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) signal pathway had an important effect on the development of pulmonary inflammation and fibrosis. GMWCNTs were then administered to the mice with a STING inhibitor (C-176). Inhibition of STING effectively decreased pulmonary inflammation and fibrosis in mice induced by GMWCNTs. Collectively, activation of the cGAS-STING signaling pathway is involved in GMWCNT-induced pulmonary inflammation and fibrosis in mice.
Collapse
Affiliation(s)
- Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Huijie Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China.
| |
Collapse
|
136
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
137
|
Wu X, Kang X, Zhang X, Xie W, Su Y, Liu X, Guo L, Guo E, Li F, Hu D, Qin X, Fu Y, Peng W, Jia J, Wang C. WEE1 inhibitor and ataxia telangiectasia and RAD3-related inhibitor trigger stimulator of interferon gene-dependent immune response and enhance tumor treatment efficacy through programmed death-ligand 1 blockade. Cancer Sci 2021; 112:4444-4456. [PMID: 34382294 PMCID: PMC8586668 DOI: 10.1111/cas.15108] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/28/2022] Open
Abstract
WEE1 plays an important role in the regulation of cell cycle G2/M checkpoints and DNA damage response (DDR). Inhibition of WEE1 can increase the instability of the genome and have anti–tumor effects in some solid tumors. However, it has certain limitations for multiple cancer cells from different lineages. Therefore, we consider the use of synthetic lethal interactions to enhance the therapeutic effect. Our experiments proved that WEE1 inhibitor (WEE1i) can activate the ataxia telangiectasia and RAD3‐related (ATR) pathway and that blockage of ATR dramatically sensitized the WEE1i‐induced cell death. The tumor‐selective synthetic lethality between bioavailable WEE1 and ATR inhibitors led to tumor remission in vivo. Mechanistically, the combination promoted the accumulation of cytosolic double‐strand DNA, which subsequently activated the stimulator of the interferon gene (STING) pathway and induced the production of type I interferon and CD8+ T cells, thereby inducing anti–tumor immunity. Furthermore, our study found that immune checkpoint programmed death‐ligand 1 is upregulated by the combination therapy, and blocking PD‐L1 further enhances the effect of the combination therapy. In summary, as an immunomodulator, the combination of WEE1i with ATR inhibitor (ATRi) and immune checkpoint blockers provides a potential new approach for cancer treatment.
Collapse
Affiliation(s)
- Xue Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Kang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Xie
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Su
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ensong Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuxia Li
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Dianxing Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Qin
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenju Peng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiedong Jia
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Changyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
138
|
Li L, Qi R, Zhang L, Yu Y, Hou J, Gu Y, Song D, Wang X. Potential biomarkers and targets of mitochondrial dynamics. Clin Transl Med 2021; 11:e529. [PMID: 34459143 PMCID: PMC8351522 DOI: 10.1002/ctm2.529] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction contributes to the imbalance of cellular homeostasis and the development of diseases, which is regulated by mitochondria-associated factors. The present review aims to explore the process of the mitochondrial quality control system as a new source of the potential diagnostic biomarkers and/or therapeutic targets for diseases, including mitophagy, mitochondrial dynamics, interactions between mitochondria and other organelles (lipid droplets, endoplasmic reticulum, endosomes, and lysosomes), as well as the regulation and posttranscriptional modifications of mitochondrial DNA/RNA (mtDNA/mtRNA). The direct and indirect influencing factors were especially illustrated in understanding the interactions among regulators of mitochondrial dynamics. In addition, mtDNA/mtRNAs and proteomic profiles of mitochondria in various lung diseases were also discussed as an example. Thus, alternations of mitochondria-associated regulators can be a new category of biomarkers and targets for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Liyang Li
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Ruixue Qi
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghaiChina
| | - Linlin Zhang
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Yuexin Yu
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Jiayun Hou
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Yutong Gu
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Dongli Song
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Xiangdong Wang
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghaiChina
| |
Collapse
|
139
|
Hong Z, Ma T, Liu X, Wang C. cGAS-STING pathway: post-translational modifications and functions in sterile inflammatory diseases. FEBS J 2021; 289:6187-6208. [PMID: 34310043 DOI: 10.1111/febs.16137] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023]
Abstract
Cytoplasmic microbial and host aberrant DNAs act as danger signals and trigger host immune responses. Upon recognition, the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) catalyzes the production of a second messenger 2'3'-cGAMP, which activates endoplasmic reticulum (ER)-associated stimulator of interferon (IFN) genes (STING) and ultimately leads to the induction of type I IFNs and inflammatory genes that collectively initiate host immune defense against microbial invasion. Inappropriate activation or suppression of this signaling pathway has been implicated in the development of some autoimmune diseases, sterile inflammation, and cancers. In this review, we describe how the activity of cGAS and STING is regulated by host post-translational modifications and summarize the recent advances of cell-specific cGAS-STING activation and its association in sterile inflammatory diseases. We also discuss key outstanding questions in the field, including how our knowledge of cGAS-STING pathway could be translated into clinical applications.
