1
|
Jing J, Sun Y, Shui Y, Wang J, Ye W, Chen R, Wu L, Xing L, Huang R, Zhou T, Zhu W, Wu Y, Zhang S, Shi J, Li Y, Liu Y, You Z. Morus alba L. alleviates influenza viral pneumonia. Evidences of its mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156574. [PMID: 40054181 DOI: 10.1016/j.phymed.2025.156574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Viral pneumonia is an infection of the lungs caused by numerous different viruses, which can lead to severe respiratory distress and even life-threatening conditions. In the absence of specific treatments for viral pneumonia, natural traditional medicines offer an alternative in terms of innovative drug therapies. Morus alba L. (common name mulberry leaf) is a Chinese medicine that has been used clinically as an antiviral. PURPOSE The therapeutic effect of M. alba on viral pneumonia was investigated along with its mechanism of action. METHODS Network pharmacology and molecular docking were used to analyze the mechanism of action of M. alba in the treatment of viral pneumonia. Histology, immunofluorescence, Western blotting, qPCR, and flow cytometry were used to evaluate the protective effect of MLE (the ethanol extract of Morus alba L.) on PR8 (A/Puerto Rico/8/1934 H1N1, a murine lung-adapted influenza A virus strain)-induced viral pneumonia. SiRNA was used to validate the relationship between the therapeutic effects of MLE on viral pneumonia and the target Syk (a crucial non-receptor tyrosine kinase). RESULTS MLE alleviated PR8-induced viral pneumonia by reducing inflammatory factor expression in the lungs, decreasing NF-κB pathway activation, slowing oxidative damage in the lungs, and inhibiting lung tissue cell apoptosis. Meanwhile, MLE for viral pneumonia was significantly associated with Syk targets. Notably, knockdown of the Syk gene not only reduced the therapeutic effect of MLE, but also suppressed PR8-induced viral pneumonia. CONCLUSION MLE can alleviate PR8-induced viral pneumonia through inhibiting the Dectin-1/Syk pathway.
Collapse
Affiliation(s)
- Junsong Jing
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Yi Sun
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Yiyang Shui
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Junyi Wang
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Wenjing Ye
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Ranran Chen
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Lianhao Wu
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Lijuan Xing
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Rongrong Huang
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; School of Public Health, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Ting Zhou
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Wenwen Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yueguo Wu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Sheng Zhang
- Center for Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
| | - Jing Shi
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.
| | - Yuanyuan Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.
| | - Yan Liu
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou, China.
| | - Zhenqiang You
- School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China.
| |
Collapse
|
2
|
Chen L, Zhu F, Gao X, Yang Y, Jin G, Zhou Y, Dong G, Zhou G. Spleen tyrosine kinase aggravates intestinal inflammation through regulating inflammatory responses of macrophage in ulcerative colitis. Int Immunopharmacol 2025; 148:114122. [PMID: 39862635 DOI: 10.1016/j.intimp.2025.114122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/01/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a persistent chronic, non-specific inflammatory disease, and macrophages play a crucial role in its pathogenesis. Spleen tyrosine kinase (Syk) is strongly associated with the pathogenesis of several inflammatory diseases. However, the role of Syk in the pathogenesis of UC is still obscure. METHODS Syk expression was analyzed in peripheral blood mononuclear cells (PBMCs) and colonic tissues of UC patients using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunofluorescence. A public database was used to analyze the expression of selected signature genes of interest in UC patients with different expressions of Syk. R788, a small molecule inhibitor of Syk, was used to treat macrophages from mice. The functions of macrophages were assessed using qRT-PCR, flow cytometry, and fluorescence microscopy. Dextran sodium sulfate (DSS)-induced colitis mice model was established to determine the role of Syk in UC. RESULTS The Syk levels were markedly increased in PBMCs, colonic tissues, and colonic mucosa lamina propria macrophages from UC patients, and positively related to disease activity. Inhibition of Syk with R788 decreased pro-inflammatory genes expression and increased anti-inflammatory genes expression in peritoneal macrophages and bone marrow macrophages. Blockade of Syk enhanced phagocytosis and bactericidal ability of macrophages. Syk promoted the production of reactive oxygen species of macrophages and M1-type macrophage polarization. Furthermore, inhibition of Syk alleviated intestinal mucosal inflammation in DSS-induced colitis model. CONCLUSIONS Syk plays a vital role in intestinal inflammation by regulating inflammatory responses of macrophages in UC. Targeting Syk may be a promising therapeutic approach for UC.
Collapse
Affiliation(s)
- Leilei Chen
- Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Xizhuang Gao
- Department of Clinical Medicine, Jining Medical University, Jining 272000, Shandong, China
| | - Yonghong Yang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, Shandong, China
| | - Guiyuan Jin
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, Shandong, China
| | - Yaqi Zhou
- Department of Clinical Medicine, Jining Medical University, Jining 272000, Shandong, China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Guangxi Zhou
- Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China.
| |
Collapse
|
3
|
Qin H, Wang J, Bai L, Ding H, Ding H, Zhang F, Han Y. Aerosol inhalation of rhIL-10 improves acute lung injury in mice by affecting pulmonary neutrophil phenotypes through neutrophil-platelet aggregates. Int Immunopharmacol 2025; 147:113948. [PMID: 39778276 DOI: 10.1016/j.intimp.2024.113948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025]
Abstract
This study investigates the therapeutic effects of recombinant human IL-10 (rhIL-10) administered via aerosol inhalation in acute lung injury (ALI), with a particular focus on neutrophils. It explores how rhIL-10, in the presence of platelets, modulates neutrophil polarization to ameliorate acute lung injury. Initially, the ALI model established in mice demonstrated that aerosol inhalation of rhIL-10 significantly mitigated the cytokine storm in the lungs, reduced pulmonary edema, and alleviated histopathological damage to lung tissue. Additionally, rhIL-10 administration was found to decrease neutrophil infiltration and platelet activation in the lungs of mice, inhibiting the formation of platelet-neutrophil aggregates (PNAs) and promoting the differentiation of neutrophils toward an anti-inflammatory phenotype in the presence of platelets. Subsequently, primary neutrophils and platelets were isolated from mouse bone marrow and blood to explore the underlying mechanisms. The results indicated that rhIL-10 promotes the expression of the signal transducer and activator of transcription 3 (STAT3) and the suppressor of cytokine signaling 3 (SOCS3) in neutrophils while inhibiting the activation of the nuclear factor kappa B (NF-κB) and the NF-κB inhibitor (IκB), which in turn enhances CD40 expression. This interaction facilitates the formation of PNAs and influences neutrophil phenotype differentiation. Furthermore, the application of the STAT3 phosphorylation inhibitor Stattic and CD40 antibody in vivo provided further validation of this potential mechanism. In conclusion, these results indicate that aerosol inhalation of rhIL-10 effectively ameliorates ALI. The underlying mechanism may involve the modulation of the neutrophil STAT/SOCS-IκB/NF-κB-CD40 signaling pathway, promoting interactions between neutrophils and platelets that facilitate the differentiation of neutrophils toward an anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Huan Qin
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiangang Wang
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | - Luyuan Bai
- Xianyang Hospital of Yan'an University, Xianyang, China
| | - Huiqin Ding
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | | | | | - Yantao Han
- School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
4
|
Warner S, Teague HL, Ramos-Benitez MJ, Panicker S, Allen K, Gairhe S, Moyer T, Parachalil Gopalan B, Douagi I, Shet A, Kanthi Y, Suffredini AF, Chertow DS, Strich JR. R406 reduces lipopolysaccharide-induced neutrophil activation. Cell Immunol 2024; 403-404:104860. [PMID: 39084187 PMCID: PMC11387147 DOI: 10.1016/j.cellimm.2024.104860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Modulating SYK has been demonstrated to have impacts on pathogenic neutrophil responses in COVID-19. During sepsis, neutrophils are vital in early bacterial clearance but also contribute to the dysregulated immune response and organ injury when hyperactivated. Here, we evaluated the impact of R406, the active metabolite of fostamatinib, on neutrophils stimulated by LPS. We demonstrate that R406 was able to effectively inhibit NETosis, degranulation, ROS generation, neutrophil adhesion, and the formation of CD16low neutrophils that have been linked to detrimental outcomes in severe sepsis. Further, the neutrophils remain metabolically active, capable of releasing cytokines, perform phagocytosis, and migrate in response to IL-8. Taken together, this data provides evidence of the potential efficacy of utilizing fostamatinib in bacterial sepsis.
