1
|
Xu H, Xu H, Li W, Liang Z, Luo W, Sheng S, Liang G, Zhang Z. Modulating the NLRP3 Inflammasome: Acitretin as a potential treatment for Sepsis-induced acute lung injury. Int Immunopharmacol 2025; 153:114504. [PMID: 40187888 DOI: 10.1016/j.intimp.2025.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Acitretin, a well-established dermatological drug primarily used for psoriasis treatment, has been clinically used for several decades. However, its potential role in modulating inflammation in sepsis remains unexplored. OBJECTIVE This study seeks to explore the impact of acitretin on sepsis-induced acute lung injury (ALI) and to elucidate the underlying mechanisms involved. METHODS In a mouse model of sepsis induced by lipopolysaccharide (LPS), we assessed the effects of acitretin on ALI. Transcriptome sequencing of lung tissue was performed to identify relevant signaling pathways. In vitro, bone marrow-derived macrophages (BMDMs) were treated with acitretin (1 μM, 5 μM and 10 μM) to evaluate its impact on NOD-, LRR- and pyrin domain-containing protein 3(NLRP3) inflammasome activation and pyroptosis. In vivo, wild-type, Nlrp3 knockout, and Gsdmd knockout mice were used to confirm the role of the NLRP3 inflammasome in mediating acitretin's effects. RESULTS Acitretin significantly mitigated sepsis-induced ALI, reducing mortality in LPS-challenged mice. Transcriptome analysis revealed that acitretin suppressed the NLRP3 inflammasome pathway in lung tissue. In vitro, acitretin dose-dependently inhibited interleukin (IL)-1β release, caspase-1 p20 production, and GSDMD cleavage in BMDMs. Furthermore, acitretin inhibited inflammasome activation by preventing ASC oligomerization and its interaction with NLRP3. In vivo, acitretin reduced lung tissue inflammation, IL-1β levels in bronchoalveolar lavage fluid, and the ratio of wet to dry in wide-type mice, but these effects were abolished in Nlrp3 and Gsdmd knockout mice. CONCLUSION Acitretin demonstrated significant anti-inflammatory properties through the suppression of the NLRP3 inflammasome, suggesting its potential as a therapeutic strategy for sepsis and related complications.
Collapse
Affiliation(s)
- Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haowen Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weifeng Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Zhiyu Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Weiwei Luo
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiying Sheng
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China.
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Zhang J, Zhang J, Yao Z, Shao W, Song Y, Tang W, Li B. GAMG ameliorates silica-induced pulmonary inflammation and fibrosis via the regulation of EMT and NLRP3/TGF-β1/Smad signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117124. [PMID: 39342756 DOI: 10.1016/j.ecoenv.2024.117124] [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: 06/06/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Silicosis is an occupational disease caused by exposure to silica characterized by pulmonary inflammation and fibrosis, for which there is a lack of effective drugs. Glycyrrhetinic acid 3-O-β-D-glucuronide (GAMG) can treat silicosis due to its anti-inflammatory and anti-fibrotic properties. Here, the effect of therapeutic interventions of GAMG was evaluated in early-stage and advanced silicosis mouse models. GAMG significantly improved fibrotic pathological changes and collagen deposition in the lungs, alleviated lung inflammation in the BALF, reduced the expression of TNF-α, IL-6, NLRP3, TGF-β1, vimentin, Col-Ⅰ, N-cadherin, and inhibited epithelial-mesenchymal transition (EMT), thereby ameliorating pulmonary fibrosis. Moreover, the dose of 100 mg/kg GAMG can effectively prevent early-stage silicosis, while that of 200 mg/kg was recommended for advanced silicosis. In vitro and in vivo study verified that GAMG can suppress EMT through the NLRP3/TGF-β1/Smad2/3 signaling pathway. Therefore, GAMG could be a promising preventive (early-stage silicosis) and therapeutic (advanced silicosis) strategy, which provides a new idea for formulating prevention and treatment strategies.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China; Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China.
| | - Jiazhen Zhang
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Zongze Yao
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Wei Shao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuanchao Song
- Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China
| | - Wenjian Tang
- Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Bo Li
- Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China.