Collapse
Affiliation(s)
- Ze Hong
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tianchi Ma
- Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xing Liu
- Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
140
|
Chang M, Xu G, Xiong C, Yang X, Yan S, Tao Y, Li H, Li Y, Yao S, Zhao Y. Alpha-lipoic acid attenuates silica-induced pulmonary fibrosis by improving mitochondrial function via AMPK/PGC1α pathway activation in C57BL/6J mice. Toxicol Lett 2021; 350:121-132. [PMID: 34252510 DOI: 10.1016/j.toxlet.2021.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/22/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022]
Abstract
Silicosis is characterized by pulmonary interstitial fibrosis that arises as a result of chronic exposure to silica. The few available treatments only delay its progression. As α-lipoic acid (ALA) has been shown to have various beneficial effects, including mitoprotective, antioxidant, and anti-inflammatory effects, we hypothesized that it may exhibit therapeutic effects in pulmonary fibrosis. Therefore, in the present study, we used a murine model of silicosis to investigate whether supplementation with exogenous ALA could attenuate silica-induced pulmonary fibrosis by improving mitochondrial function. ALA was administered to the model mice via continuous intragastric administration for 28 days, and then the antioxidant and mitoprotective effects of ALA were evaluated. The results showed that ALA decreased the production of reactive oxygen species, protected mitochondria from silica-induced dysfunction, and inhibited extracellular matrix deposition. ALA also decreased hyperglycemia and hyperlipidemia. Activation of the mitochondrial AMPK/PGC1α pathway might be responsible for these ALA-mediated anti-fibrotic effects. Exogenous ALA blocked oxidative stress by activating NRF2. Taken together, these findings demonstrate that exogenous ALA effectively prevents the progression of silicosis in a murine model, likely by stimulating mitochondrial biogenesis and endogenous antioxidant responses. Therefore, ALA can potentially delay the progression of silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Meiyu Chang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Guangcui Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Cheng Xiong
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Xuesi Yang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Sensen Yan
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yingjun Tao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Haibin Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yuchun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yingzheng Zhao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China.
| |
Collapse
|
141
|
Liu Q, Wu J, Zhang X, Li X, Wu X, Zhao Y, Ren J. Circulating mitochondrial DNA-triggered autophagy dysfunction via STING underlies sepsis-related acute lung injury. Cell Death Dis 2021; 12:673. [PMID: 34218252 PMCID: PMC8254453 DOI: 10.1038/s41419-021-03961-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023]
Abstract
The STING pathway and its induction of autophagy initiate a potent immune defense response upon the recognition of pathogenic DNA. However, this protective response is minimal, as STING activation worsens organ damage, and abnormal autophagy is observed during progressive sepsis. Whether and how the STING pathway affects autophagic flux during sepsis-induced acute lung injury (sALI) are currently unknown. Here, we demonstrate that the level of circulating mtDNA and degree of STING activation are increased in sALI patients. Furthermore, STING activation was found to play a pivotal role in mtDNA-mediated lung injury by evoking an inflammatory storm and disturbing autophagy. Mechanistically, STING activation interferes with lysosomal acidification in an interferon (IFN)-dependent manner without affecting autophagosome biogenesis or fusion, aggravating sepsis. Induction of autophagy or STING deficiency alleviated lung injury. These findings provide new insights into the role of STING in the regulatory mechanisms behind extrapulmonary sALI.
Collapse
Affiliation(s)
- Qinjie Liu
- Research Institute of General Surgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, China
| | - Jie Wu
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Xufei Zhang
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xuanheng Li
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Nanjing, China.
| | - Yun Zhao
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, China.