Collapse
Affiliation(s)
- Seth Warner
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Marcos J Ramos-Benitez
- Basic Science Department, Microbiology Division, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Sumith Panicker
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiana Allen
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Salina Gairhe
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Moyer
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bindu Parachalil Gopalan
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Iyadh Douagi
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; NIH Center for Human Immunology, Inflammation, and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arun Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony F Suffredini
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Chertow
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Shi S, Deng R, Huang R, Zhou S. Bergapten attenuates sepsis-induced acute lung injury in mice by regulating Th17/Treg balance. Inhal Toxicol 2024; 36:421-430. [PMID: 39420573 DOI: 10.1080/08958378.2024.2400479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The abnormality of the immune system caused by infection is a contributor to the organ dysfunctions associated with sepsis. The balance between Th17/Treg cells is essential for maintaining immune homeostasis. Bergapten is a natural furocoumarin and has been reported to alleviate the Th17/Treg imbalance. Here, we explored the effects of bergapten on the inflammation and immune state in mouse models of sepsis. METHODS The model was established using the cecal ligation and puncture method. Mice were administered 30 mg/kg bergapten. Histological examination, RT-qPCR, enzyme-linked immunosorbent assay, immunoblotting, immunofluorescence, immunohistochemistry, and flow cytometry were used to evaluate the effects of bergapten in vivo. RESULTS Bergapten ameliorated lung damage, reduced lung wet/dry weight ratio, inhibited myeloperoxidase activity, and reduced inflammatory cell infiltration. Bergapten also restrained sepsis-induced inflammation via inhibition of inflammatory cytokines and NF-κB signaling. These effects were accompanied by the restored Th17/Treg balance induced by bergapten. Bergapten decreased the number of Th17 cells and elevated the number of Tregs, and this effect was mediated by the signal transducer and activator of transcription 5 (STAT5)/Forkhead box P3 (Foxp3) and STAT3/retinoid-related orphan receptor-γt (RORγt) pathways. CONCLUSIONS Bergapten exerted anti-inflammatory effects in acute lung injury by improving the Th17/Treg balance, which suggested a potential of bergapten as an immunomodulatory drug treating sepsis-associated diseases.
Collapse
Affiliation(s)
- Shanqiu Shi
- Department of Emergency Medicine, Hanzhong Central Hospital, Hanzhong, China
| | - Rui Deng
- Multimodal Therapy Department of Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Renchun Huang
- Department of Emergency Medicine, Hanzhong Central Hospital, Hanzhong, China
| | - Shitai Zhou
- Department of Emergency Medicine, Hanzhong Central Hospital, Hanzhong, China
| |
Collapse
|
6
|
Li K, Deng Z, Lei C, Ding X, Li J, Wang C. The Role of Oxidative Stress in Tumorigenesis and Progression. Cells 2024; 13:441. [PMID: 38474405 PMCID: PMC10931308 DOI: 10.3390/cells13050441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress refers to the imbalance between the production of reactive oxygen species (ROS) and the endogenous antioxidant defense system. Its involvement in cell senescence, apoptosis, and series diseases has been demonstrated. Advances in carcinogenic research have revealed oxidative stress as a pivotal pathophysiological pathway in tumorigenesis and to be involved in lung cancer, glioma, hepatocellular carcinoma, leukemia, and so on. This review combs the effects of oxidative stress on tumorigenesis on each phase and cell fate determination, and three features are discussed. Oxidative stress takes part in the processes ranging from tumorigenesis to tumor death via series pathways and processes like mitochondrial stress, endoplasmic reticulum stress, and ferroptosis. It can affect cell fate by engaging in the complex relationships between senescence, death, and cancer. The influence of oxidative stress on tumorigenesis and progression is a multi-stage interlaced process that includes two aspects of promotion and inhibition, with mitochondria as the core of regulation. A deeper and more comprehensive understanding of the effects of oxidative stress on tumorigenesis is conducive to exploring more tumor therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Changshan Wang
- Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, 49 Xilingol South Road, Yu Quan District, Hohhot 010020, China; (K.L.); (Z.D.); (C.L.); (X.D.); (J.L.)
| |
Collapse
|
7
|
Wang S, Ma J, Li X, Xian X, Tan G, Cai H, Yang B, Zhang A, Guo J, Gu G, Meng Z, Fu B. EGR-1 Contributes to Pulmonary Edema by Regulating the Epithelial Sodium Channel in Lipopolysaccharide-Induced Acute Lung Injury. Immunol Invest 2023; 52:925-939. [PMID: 37732637 DOI: 10.1080/08820139.2023.2256778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Acute lung injury (ALI) is a common lung disease with increasing morbidity and mortality rates due to the lack of specific drugs. Impaired alveolar fluid clearance (AFC) is a primary pathological feature of ALI. Epithelial sodium channel (ENaC) is a primary determinant in regulating the transport of Na+ and the clearance of alveolar edema fluid. Therefore, ENaC is an important target for the development of drugs for ALI therapy. However, the role of ENaC in the progression of ALI remains unclear. Inhibition of early growth response factor (EGR-1) expression has been reported to induce a protective effect on ALI; therefore, we evaluated whether EGR-1 participates in the progression of ALI by regulating ENaC-α in alveolar epithelium. We investigated the potential mechanism of EGR-1-mediated regulation of ENaC in ALI. We investigated whether EGR-1 aggravates the pulmonary edema response in ALI by regulating ENaC. ALI mouse models were established by intrabronchial injection of lipopolysaccharides (LPS). Lentiviruses with EGR-1 knockdown were transfected into LPS-stimulated A549 cells. We found that EGR-1 expression was upregulated in the lung tissues of ALI mice and in LPS-induced A549 cells, and was negatively correlated with ENaC-α expression. Knockdown of EGR-1 increased ENaC-α expression and relieved cellular edema in ALI. Moreover, EGR-1 regulated ENaC-α expression at the transcriptional level, and correspondingly promoted pulmonary edema and aggravated ALI symptoms. In conclusion, our study demonstrated that EGR-1 could promote pulmonary edema by downregulating ENaC-α at the transcriptional level in ALI. Our study provides a new potential therapeutic strategy for treatment of ALI.