| |
Collapse
|
3
|
Chen PR, Li CY, Yazal T, Chen IC, Liu PL, Chen YT, Liu CC, Lo J, Lin TC, Chang CT, Wu HE, Chen YR, Cheng WC, Chiu CC, Chen CS, Wang SC. Protective effects of nordalbergin against LPS-induced endotoxemia through inhibiting MAPK/NF-κB signaling pathway, NLRP3 inflammasome activation, and ROS production. Inflamm Res 2024; 73:1657-1670. [PMID: 39052062 DOI: 10.1007/s00011-024-01922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Nordalbergin is a coumarin extracted from Dalbergia sissoo DC. To date, the biological effects of nordalbergin have not been well investigated. To investigate the anti-inflammatory responses and the anti-oxidant abilities of nordalbergin using lipopolysaccharide (LPS)-activated macrophages and LPS-induced sepsis mouse model. MATERIALS AND METHODS Production of nitrite oxide (NO), prostaglandin E2 (PGE2), pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β), reactive oxygen species (ROS), tissue damage and serum inflammatory markers, and the activation of the NLRP3 inflammasome were examined. RESULTS Our results indicated that nordalbergin reduced the production of NO and pro-inflammatory cytokines in vitro and ex vivo. Nordalbergin also suppressed iNOS and cyclooxygenase-2 expressions, decreased NF-κB activity, and attenuated MAPKs signaling pathway activation by decreasing JNK and p38 phosphorylation by LPS-activated J774A.1 macrophages. Notably, nordalbergin diminished NLRP3 inflammasome activation via repressing the maturation of IL-1β and caspase-1 and suppressing ROS production by LPS/ATP- and LPS/nigericin-activated J774A.1 macrophages. Furthermore, nordalbergin exhibited protective effects against the infiltration of inflammatory cells and also inhibited the levels of organ damage markers (AST, ALT, BUN) by LPS-challenged mice. CONCLUSION Nordalbergin possesses anti-inflammatory effects in macrophage-mediated innate immune responses, alleviates ROS production, decreases NLRP3 activation, and exhibits protective effects against LPS-induced tissue damage in mice.
Collapse
Grants
- NSTC 111-2218-E-037-001, NSTC 111-2314-B-037-071-MY3, NSTC 112-2314-B-037-127, NSTC 112-2314-B-037-128, NSTC 112-2926-I-037-501-G, NSTC 112-2314-B-037-089, NSTC 112-2311-B-039 -001, NSTC 112-2622-E-039-001, NSTC 111-2622-E-039-004 and NSTC 112-2218-E-037-001 National Science and Technology Council, Taiwan, R.O.C.
- KT113P010 NTHU-KMU Joint Research Project
- CMU111-IP-04 and CMU112-MF-25 China Medical University, Taiwan
- DMR-112-056, DMR-113-190, and DMR-113-191 China Medical University Hospital
Collapse
Affiliation(s)
- Pin-Rong Chen
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
| | - Taha Yazal
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - I-Chen Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427213, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Chi Mei Medical Center, Tainan, 71004, Taiwan
| | - Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Tzu-Chieh Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Tang Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yuan-Ru Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40403, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Academia Sinica, Taipei, 40403, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shu-Chi Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
4
|
李 明, 张 玮, 华 梦. [Bardoxolone methyl alleviates acute liver injury in mice by inhibiting NLRP3 inflammasome activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1662-1669. [PMID: 39505333 PMCID: PMC11744081 DOI: 10.12122/j.issn.1673-4254.2024.09.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To investigate the inhibitory effect of bardoxolone methyl (CDDO-Me) on activation of NLRP3 inflammasome and its mechanism for alleviating acute liver injury (ALI). METHODS Mouse bone marrow-derived macrophages (BMDM) and THP-1 cells were pre-treated with CDDO-Me followed by treatment with Nigericin, ATP, MSU, intracellular LPS transfection for activation of NLRP3 inflammasomes, or poly A: T for activation of AIM2 inflammasomes. The levels of caspase-1 and IL-1β in the cell culture supernatant was determined with Western blotting and ELISA to assess the inhibitory effect of CDDO-Me on NLRP3 inflammasomes and its specificity. In the animal experiment, male C57BL/6J mouse models of acetaminophen-induced ALI were treated with low-dose (20 mg/kg) and high-dose (40 mg/kg) CDDO-Me, and the changes in serum levels of IL-1β, TNF- α, AST and ALT were measured by ELISA and liver tissue pathology was observed using HE staining. RESULTS In mouse BMDM and THP-1 cells, CDDO-Me dose-dependently inhibited the activation of NLRP3 inflammasomes without significantly affecting the secretion of non-inflammasome-related inflammatory factors IL-6 and TNF-α or AIM2 inflammasome activation. In the mouse models of ALI, CDDO-Me treatment at both the low and high doses significantly reduced serum levels of IL-1β, AST and ALT, ameliorated histological changes and reduced inflammatory cell infiltration in the liver tissue, and the effects exhibited a distinct dose dependence. CONCLUSION CDDO-Me can specifically inhibit the activation of NLRP3 inflammasomes to alleviate acetaminophen-induced ALI in mice.