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, China.
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, China.
- Research Institute of General Surgery, Jinling Hospital, Nanjing, China.
| |
Collapse
|
142
|
Wilkinson ML, Gow AJ. Effects of fatty acid nitroalkanes on signal transduction pathways and airway macrophage activation. Innate Immun 2021; 27:353-364. [PMID: 34375151 PMCID: PMC8419298 DOI: 10.1177/17534259211015330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Fatty acid nitroalkenes are reversibly-reactive electrophiles that are endogenously detectable at nM concentrations and display anti-inflammatory, pro-survival actions. These actions are elicited through the alteration of signal transduction proteins via a Michael addition on nucleophilic cysteine thiols. Nitrated fatty acids (NO2-FAs), like 9- or 10-nitro-octadec-9-enolic acid, will act on signal transduction proteins directly or on key regulatory proteins to cause an up-regulation or down-regulation of the protein's expression, yielding an anti-inflammatory response. These responses have been characterized in many organ systems, such as the cardiovascular system, with the pulmonary system less well defined. Macrophages are one of the most abundant immune cells in the lung and are essential in maintaining lung homeostasis. Despite this, macrophages can play a role in both acute and chronic lung injury due to up-regulation of anti-inflammatory signal transduction pathways and down-regulation of pro-inflammatory pathways. Through their propensity to alter signal transduction pathways, NO2-FAs may be able to reduce macrophage activation during pulmonary injury. This review will focus on the implications of NO2-FAs on macrophage activation in the lung and the signal transduction pathways that may be altered, leading to reduced pulmonary injury.
Collapse
Affiliation(s)
- Melissa L Wilkinson
- Department of Pharmacology and Toxicology, The State University of New Jersey, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, The State University of New Jersey, USA
| |
Collapse
|
143
|
Liu J, Jia Z, Gong W. Circulating Mitochondrial DNA Stimulates Innate Immune Signaling Pathways to Mediate Acute Kidney Injury. Front Immunol 2021; 12:680648. [PMID: 34248963 PMCID: PMC8264283 DOI: 10.3389/fimmu.2021.680648] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/07/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial dysfunction is increasingly considered as a critical contributor to the occurrence and progression of acute kidney injury (AKI). However, the mechanisms by which damaged mitochondria mediate AKI progression are multifactorial and complicated. Mitochondrial DNA (mtDNA) released from damaged mitochondria could serve as a danger-associated molecular pattern (DAMP) and activate the innate immune system through STING, TLR9, NLRP3, and some other adaptors, and further mediate tubular cell inflammation and apoptosis. Accumulating evidence has demonstrated the important role of circulating mtDNA and its related pathways in the progression of AKI, and regulating the proteins involved in these pathways may be an effective strategy to reduce renal tubular injury and alleviate AKI. Here, we aim to provide a comprehensive overview of recent studies on mtDNA-mediated renal pathological events to provide new insights in the setting of AKI.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| |
Collapse
|
144
|
Abstract
cGAS (cytosolic DNA sensor cyclic AMP-GMP synthase)-STING (stimulator of interferon genes) signaling is critical for sensing cytosolic DNA to initiate host immune responses against invading pathogens and cancer. However, inappropriate activation of STING signaling causes severe and often fatal autoimmune or autoinflammatory diseases. Hence, STING is an attractive drug target for the treatment of STING-driven autoimmune and inflammatory disorders. Therefore, there is a need to identify lead compounds that effectively inhibit human STING for further drug development. Here, we identified and characterized a STING-specific inhibitor SN-011 with high efficiency, specificity, and safety, paving the way for therapeutically manipulating STING-mediated clinical diseases. Cytosolic DNA activates cGAS (cytosolic DNA sensor cyclic AMP-GMP synthase)-STING (stimulator of interferon genes) signaling, which triggers interferon and inflammatory responses that help defend against microbial infection and cancer. However, aberrant cytosolic self-DNA in Aicardi–Goutière’s syndrome and constituently active gain-of-function mutations in STING in STING-associated vasculopathy with onset in infancy (SAVI) patients lead to excessive type I interferons and proinflammatory cytokines, which cause difficult-to-treat and sometimes fatal autoimmune disease. Here, in silico docking identified a potent STING antagonist SN-011 that binds with higher affinity to the cyclic dinucleotide (CDN)-binding pocket of STING than endogenous 2′3′-cGAMP. SN-011 locks STING in an open inactive conformation, which inhibits interferon and inflammatory cytokine induction activated by 2′3′-cGAMP, herpes simplex virus type 1 infection, Trex1 deficiency, overexpression of cGAS-STING, or SAVI STING mutants. In Trex1−/− mice, SN-011 was well tolerated, strongly inhibited hallmarks of inflammation and autoimmunity disease, and prevented death. Thus, a specific STING inhibitor that binds to the STING CDN-binding pocket is a promising lead compound for STING-driven disease.