Collapse
Affiliation(s)
- Song Wang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jing Ma
- Office of Academic Research, Liaocheng People's Hospital, Liaocheng, China
| | - Xin Li
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Xinmiao Xian
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guikun Tan
- Pharmacy Department, Liaocheng Woman and Child Health Care Hospital, Liaocheng, China
| | - Hongwei Cai
- Department of Critical Care Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Bingwu Yang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Anqi Zhang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jianran Guo
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guohao Gu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Zhen Meng
- Biomedical Laboratory, Medical School of Liaocheng University, Liaocheng, China
| | - Bo Fu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
8
|
Zhou Y, Zhang Y, Yu W, Qin Y, He H, Dai F, Wang Y, Zhu F, Zhou G. Immunomodulatory role of spleen tyrosine kinase in chronic inflammatory and autoimmune diseases. Immun Inflamm Dis 2023; 11:e934. [PMID: 37506139 PMCID: PMC10373573 DOI: 10.1002/iid3.934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The high prevalence of chronic inflammatory diseases or autoimmune reactions is a major source of concern and affects the quality of life of patients. Chronic inflammatory or autoimmune diseases are associated with many diseases in humans, including asthma, rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease and cancer. Splenic tyrosine kinase (SYK) is a non-receptor tyrosine kinase that plays an important role in immune receptor signalling in immune and inflammatory responses. METHODS This is a review article in which we searched for keywords "splenic tyrosine kinase", "inflammation" and "autoimmune diseases" in published literature such as Pubmed and Web of Science to collect relevant information and then conducted a study focusing on the latest findings on the involvement of SYK in chronic inflammatory or autoimmune diseases. RESULTS This paper reviews the regulation of Fcγ, NF-κB, B cell and T cell-related signalling pathways by SYK, which contributes to disease progression in chronic inflammatory and autoimmune diseases such as airway fibrosis, inflammatory skin disease and inflammatory bowel disease. CONCLUSION This paper shows that SYK plays an important role in chronic inflammatory and autoimmune diseases. syk targets hematological, autoimmune and other inflammatory diseases and therefore, inhibition of SYK expression or blocking its related pathways may provide new ideas for clinical prevention and treatment of inflammatory or autoimmune diseases.
Collapse
Affiliation(s)
- Yaqi Zhou
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yaowen Zhang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Wei Yu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yufen Qin
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Heng He
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fengxian Dai
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yibo Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
9
|
Jiang X, Sun J, Guo S, Zhao Z, Chen Y, Cao J, Liu Y, Cheng G, Tian L, Li Y. Elsholtzia bodinieri Vaniot ameliorated acute lung injury in mice by regulating pyroptosis, inflammation, oxidative stress and macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116232. [PMID: 36764561 DOI: 10.1016/j.jep.2023.116232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Elsholtzia bodinieri Vaniot, perennial herbs, a traditional Yunnan Chinese herbal medicine. Its whole herb can be used as commonly used herbs to cure fever, headache, inflammation, indigestion etc., and its tender tip can also be used as tea in Yunnan of China. However, the protective mechanism of Elsholtzia bodinieri Vaniot on acute lung injury (ALI) still needs to be explored. AIM OF STUDY ALI is characterized by acute respiratory inflammation, which remains a significant source of morbidity and mortality. The current study with the aim of determining the therapeutic the efficacy of E. bodinieri Vaniot on lipopolysaccharide-induced ALI, moreover uncovered the underlying gene-regulated framework, so E. bodinieri Vaniot might serve as functional food for adjuvant therapy or therapeutic agent. MATERIALS AND METHODS These potential pharmacological targets of E. bodinieri Vaniot against ALI were analyzed by multiple bioinformatics databases. E. bodinieri Vaniot methanol extract (EBE) was obtained by ultrasonic-assisted extraction method, and detected by UHPLC-ESI-HRMS/MS. These pyroptosis, inflammation and oxidative stress associated factors were measured using ELISA assay, western blotting, and histopathological examination to assess the effects of EBE. EcoTyper and immunofluorescence staining were employed to estimate macrophage polarization states in ALI lungs tissue. RESULTS In ALI lung tissues, EBE treatment could increase B cell leukemia/lymphoma 2 (BCL2) to inhibit pyroptosis, downregulate prostaglandin-endoperoxide synthase 2 (PTGS2) to attenuate inflammation, upregulating NAD(P)H dehydrogenase, quinone 1 (NQO1) to alleviate oxidative stress and induce macrophage polarization toward the M2 phenotype. CONCLUSION E. bodinieri Vaniot ameliorated ALI thought regulating pyroptosis, inflammation, oxidative stress and macrophage polarization, as well as could be a promising source for therapeutic agent.
Collapse
Affiliation(s)
- Xiaoqian Jiang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jin Sun
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Shancheng Guo
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhiye Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yuxu Chen
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jianxin Cao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yaping Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Guiguang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Lei Tian
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Ye Li
- School of Medicine, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
10
|
Hong X, Zhou Y, Zhu Z, Li Y, Li Z, Zhang Y, Hu X, Zhu F, Wang Y, Fang M, Huang Y, Shen T. Environmental endocrine disruptor Bisphenol A induces metabolic derailment and obesity via upregulating IL-17A in adipocytes. ENVIRONMENT INTERNATIONAL 2023; 172:107759. [PMID: 36696794 DOI: 10.1016/j.envint.2023.107759] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Bisphenol A (BPA), a ubiquitous environmental endocrine disruptor, has been extensively demonstrated to be associated with metabolic disorders, including obesity and type 2 diabetes mellitus. However, the underlying mechanism underpinning the environmental etiology of chronic metabolic disorders has not been sufficiently elucidated. OBJECTIVES This study is designed to explore the toxicological pathogenesis of chronic inflammation in BPA exposure during obesity. METHODS We investigated the role of IL-17A in the association of BPA exposure and obesity from human cross-sectional study to animal models, including genetically modified IL-17A-/- mice. RESULTS Here, our work started from case-control observation that BPA exposure was significantly associated with risk of obesity (odds ratio = 4.72, 95%CI: 3.18 - 11.18, P < 0.01), metabolic disorder and levels of interleukin-17A (IL-17A) in human adipose (estimated changes β = 0.46, 95%CI: 0.15 - 1.01, P < 0.01) with bariatric surgery. Animal model fed with high-fat diet (HFD) confirmed that BPA exposure aggravated body weight gain and insulin resistance, concurrent with much heightened inflammatory responses in the adipose tissue including increase in IL-17A and macrophage polarization towards M1 stage. Genetically modified IL-17A ablated mice (IL-17A-/-) showed reversed adipose tissue inflammation response, improved macrophage polarization homeostasis, along with insulin sensitivity in both HFD group alone or much more significantly the HFD + BPA group. Moreover, mediation analysis in human epidemiological investigation demonstrated that plasma IL-17A attributed up to 30.01% mediating role in the associations between BPA exposure and obesity risk. DISCUSSION This research paradigm from human to animal provides strong evidence for the elucidation of IL-17A moderating inflammation and insulin resistance in obesity. Such findings reiterate the obesogenic role of environmental endocrine disruptor BPA in metabolic disorders and unveils the potential toxicological mechanisms underpinning such effect.