Collapse
Affiliation(s)
- 明远 李
- 蚌埠医科大学慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical University, Bengbu 233030, China
- 蚌埠医科大学第一附属医院检验科,安徽 蚌埠 233004Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - 玮 张
- 蚌埠医科大学慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical University, Bengbu 233030, China
- 蚌埠医科大学第一附属医院检验科,安徽 蚌埠 233004Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - 梦晴 华
- 蚌埠医科大学慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical University, Bengbu 233030, China
| |
Collapse
|
5
|
Tan S, Gu Y, Zhu Y, Luo C, Li Z, Lin H, Zhang W. Insights from bioinformatics analysis reveal that lipopolysaccharide induces activation of chemokine-related signaling pathways in human nasal epithelial cells. Sci Rep 2024; 14:7672. [PMID: 38561377 PMCID: PMC10984988 DOI: 10.1038/s41598-024-58317-y] [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: 01/03/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Lipopolysaccharide (LPS) is known to elicit a robust immune response. This study aimed to investigate the impact of LPS on the transcriptome of human nasal epithelial cells (HNEpC). HNEpC were cultured and stimulated with LPS (1 μg/mL) or an equivalent amount of normal culture medium. Subsequently, total RNA was extracted, purified, and sequenced using next-generation RNA sequencing technology. Differentially expressed genes (DEGs) were identified and subjected to functional enrichment analysis. A protein-protein interaction (PPI) network of DEGs was constructed, followed by Ingenuity Pathway Analysis (IPA) to identify molecular pathways influenced by LPS exposure on HNEpC. Validation of key genes was performed using quantitative real-time PCR (qRT-PCR). A total of 97 DEGs, comprising 48 up-regulated genes and 49 down-regulated genes, were identified. Results from functional enrichment analysis, PPI, and IPA indicated that DEGs were predominantly enriched in chemokine-related signaling pathways. Subsequent qRT-PCR validation demonstrated significant upregulation of key genes in these pathways in LPS-treated HNEpC compared to control cells. In conclusion, LPS intervention profoundly altered the transcriptome of HNEpC, potentially exacerbating inflammatory responses through the activation of chemokine-related signaling pathways.
Collapse
Affiliation(s)
- Shaolin Tan
- Postgraduate Training Base of Shanghai Sixth People's Hospital, Jinzhou Medical University, Shanghai, China
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yuelong Gu
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Ying Zhu
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Chunyu Luo
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Zhipeng Li
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Hai Lin
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| | - Weitian Zhang
- Department of Otolaryngology-Head and Neck Surgery & Allergy Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Otolaryngological Institute, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| |
Collapse
|
6
|
Liu X, Chen L, Peng W, Deng H, Ni H, Tong H, Hu H, Wang S, Qian J, Liang A, Chen K. Th17/Treg balance: the bloom and wane in the pathophysiology of sepsis. Front Immunol 2024; 15:1356869. [PMID: 38558800 PMCID: PMC10978743 DOI: 10.3389/fimmu.2024.1356869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Sepsis is a multi-organ dysfunction characterized by an unregulated host response to infection. It is associated with high morbidity, rapid disease progression, and high mortality. Current therapies mainly focus on symptomatic treatment, such as blood volume supplementation and antibiotic use, but their effectiveness is limited. Th17/Treg balance, based on its inflammatory property, plays a crucial role in determining the direction of the inflammatory response and the regression of organ damage in sepsis patients. This review provides a summary of the changes in T-helper (Th) 17 cell and regulatory T (Treg) cell differentiation and function during sepsis, the heterogeneity of Th17/Treg balance in the inflammatory response, and the relationship between Th17/Treg balance and organ damage. Th17/Treg balance exerts significant control over the bloom and wanes in host inflammatory response throughout sepsis.
Collapse
Affiliation(s)
- Xinyong Liu
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Longwang Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Peng
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongsheng Deng
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongying Ni
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongjie Tong
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hangbo Hu
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shengchao Wang
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jin Qian
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Andong Liang
- Nursing Faculty, School of Medicine, Jinhua Polytechnic, Jinhua, China
| | - Kun Chen
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|