Collapse
|
145
|
He WR, Cao LB, Yang YL, Hua D, Hu MM, Shu HB. VRK2 is involved in the innate antiviral response by promoting mitostress-induced mtDNA release. Cell Mol Immunol 2021; 18:1186-1196. [PMID: 33785841 PMCID: PMC8093274 DOI: 10.1038/s41423-021-00673-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/07/2021] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial stress (mitostress) triggered by viral infection or mitochondrial dysfunction causes the release of mitochondrial DNA (mtDNA) into the cytosol and activates the cGAS-mediated innate immune response. The regulation of mtDNA release upon mitostress remains uncharacterized. Here, we identified mitochondria-associated vaccinia virus-related kinase 2 (VRK2) as a key regulator of this process. VRK2 deficiency inhibited the induction of antiviral genes and caused earlier and higher mortality in mice after viral infection. Upon viral infection, VRK2 associated with voltage-dependent anion channel 1 (VDAC1) and promoted VDAC1 oligomerization and mtDNA release, leading to the cGAS-mediated innate immune response. VRK2 was also required for mtDNA release and cGAS-mediated innate immunity triggered by nonviral factors that cause Ca2+ overload but was not required for the cytosolic nucleic acid-triggered innate immune response. Thus, VRK2 plays a crucial role in the mtDNA-triggered innate immune response and may be a potential therapeutic target for infectious and autoimmune diseases associated with mtDNA release.
Collapse
Affiliation(s)
- Wen-Rui He
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China
| | - Li-Bo Cao
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yu-Lin Yang
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China
| | - Duo Hua
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ming-Ming Hu
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Hong-Bing Shu
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
146
|
李 永, 崔 淑, 孟 卫, 胡 海, 王 琛. [Mitochondrial DNA and cGAS-STING Innate Immune Signaling Pathway: Latest Research Progress]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2021; 52:387-395. [PMID: 34018355 PMCID: PMC10409199 DOI: 10.12182/20210560501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 11/23/2022]
Abstract
Mitochondria are important organelles that present extensively in cells, serving diverse functions. In addition to controlling cell energy production and metabolism, mitochondria are also involved in various biological processes, including anti-infection, apoptosis, and autophagy. Harmful stimuli from external environment or those generated by the cells themselves can damage mitochondria and cause mitochondrial stress response, during which the mitochondrial matrix containing mitochondrial DNA (mtDNA) can leak into the cytoplasm. Cytoplasmic mtDNA, acting as a damage-associated molecular pattern (DAMP), can activate a panel of DNA sensors and elicit innate immune response in organisms. Cyclic GMP-AMP synthase (cGAS), a key intracellular DNA sensor, can catalyze the conversion of GTP and ATP to cyclic GMP-AMP (2'3'-cGAMP), which serves as second messenger to bind and activate stimulator of interferon gene (STING), an endoplasmic adaptor protein. Beyond its critical roles in anti-microbial immunity, cGAS-STING pathway also serves important functions in many pathological and physiological processes such as autoimmunity, tumor and senescence. In this review, we focus on how the mtDNA released during mitochonrial stress response activates the cGAS-STING innate immune signaling pathway and the associated diseases, in order to help promote basic research about the role of mitochondria in innate immunity and provide new strategies for developing mitochondria-targeting drugs.