Collapse
Affiliation(s)
- Xu Hong
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yi Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Zhiyuan Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yuting Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Zuo Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yuheng Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Xinxin Hu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Fuhai Zhu
- Health Management Center, Second Affiliated Hospital, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yong Wang
- Department of General Surgery, Second Affiliated Hospital, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China.
| | - Tong Shen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China.
| |
Collapse
|
11
|
Chen L, Liu X, Wang X, Lu Z, Ye Y. Berberine Alleviates Acute Lung Injury in Septic Mice by Modulating Treg/Th17 Homeostasis and Downregulating NF-κB Signaling. Drug Des Devel Ther 2023; 17:1139-1151. [PMID: 37077411 PMCID: PMC10108910 DOI: 10.2147/dddt.s401293] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
Purpose A common complication of sepsis is acute lung injury (ALI), which is associated with an acute onset, rapid disease changes, and high mortality. Regulatory T (Treg) and T helper 17 (Th17) cells comprise CD4+ T cell subsets, which strongly influence inflammation during ALI. In this study, we investigated the effect of berberine (BBR), an antioxidant, anti-inflammatory, and immunomodulatory drug, on the inflammatory response and immune state in mice with sepsis. Methods A mouse model of cecal ligation and puncture (CLP) was established. The mice were intragastrically administered 50 mg/kg BBR. We used histological techniques to evaluate inflammatory tissue injury and flow cytometry for analyzing Treg/Th17 levels. We also assessed NF-κB signaling pathways by Western blotting assays and immunofluorescence staining. Enzyme-linked immunosorbent assay (ELISA) was performed to measure the content of cytokines. Results Treatment with BBR considerably mitigated lung injury while improving survival, post-cecal ligation, and puncture (CLP). Treatment with BBR ameliorated pulmonary edema and hypoxemia in septic mice and inhibited the NF-κB signaling pathway. BBR also increased Treg cells and decreased Th17 proportions in the spleen and lung tissue of CLP-treated mice. Blocking Treg cells weakened the protective effect of BBR on sepsis-associated lung injury. Conclusion Overall, these results suggested that BBR is a potential therapeutic agent for sepsis.
Collapse
Affiliation(s)
- Longwang Chen
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Xinyong Liu
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, People’s Republic of China
| | - Xuetao Wang
- Department of Intensive Care Unit, Wenzhou Longwan District First People’s Hospital, Wenzhou, Zhejiang, People’s Republic of China
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Yumei Ye
- Department of Ultrasound Imaging, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Correspondence: Yumei Ye, Department of Ultrasound Imaging, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China, Tel +860577-5557-9410, Email
| |
Collapse
|
12
|
Fu H, Zhu H. Geniposidic acid protects lipopolysaccharide-induced acute lung injury via the TLR4/MyD88 signaling pathway in vitro and in vivo. Immunopharmacol Immunotoxicol 2022; 44:984-992. [PMID: 35770920 DOI: 10.1080/08923973.2022.2096465] [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: 12/13/2022]
Abstract
BACKGROUND Acute lung injury (ALI) is a common respiratory disease and is a serious threat to human health due to the lack of effective treatment. Geniposidic acid (GPA) is an iridoid glucoside extracted from Gardeniae jasminoides Ellis and can treat inflammation-related diseases. This study aimed to investigate the regulatory functions of GPA on lipopolysaccharide (LPS)-induced ALI and its potential mechanism, providing effective strategies for the clinical treatment of ALI. METHODS ALI models were constructed by LPS in Sprague-Dawley rats and pulmonary epithelial cells. The function of GPA was investigated by hematoxylin-eosin staining, lung function assessment, Western blot, Masson staining, and Sirius Red staining, quantitative real-time PCR, enzyme-linked immunosorbent assay, cell counting kit-8 assay, apoptosis analysis, and immunofluorescence assays. RESULTS Functionally, GPA increased survival, relieved pulmonary epithelial function in response to LPS, repressed pulmonary fibrosis and inflammation caused by ALI in vivo; GPA also repressed pulmonary epithelial cell injury and inflammation induced by LPS in vitro. Mechanistically, GPA decreased the protein levels of TLR4 and MyD88 and accelerated the nuclear export of p65, suggesting that GPA repressed the activation of p65. CONCLUSION GPA protected LPS-induced ALI through the TLR4/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Hui Fu
- Department of Pediatrics, Changzhou Second People's Hospital, Changzhou, China
| | - Hui Zhu
- Department of Pediatrics, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| |
Collapse
|
13
|
Luan R, Ding D, Yang J. The protective effect of natural medicines against excessive inflammation and oxidative stress in acute lung injury by regulating the Nrf2 signaling pathway. Front Pharmacol 2022; 13:1039022. [PMID: 36467050 PMCID: PMC9709415 DOI: 10.3389/fphar.2022.1039022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/02/2022] [Indexed: 09/29/2023] Open
Abstract
Acute lung injury (ALI) is a common critical disease of the respiratory system that progresses into acute respiratory distress syndrome (ARDS), with high mortality, mainly related to pulmonary oxidative stress imbalance and severe inflammation. However, there are no clear and effective treatment strategies at present. Nuclear factor erythroid 2-related factor 2(Nrf2) is a transcription factor that interacts with multiple signaling pathways and regulates the activity of multiple oxidases (NOX, NOS, XO, CYP) related to inflammation and apoptosis, and exhibits antioxidant and anti-inflammatory roles in ALI. Recently, several studies have reported that the active ingredients of natural medicines show protective effects on ALI via the Nrf2 signaling pathway. In addition, they are cheap, naturally available, and possess minimal toxicity, thereby having good clinical research and application value. Herein, we summarized various studies on the protective effects of natural pharmaceutical components such as polyphenols, flavonoids, terpenoids, alkaloids, and polysaccharides on ALI through the Nrf2 signaling pathway and demonstrated existing gaps as well as future perspectives.
Collapse
|
14
|
Cooper N, Ghanima W, Hill QA, Nicolson PLR, Markovtsov V, Kessler C. Recent advances in understanding spleen tyrosine kinase (SYK) in human biology and disease, with a focus on fostamatinib. Platelets 2022; 34:2131751. [DOI: 10.1080/09537104.2022.2131751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nichola Cooper
- Clinical Reader in Immune Haematology and Honorary Consultant, Faculty of Medicine, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Waleed Ghanima
- Head of Research and Consultant Haematologist, Department of Hemato-oncology, Østfold Hospital, and Department of Hematology, Institute of Clinical Medicine, Oslo University, Oslo, Norway
| | - Quentin A Hill
- Consultant Haematologist, Department of Haematology, Leeds Teaching Hospitals, Leeds, UK
| | - Phillip LR Nicolson
- Clinical Lecturer in Haematology, Institute of Cardiovascular Sciences, University of Birmingham, and Department of Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Vadim Markovtsov
- Translational Biology, Rigel Pharmaceuticals, South San Francisco, CA, USA
| | - Craig Kessler
- Medicine and Pathology, Director, Division of Coagulation, Director, Cellular and Therapeutic Apheresis and Cellular Collection, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
15
|
Tang J, Suo L, Li F, Bian K, Yang C, Wang Y. Transcriptome profiling of lung immune responses potentially related to acute respiratory distress syndrome in forest musk deer. BMC Genomics 2022; 23:701. [PMID: 36221054 PMCID: PMC9552132 DOI: 10.1186/s12864-022-08917-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 09/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background Forest musk deer is an endangered species globally. The death of captive forest musk deer can be caused by certain respiratory system diseases. Acute respiratory distress syndrome (ARDS) is a huge threat to the life of forest muck deer that breed in our department. Methods Lung histopathologic analysis was conducted by hematoxylin and eosin (HE) staining. The lung gene changes triggered by ARDS were examined by RNA sequencing and related bioinformatics analysis in forest musk deer. The potential functions of unigenes were investigated by NR, SwissProt KOG, GO, and KEGG annotation analyses. Vital biological processes or pathways in ARDS were examined by GO and KEGG enrichment analyses. Results A total of 3265 unigenes were differentially expressed (|log2fold-change|> 2 and adjusted P value < 0.01) in lung tissues of 3 forest musk deer with ARDS compared with normal lung tissues of the non-ARDS group. These differentially expressed unigenes (DEGs) played crucial roles in immunity and defense responses to pathogens. Moreover, we identified the DEGs related to one or more of the following biological processes: lung development, immunity, and bacterial/viral/fungal infection. And six DEGs that might be involved in lung injury caused by immune dysregulation or viral/fungal infection were identified. Conclusion ARDS-mediated lung gene alterations were identified in forest musk deer. Moreover, multiple genes involved in lung development and lung defense responses to bacteria/viruses/fungi in ARDS were filtered out in forest musk deer. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08917-7.
Collapse
Affiliation(s)
- Jie Tang
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China
| | - Lijuan Suo
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China
| | - Feiran Li
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China
| | - Kun Bian
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China
| | - Chao Yang
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China.
| | - Yan Wang
- Shaanxi Institute of Zoology, Xi'an710032, Shaanxi, China.,Shaanxi Provincial Field Observation & Research Station for Golden Monkey, Giant Panda and Biodiversity, Xi'an 723400, Shaanxi, China
| |
Collapse
|
16
|
Zeng M, Zhang B, Ren Y, Wang S, Guo P, Liu M, Zhang Q, Jia J, Li J, Zheng X, Feng W. A sesquiterpene isolated from the stems and leaves of Dioscorea opposita thunb. Transforms the composition of immune cells through ERβ in a mouse model of LPS-induced lung injury. Heliyon 2022; 8:e10500. [PMID: 36105471 PMCID: PMC9465438 DOI: 10.1016/j.heliyon.2022.e10500] [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: 10/11/2021] [Revised: 01/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Acute lung injury (ALI) is a common critical disease with a high mortality rate. Natural products have marked efficacy in the prevention and treatment of ALI, in addition, estrogen and its receptors are involved in the pathogenesis and development of lung injury. Our previous research shows that sesquiterpenes isolated from the stems and leaves of Dioscorea opposita Thunb. have anti-inflammatory and estrogenic-like activity. In the present study, sesquiterpene (A1) is a natural extract from the stems and leaves of Dioscorea opposita Thunb. with a view to determining whether A1 can improve lung function in a mouse model of LPS-induced ALI and exploring the involvement of the estrogen receptor β (ERβ) pathway. A1 (20 or 40 mg/kg, i. g., 2 times/day) was administered for 3 d, followed by the induction of ALI via an intratracheal LPS drip (5 mg/kg/2 h). The lung function and levels of inflammation, immune cells, apoptosis, and ERβ expression were examined. The antagonistic activity of specific ERβ blocker (THC, 1 μM) against A1 (20 μM) in co-cultured BEAS-2B cells and splenic lymphocytes induced with LPS (1 μg/mL, 24 h) was also investigated to assess whether the observed effects of A1 were mediated by ERβ. A1 improved lung function, regulated the immune system, and decreased inflammation and apoptosis. Moreover, A1 increased the expression of ERβ in LPS-induced mice, and antagonism of ERβ decreased the protective effects of A1 in a co-culture system. A1 had anti-ALI effects that might partially mediated through ERβ signaling. Our data provide molecular justification for the use of A1 in the treatment of ALI.
Collapse
Affiliation(s)
- Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yingjie Ren
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Pengli Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Qinqin Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jinyue Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| |
Collapse
|
17
|
Piperlongumin Improves Survival in the Mouse Model of Sepsis: Effect on Coagulation Factors and Lung Inflammation. Inflammation 2022; 45:2513-2528. [PMID: 35831643 PMCID: PMC9281243 DOI: 10.1007/s10753-022-01709-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/22/2022] [Indexed: 11/05/2022]
Abstract
Excessive inflammation and coagulation contribute to high morbidity and mortality in sepsis. Many studies have indicated the role of piperlongumine (PL) in anti-inflammation, but its effect on coagulation remains uncertain. Here, we explore whether PL could moderate coagulation indicators and alleviate lung inflammation during sepsis. RAW264.7 cells were induced by lipopolysaccharide (LPS) and treated with PL. Inflammatory and coagulation indicators, cell function and signaling, were evaluated in cells. Cecal ligation and puncture (CLP) mice were treated with PL by gavage. The harvested lungs and plasma were used to assess inflammation and coagulation indicators. As a result, PL increased the survival rate and reduced the concentrations of tissue factor (TF), plasminogen activator inhibitor 1 (PAI-1), thrombin-antithrombin complex (TAT), D-dimer, interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α in CLP mice, with fibrinogen in reverse. Moreover, the PL alleviated inflammation, fibrin deposition, and lung injury in the lungs of CLP mice. In vitro, PL downregulated the expression of TF, PAI-1, IL-6, TNF-α, and IL-1β in RAW264.7 cells induced by LPS. Furthermore, PL inhibited the phosphorylation of the AKT/mTOR signaling pathway's key proteins and suppressed the nuclear translocation of p-STAT3 in LPS-stimulated RAW264.7 cells. In conclusion, this study suggests that PL may modulate coagulation indicators and improve lung inflammation through AKT/mTOR signaling pathway in sepsis.