Collapse
Affiliation(s)
- 永兴 李
- 中国药科大学 天然药物活性组分与药效国家重点实验室 (中国药科大学生命科学与技术学院) (南京 210009)State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - 淑方 崔
- 中国药科大学 天然药物活性组分与药效国家重点实验室 (中国药科大学生命科学与技术学院) (南京 210009)State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - 卫 孟
- 中国药科大学 天然药物活性组分与药效国家重点实验室 (中国药科大学生命科学与技术学院) (南京 210009)State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - 海洋 胡
- 中国药科大学 天然药物活性组分与药效国家重点实验室 (中国药科大学生命科学与技术学院) (南京 210009)State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - 琛 王
- 中国药科大学 天然药物活性组分与药效国家重点实验室 (中国药科大学生命科学与技术学院) (南京 210009)State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
147
|
Lescoat A, Cavalin C, Lecureur V, Jégo P. [Toward a better understanding of the etiology of systemic autoimmune diseases : should a systemic disease still be defined as a "diffuse inflammatory disease of unknown origin" in 2021? Example of crystalline silica exposure]. Rev Med Interne 2021; 42:233-236. [PMID: 33781611 DOI: 10.1016/j.revmed.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/14/2021] [Indexed: 10/21/2022]
Affiliation(s)
- A Lescoat
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France; Department of Internal Medicine and Clinical Immunology, CHU Rennes, University of Rennes 1, Rennes, France.
| | - C Cavalin
- Institut de Recherche Interdisciplinaire en Sciences Sociales (IRISSO), UMR CNRS-INRA 7170-1427, Université Paris-Dauphine, Paris, France; Centre d'études de l'emploi et du travail (CEET, CNAM), Noisy-le-Grand, France; Laboratoire interdisciplinaire d'évaluation des politiques publiques (LIEPP) de Sciences Po, Paris, France
| | - V Lecureur
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - P Jégo
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France; Department of Internal Medicine and Clinical Immunology, CHU Rennes, University of Rennes 1, Rennes, France
| |
Collapse
|
148
|
Li C, Zhang L, Qian D, Cheng M, Hu H, Hong Z, Cui Y, Yu H, Wang Q, Zhu J, Meng W, Xu JF, Sun Y, Zhang P, Wang C. RNF111-facilitated neddylation potentiates cGAS-mediated antiviral innate immune response. PLoS Pathog 2021; 17:e1009401. [PMID: 33720974 PMCID: PMC7959372 DOI: 10.1371/journal.ppat.1009401] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthetase (cGAS) has emerged as a fundamental component fueling the anti-pathogen immunity. Because of its pivotal role in initiating innate immune response, the activity of cGAS must be tightly fine-tuned to maintain immune homeostasis in antiviral response. Here, we reported that neddylation modification was indispensable for appropriate cGAS-STING signaling activation. Blocking neddylation pathway using neddylation inhibitor MLN4924 substantially impaired the induction of type I interferon and proinflammatory cytokines, which was selectively dependent on Nedd8 E2 enzyme Ube2m. We further found that deficiency of the Nedd8 E3 ligase Rnf111 greatly attenuated DNA-triggered cGAS activation while not affecting cGAMP induced activation of STING, demonstrating that Rnf111 was the Nedd8 E3 ligase of cGAS. By performing mass spectrometry, we identified Lys231 and Lys421 as essential neddylation sites in human cGAS. Mechanistically, Rnf111 interacted with and polyneddylated cGAS, which in turn promoted its dimerization and enhanced the DNA-binding ability, leading to proper cGAS-STING pathway activation. In the same line, the Ube2m or Rnf111 deficiency mice exhibited severe defects in innate immune response and were susceptible to HSV-1 infection. Collectively, our study uncovered a vital role of the Ube2m-Rnf111 neddylation axis in promoting the activity of the cGAS-STING pathway and highlighted the importance of neddylation modification in antiviral defense.