Collapse
|
18
|
Yang B, Ma L, Wei Y, Cui Y, Li X, Wei Y, Zhang S, Zhang L, Zhou H, Wang G, Li X. Isorhamnetin alleviates lipopolysaccharide-induced acute lung injury by inhibiting mTOR signaling pathway. Immunopharmacol Immunotoxicol 2022; 44:387-399. [PMID: 35306954 DOI: 10.1080/08923973.2022.2052892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Aim: Acute Lung Injury (ALI) is an acute hypoxic respiratory insufficiency caused by various traumatic factors, manifested as progressive hypoxemia and respiratory distress, and lung imaging shows a heterogeneous osmotic outbreak. Isorhamnetin (ISO) is a flavonoid compound isolated and purified from medicinal plants, such as Hippophae rhamnoides L. and Ginkgo, and has multiple pharmacological functions, such as anti-tumor, anti-myocardial hypoxia, and cardiovascular protection. Our previous study has shown that ISO could attenuate lipopolysaccharide (LPS)-induced acute lung injury in mice, but its mechanism is not clear.Methods: In this study, we used LPS-induced mouse and cell models to research the mechanism of ISO alleviating acute lung injury.Results: The results showed that ISO could attenuate the injury of type II alveolar epithelial cells by inhibiting the TLR4/NF-κB pathway. Further studies showed that ISO could inhibit the activation of mTOR signal in vivo and in vitro and promote autophagy in alveolar epithelial cells to reduce lung injury caused by LPS. In addition, ISO could inhibit LPS-induced epithelial cell apoptosis.Conclusion: Overall, ISO could suppress injury and apoptosis of epithelial cells and activate autophagy to protect epithelial cells via inhibiting mTOR signal and attenuating LPS-induced acute lung injury in mice.
Collapse
Affiliation(s)
- Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ling Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Yuli Wei
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Yunyao Cui
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Yiying Wei
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Shanshan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Liang Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Guangshun Wang
- Department of Thoracic Surgery, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| |
Collapse
|
19
|
Han S, Yuan R, Cui Y, He J, Wang QQ, Zhuo Y, Yang S, Gao H. Hederasaponin C Alleviates Lipopolysaccharide-Induced Acute Lung Injury In Vivo and In Vitro Through the PIP2/NF-κB/NLRP3 Signaling Pathway. Front Immunol 2022; 13:846384. [PMID: 35281058 PMCID: PMC8913935 DOI: 10.3389/fimmu.2022.846384] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/04/2022] Open
Abstract
Gene transcription is governed by epigenetic regulation that is essential for the pro-inflammatory mediators surge following pathological triggers. Acute lung injury (ALI) is driven by pro-inflammatory cytokines produced by the innate immune system, which involves the nod-like receptor 3 (NLRP3) inflammasome and nuclear factor-κB (NF-κB) pathways. These two pathways are interconnected and share a common inducer the phosphatidylinositol 4,5-bisphosphate (PIP2), an epigenetic regulator of (Ribosomal ribonucleic acid (rRNA) gene transcription, to regulate inflammation by the direct inhibition of NF-κB phosphorylation and NLRP3 inflammasome activation. Herein, we report that hederasaponin C (HSC) exerted a therapeutic effect against ALI through the regulation of the PIP2/NF-κB/NLRP3 signaling pathway. In lipopolysaccharide (LPS)/lipopolysaccharide + adenosine triphosphate (LPS+ATP)-stimulated macrophages, our results showed that HSC remarkably inhibited the secretion of interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-α (TNF-α). Moreover, HSC inhibited NF-κB/p65 nuclear translocation and the binding of PIP2 to transforming growth factor-β activated kinase 1 (TAK1). The intracellular calcium (Ca2+) level was decreased by HSC via the PIP2 signaling pathway, which subsequently inhibited the activation of NLRP3 inflammasome. HSC markedly alleviated LPS-induced ALI, restored lung function of mice, and rescued ALI-induced mice death. In addition, HSC significantly reduced the level of white blood cells (WBC), neutrophils, and lymphocytes, as well as pro-inflammatory mediators like IL-6, IL-1β, and TNF-α. Hematoxylin and eosin (H&E) staining results suggested HSC has a significant therapeutic effect on lung injury of mice. Interestingly, the PIP2/NF-κB/NLRP3 signaling pathway was further confirmed by the treatment of HSC with ALI, which is consistent with the treatment of HSC with LPS/LPS+ATP-stimulated macrophages. Overall, our findings revealed that HSC demonstrated significant anti-inflammatory activity through modulating the PIP2/NF-κB/NLRP3 axis in vitro and in vivo, suggesting that HSC is a potential therapeutic agent for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Shan Han
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Renyikun Yuan
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yushun Cui
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qin-Qin Wang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Youqiong Zhuo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Guangxi University of Chinese Medicine, Nanning, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Hongwei Gao,
| |
Collapse
|
20
|
Li R, Hu X, Chen H, Zhao Y, Gao X, Yuan Y, Guo H, Huang H, Zou X, Qi H, Liu H, Shang Y. Role of Cholinergic Anti-Inflammatory Pathway in Protecting Sepsis-Induced Acute Lung Injury through Regulation of the Conventional Dendritic Cells. Mediators Inflamm 2022; 2022:1474891. [PMID: 35125962 PMCID: PMC8813293 DOI: 10.1155/2022/1474891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway connects the immune response system and the nervous system via the vagus nerve. The key regulatory receptor is the α7-subtype of the nicotinic acetylcholine receptor (α7nAChR). Cholinergic anti-inflammatory pathway has been proved to be effective in suppressing the inflammation responses in acute lung injury (ALI). Dendritic cells (DCs), the important antigen-presenting cells, also express the α7nAChR. Past studies have indicated that reducing the quantity of mature conventional DCs and inhibiting the maturation of pulmonary DCs may prove effective for the treatment of ALI. However, the effects of cholinergic anti-inflammatory pathway on maturation, function, and quantity of DCs and conventional DCs in ALI remain unclear. OBJECTIVE It was hypothesized that cholinergic anti-inflammatory pathway may inhibit the inflammatory response of ALI by regulating maturation, phenotype, and quantity of DCs and conventional DCs. METHODS GTS-21 (GTS-21 dihydrochloride), an α7nAchR agonist, was prophylactically administered in sepsis-induced ALI mouse model and LPS-primed bone marrow-derived dendritic cells. The effects of GTS-21 were observed with respect to maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2 (type 2 conventional DCs) and the release of DC-related proinflammatory cytokines in vivo and in vitro. RESULTS The results of the present study revealed that GTS-21 treatment decreased the maturation of DCs and the production of DC-related proinflammatory cytokines in vitro and in sepsis-induced ALI mouse model; it reduced the quantity of CD11c+MHCII+ conventional DCs and CD11c+CD11b+ conventional DCs2 in vivo experiment. CONCLUSIONS Cholinergic anti-inflammatory pathway contributes to the reduction in the inflammatory response in ALI by regulating maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Huibin Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Yue Zhao
- Department of Critical Care Medicine, Jin Yin-tan Hospital, Wuhan, Hubei 430048, China
| | - Xuehui Gao
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Huiling Guo
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haiyan Huang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Liu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
21
|
Xie H, Xie S, Wang M, Wei H, Huang H, Xie A, Li J, Fang C, Shi F, Yang Q, Qi Y, Yin Z, Wang X, Huang J. Properties and Roles of γδT Cells in Plasmodium yoelii nigeriensis NSM Infected C57BL/6 Mice. Front Cell Infect Microbiol 2022; 11:788546. [PMID: 35127555 PMCID: PMC8811364 DOI: 10.3389/fcimb.2021.788546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background Many kinds of immune cells are involved in malaria infection. γδT cells represent a special type of immune cell between natural and adaptive immune cells that play critical roles in anti-parasite infection. Methods In this study, malaria infection model was constructed. Distribution of γδT cells in various immune organs and dynamic changes of γδT cells in the spleens of C57BL/6 mice after infection were detected by flow cytometry. And activation status of γδT cells was detected by flow cytometry. Then γδT cells in naive and infected mice were sorted and performed single-cell RNA sequencing (scRNA-seq). Finally, γδTCR KO mice model was constructed and the effect of γδT cell depletion on mouse T and B cell immunity against Plasmodium infection was explored. Results Here, splenic γδT cells were found to increase significantly on day 14 after Plasmodium yoelii nigeriensis NSM infection in C57BL/6 mice. Higher level of CD69, ICOS and PD-1, lower level of CD62L, and decreased IFN-γ producing after stimulation by PMA and ionomycin were found in γδT cells from infected mice, compared with naive mice. Moreover, 11 clusters were identified in γδT cells by scRNA-seq based t-SNE analysis. Cluster 4, 5, and 7 in γδT cells from infected mice were found the expression of numerous genes involved in immune response. In the same time, the GO enrichment analysis revealed that the marker genes in the infection group were involved in innate and adaptive immunity, pathway enrichment analysis identified the marker genes in the infected group shared many key signalling molecules with other cells or against pathogen infection. Furthermore, increased parasitaemia, decreased numbers of RBC and PLT, and increased numbers of WBC were found in the peripheral blood from γδTCR KO mice. Finally, lower IFN-γ and CD69 expressing CD4+ and CD8+ T cells, lower B cell percentage and numbers, and less CD69 expressing B cells were found in the spleen from γδTCR KO infected mice, and lower levels of IgG and IgM antibodies in the serum were also observed than WT mice. Conclusions Overall, this study demonstrates the diversity of γδT cells in the spleen of Plasmodium yoelii nigeriensis NSM infected C57BL/6 mice at both the protein and RNA levels, and suggests that the expansion of γδT cells in cluster 4, 5 and 7 could promote both cellular and humoral immune responses.
Collapse
Affiliation(s)
- Hongyan Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shihao Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixia Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - He Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anqi Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiajie Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feihu Shi
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Quan Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhinan Yin
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Xinhua Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xinhua Wang, ; Jun Huang,
| | - Jun Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xinhua Wang, ; Jun Huang,
| |
Collapse
|
22
|
Cao F, Wang C, Long D, Deng Y, Mao K, Zhong H. Network-Based Integrated Analysis of Transcriptomic Studies in Dissecting Gene Signatures for LPS-Induced Acute Lung Injury. Inflammation 2021; 44:2486-2498. [PMID: 34462829 PMCID: PMC8405180 DOI: 10.1007/s10753-021-01518-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/07/2021] [Indexed: 10/26/2022]
Abstract
Acute lung injury (ALI) is a type of serious clinical syndrome leading to morbidity and mortality. However, the precise pathogenesis of ALI remains elusive. Here, we implemented an integrative meta-analysis of six GEO microarray studies with 76 samples in the ALI mouse model. A total of 958 differentially expressed genes (DEGs) were identified in LPS relative to normal samples. Then, a network-based meta-analysis was used to mine core DEGs and to unfold the interactions among these genes. We found that Ebi3 was the top upregulated genes in the LPS-induced ALI. GO, KEGG, and GSEA analyses were performed for functional annotation. qRT-PCR revealed augmented expression of six candidate genes (Stat1, Syk, Jak3, Rac2, Ripk1, and Traf6) in the established ALI mouse model with LPS exposure. Taken together, our study investigated comprehensively hub DEGs and their networks for LPS-stimulated ALI, which might afford an additional approach to determine biomarkers and therapeutic targets and explore the molecular pathophysiology toward ALI.
Collapse
Affiliation(s)
- Fang Cao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Huichuan District, 149 Dalian Road, Zunyi, Guizhou, 563003 China
| | - Chunyan Wang
- Department of Gastroenterology, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology, Chengdu, 610000 Sichuan China
| | - Danling Long
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 Hubei China
| | - Yujuan Deng
- School of Computer Science and Engineering, Shijiazhuang University, Shijiazhuang, Hebei China
| | - Kaimin Mao
- Department of Critical Care Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200127 China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Wuhan, 430072 Hubei China
| |
Collapse
|
23
|
Chu SJ, Tang SE, Pao HP, Wu SY, Liao WI. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol 2021; 12:752507. [PMID: 34658893 PMCID: PMC8514687 DOI: 10.3389/fphar.2021.752507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
Protease-activated receptor (PAR)-1 is a thrombin-activated receptor that plays an essential role in ischemia/reperfusion (IR)-induced acute inflammation. PAR-1 antagonists have been shown to alleviate injuries in various IR models. However, the effect of PAR-1 antagonists on IR-induced acute lung injury (ALI) has not yet been elucidated. This study aimed to investigate whether PAR-1 inhibition could attenuate lung IR injury. Lung IR was induced in an isolated perfused rat lung model. Male rats were treated with the specific PAR-1 antagonist SCH530348 (vorapaxar) or vehicle, followed by ischemia for 40 min and reperfusion for 60 min. To examine the role of PAR-1 and the mechanism of SCH530348 in lung IR injury, western blotting and immunohistochemical analysis of lung tissue were performed. In vitro, mouse lung epithelial cells (MLE-12) were treated with SCH530348 or vehicle and subjected to hypoxia-reoxygenation (HR). We found that SCH530348 decreased lung edema and neutrophil infiltration, attenuated thrombin production, reduced inflammatory factors, including cytokine-induced neutrophil chemoattractant-1, interleukin-6 and tumor necrosis factor-α, mitigated lung cell apoptosis, and downregulated the phosphoinositide 3-kinase (PI3K), nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in IR-injured lungs. In addition, SCH530348 prevented HR-induced NF-κB activation and inflammatory chemokine production in MLE12 cells. Our results demonstrate that SCH530348 exerts protective effects by blocking PAR-1 expression and modulating the downstream PI3K, NF-κB and MAPK pathways. These findings indicate that the PAR-1 antagonist protects against IR-induced ALI and is a potential therapeutic candidate for lung protection following IR injury.
Collapse
Affiliation(s)
- Shi-Jye Chu
- Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
24
|
Hughes DM, Toste C, Nelson C, Escalon J, Blevins F, Shah B. Transitioning From Thrombopoietin Agonists to the Novel SYK Inhibitor Fostamatinib: A Multicenter, Real-World Case Series. J Adv Pract Oncol 2021; 12:508-517. [PMID: 34430061 PMCID: PMC8299792 DOI: 10.6004/jadpro.2021.12.5.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Fostamatinib is an oral spleen tyrosine kinase (SYK) inhibitor used for the treatment of adult patients with chronic immune thrombocytopenia (ITP) who have had an insufficient response to a previous treatment. Clinical and operational barriers may exist that warrant bridging or switching from thrombopoietin receptor agonists (TPO-RAs), such as volatility or unpredictability of platelets, adverse events, and quality of life or patient preference. While fostamatinib demonstrated durable platelet responses, the safety and efficacy in combination, bridging, and/or switching with TPO-RAs is not well documented. The objective of this article is to provide guidance from real-world case studies for a safe and effective strategy for the transitioning of patients from TPO-RAs to fostamatinib, with some degree of overlap between the two agents. Currently, the evidence does not exist to guide the safe and effective use of combination therapy or transition between therapies in ITP. This case series highlights the importance to further understand the complexities of managing this disease, as well as successfully combining, bridging, and/or switching patients over to fostamatinib without the need for rescue therapy or increase in adverse events. The need for real-world evidence that guides providers on the safety and efficacy of short- and long-term combination therapy of fostamatinib and TPO-RAs is crucial. The rationale for combination therapy is to target different pathways, platelet destruction, and production without added toxicities. Additionally, gradual tapering off of TPO-RAs may provide a more favorable clinical outcome when switching to fostamatinib. The need for additional data is necessary to provide clinicians with guidance when managing patients with ITP.