Collapse
Affiliation(s)
- Chenhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lele Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Qian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mingxing Cheng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ze Hong
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ye Cui
- Division of Immunology, The Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Huansha Yu
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jin-fu Xu
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Sun
- Cancer Institute of the 2 affiliated hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- * E-mail: (YS); (PZ); (CW)
| | - Peng Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- * E-mail: (YS); (PZ); (CW)
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- * E-mail: (YS); (PZ); (CW)
| |
Collapse
|
149
|
Pu F, Chen F, Liu J, Zhang Z, Shao Z. Immune Regulation of the cGAS-STING Signaling Pathway in the Tumor Microenvironment and Its Clinical Application. Onco Targets Ther 2021; 14:1501-1516. [PMID: 33688199 PMCID: PMC7935450 DOI: 10.2147/ott.s298958] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/19/2021] [Indexed: 12/25/2022] Open
Abstract
As a DNA receptor in the cytoplasm, cyclic GMP-AMP synthase (cGAS) contributes to the recognition of abnormal DNA in the cytoplasm and contributes to the stimulator of interferon genes (STING) signaling pathway. cGAS could mediate the expression of interferon-related genes, inflammatory-related factors, and downstream chemokines, thus initiating the immune response. The STING protein is a key effector downstream of the DNA receptor pathway. It is widely expressed across cell types such as immune cells, tumor cells, and stromal cells and plays a role in signal transduction for cytoplasmic DNA sensing and immunity. STING agonists, as novel agonists, are used in preclinical research and in the treatment of various tumors via clinical trials and have displayed attractive application prospects. Studying the cGAS-STING signaling pathway will deepen our understanding of tumor immunity and provide a basis for the research and development of antitumor drugs.
Collapse
Affiliation(s)
- Feifei Pu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fengxia Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, People's Republic of China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
150
|
Chauhan PS, Wagner JG, Benninghoff AD, Lewandowski RP, Favor OK, Wierenga KA, Gilley KN, Ross EA, Harkema JR, Pestka JJ. Rapid Induction of Pulmonary Inflammation, Autoimmune Gene Expression, and Ectopic Lymphoid Neogenesis Following Acute Silica Exposure in Lupus-Prone Mice. Front Immunol 2021; 12:635138. [PMID: 33732257 PMCID: PMC7959771 DOI: 10.3389/fimmu.2021.635138] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Occupational exposure to crystalline silica (cSiO2) is etiologically associated with systemic lupus erythematosus (lupus) and other autoimmune diseases. cSiO2's autoimmune effects in humans can be mimicked chronically in female lupus-prone NZBWF1 mice following repeated exposure to the particle. However, the immediate and short-term effects of cSiO2 in this widely used model of autoimmune disease are not well-understood. In the present study, we tested the hypothesis that a single acute cSiO2 dose triggers early presentation of cellular, histopathological, transcriptomic, and protein biomarkers of inflammation and autoimmunity in lupus-prone mice. Eight-week old female NZBWF1 mice were intranasally instilled once with 2.5 mg cSiO2 or saline vehicle and necropsied at 1, 7, 14, 21, and 28 d post-instillation (PI). Analyses of bronchoalveolar lavage fluid (BALF) and lung tissue revealed that by 7 d PI, acute cSiO2 exposure persistently provoked: (i) robust recruitment of macrophages, neutrophils, and lymphocytes into the alveoli, (ii) cell death as reflected by increased protein, double-stranded DNA, and lactate dehydrogenase activity, (iii) elevated secretion of the cytokines IL-1α, IL-1β, IL-18, TNF-α, IL-6, MCP-1, and B cell activation factor (BAFF), and (iv) upregulation of genes associated with chemokines, proinflammatory cytokines, lymphocyte activation, and type I interferon signaling. The appearance of these endpoints was subsequently followed by the emergence in the lung of organized CD3+ T cells (14 d PI) and CD45R+ B cells (21 d PI) that were indicative of ectopic lymphoid structure (ELS) development. Taken together, acute cSiO2 exposure triggered a rapid onset of autoimmune disease pathogenesis that was heralded in the lung by unresolved inflammation and cell death, proinflammatory cytokine production, chemokine-driven recruitment of leukocytes, an interferon response signature, B and T cell activation, and ELS neogenesis. This short-term murine model provides valuable new insight into potential early mechanisms of cSiO2-induced lupus flaring and, furthermore, offers a rapid venue for evaluating interventions against respirable particle-triggered inflammation and autoimmunity.
Collapse
Affiliation(s)
- 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
| | - James G. Wagner
- 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
| | - Abby D. Benninghoff
- Department of Animal, Dairy and Veterinary Sciences, School of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Ryan P. Lewandowski
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Olivia K. Favor
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pharmacology and 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
| | - Kristen N. Gilley
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Elizabeth A. Ross
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 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
- Department of Pharmacology and Toxicology, 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, East Lansing, MI, United States
| |
Collapse
|