Collapse
Affiliation(s)
- David M Hughes
- Department of Pharmacy, Boston Medical Center, Boston, Massachusetts
| | | | | | | | - Frances Blevins
- Department of Medicine, Boston University, Boston, Massachusetts
| | - Bhavesh Shah
- Department of Pharmacy, Boston Medical Center, Boston, Massachusetts
| |
Collapse
|
25
|
Kleinveld DJB, Botros L, Maas MAW, Kers J, Aman J, Hollmann MW, Juffermans NP. Bosutinib reduces endothelial permeability and organ failure in a rat polytrauma transfusion model. Br J Anaesth 2021; 126:958-966. [PMID: 33685634 PMCID: PMC8258973 DOI: 10.1016/j.bja.2021.01.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Trauma-induced shock is associated with endothelial dysfunction. We examined whether the tyrosine kinase inhibitor bosutinib as an adjunct therapy to a balanced blood component resuscitation strategy reduces trauma-induced endothelial permeability, thereby improving shock reversal and limiting transfusion requirements and organ failure in a rat polytrauma transfusion model. METHODS Male Sprague-Dawley rats (n=13 per group) were traumatised and exsanguinated until a MAP of 40 mm Hg was reached, then randomised to two groups: red blood cells, plasma and platelets in a 1:1:1 ratio with either bosutinib or vehicle. Controls were randomised to sham (median laparotomy, no trauma) with bosutinib or vehicle. Organs were harvested for histology and wet/dry (W/D) weight ratio. RESULTS Traumatic injury resulted in shock, with higher lactate levels compared with controls. In trauma-induced shock, the resuscitation volume needed to obtain a MAP of 60 mm Hg was lower in bosutinib-treated animals (2.8 [2.7-3.2] ml kg-1) compared with vehicle (6.1 [5.1-7.2] ml kg-1, P<0.001). Lactate levels in the bosutinib group were 2.9 [1.7-4.8] mM compared with 6.2 [3.1-14.1] mM in the vehicle group (P=0.06). Bosutinib compared with vehicle reduced lung vascular leakage (W/D ratio of 5.1 [4.6-5.3] vs 5.7 [5.4-6.0] (P=0.046) and lung injury scores (P=0.027). CONCLUSIONS Bosutinib as an adjunct therapy to a balanced transfusion strategy reduced resuscitation volume, improved shock reversal, and reduced vascular leak and organ injury in a rat polytrauma model.
Collapse
Affiliation(s)
- Derek J B Kleinveld
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Liza Botros
- Department of Pulmonary Diseases, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M Adrie W Maas
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Pathology, Leiden University Medical Center, University of Leiden, Leiden, The Netherlands; Van't Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, The Netherlands; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jurjan Aman
- Department of Pulmonary Diseases, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Trauma Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Intensive Care Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Li J, Wang K, Huang B, Li R, Wang X, Zhang H, Tang H, Chen X. The receptor for advanced glycation end products mediates dysfunction of airway epithelial barrier in a lipopolysaccharides-induced murine acute lung injury model. Int Immunopharmacol 2021; 93:107419. [PMID: 33548580 DOI: 10.1016/j.intimp.2021.107419] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Airway epithelial cells (AECs) act as the first barrier protecting against invasion of environment agents and maintain integrity of lung structure and function. Dysfunction of airway epithelial barrier has been shown to be involved in ALI/ARDS pathogenesis. Yet, the exact mechanism is still obscure. Our study evaluated whether the receptor for advanced glycation end products (RAGE) mediates impaired airway epithelial barrier in LPS-induced murine ALI model. METHODS Male BALB/c mice were subjected to intratracheal instillation of LPS to generate an ALI model. Inhibitors of RAGE, FPS-ZM1 and Azeliragon were respectively given to the mice through intraperitoneal injection. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected for further analysis. RESULTS LPS exposure led to markedly increased expression of RAGE and its ligands HMGB1, HSP70, S100b. Treatment of FPS-ZM1 or Azeliragon not only effectively descended the expression of RAGE and its ligands but also attenuated LPS-induced neutrophil-predominant airway inflammation and injury, decreased levels of IL-6, IL-1β and TNF-α in BALF, alleviated increased alveolar-capillary permeability and pulmonary edema. LPS stimulation significantly impaired the integrity of airway epithelium, paralleled with dislocation of adheren junction (AJ) protein E-cadherin at cell-cell contacts and down-expression of both AJ and tight junction (TJ) proteins Claudin-2 and occludin, all of which were dramatically rescued by RAGE inhibition. CONCLUSION RAGE signaling mediates airway epithelial barrier dysfunction in a LPS-induced ALI murine model.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Kai Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Bo Huang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Xilong Wang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Hailing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| |
Collapse
|
27
|
Wang T, Lin S, Liu R, Li H, Liu Z, Zhang X, Xu H, Li Q, Bi K. Metabolomic profile perturbations of serum, lung, bronchoalveolar lavage fluid, spleen and feces in LPS-induced acute lung injury rats based on HPLC-ESI-QTOF-MS. Anal Bioanal Chem 2020; 412:1215-1234. [PMID: 31940090 DOI: 10.1007/s00216-019-02357-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
Acute lung injury (ALI) is a clinically common and serious disease, underscoring the urgent need for clarification of its pathogenesis. According to traditional Chinese medicine (TCM) theories on the "lung-spleen-intestine axis" and its correlation with ALI, a high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (HPLC-QTOF-MS) metabolomic platform was applied to identify biomarkers from five bio-samples of control and model rats challenged with intratracheally administered lipopolysaccharide (LPS) based on multivariate mathematical statistical analysis. As a result, 19, 24, 24, 15 and 29 altered metabolites were identified in serum, lung, bronchoalveolar lavage fluid (BALF), spleen and feces samples, respectively. Metabolic pathway analysis showed that linoleic acid, sphingolipid, glycerophospholipid and bile acid metabolism pathways were mainly altered by ALI. Additionally, ROC curves were applied to assess the specificity and sensitivity of the biomarkers. ALI characteristic metabolomic spectra were then established to differentiate the control from the model group with a similarity discriminative threshold of 0.7. Additionally, to compare the metabolomic profiles of the five bio-samples and establish metabolic similarities and differences among them, correlation analysis was conducted in order to delineate an objective law of endogenous linkage along the lung-spleen-intestine axis. Therefore, this study provides insights into the mechanisms involved in ALI from a metabolomics perspective, which can be applied in characterization of the mechanism and early disease detection. Graphical abstract.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Song Lin
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Hua Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Zihan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xinnong Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China.
| |
Collapse
|