1
|
Wang Q, He W, Zhou Y, Feng R, Wang Y, Liu L, Yuan Y, Dai J, Liu Y, Zhang X. Polystyrene nanoplastics aggravate house dust mite induced allergic airway inflammation through EGFR/ERK-dependent lung epithelial barrier dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118329. [PMID: 40381396 DOI: 10.1016/j.ecoenv.2025.118329] [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: 12/18/2024] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Concerns that airborne micro- and nanoplastics (MNPs) may impair human respiratory health are rising. However, the specific effects of MNPs on allergic asthma remain insufficiently explored. This study developed an allergic asthma model using house dust mite (HDM), and mice were exposed to 50 μg polystyrene nanoparticles (PS-NPs) at three-days interval. Additionally, the effects and potential mechanisms of PS-NPs exposure (25, 50 and 100 μg/mL) on lung epithelial barrier dysfunction were explored using mouse lung epithelial type II (MLE-12) and A549 cells. The pathological changes of airway tissue and the increase of inflammatory response confirmed that exposure to PS-NPs significantly aggravated allergic asthma in mice. Importantly, in the presence of HDM sensitization, the accumulation of PS-NPs in the alveolar region was increased, leading to lung epithelial barrier dysfunction and more Th2-mediated eosinophilic inflammation, characterized by elevated IL-4, IL-13, immunoglobulin E (Ig E) and eosinophils. The activation of the epidermal growth factor receptor (EGFR) pathway and its downstream extracellular regulating kinase (ERK) was investigated using transcriptomic sequencing to elucidate the effects of PS-NPs exposure on lung epithelial barrier dysfunction. Furthermore, an EGFR-specific inhibitor AG1478 was employed to confirm the role of the EGFR/ERK pathway in lung epithelial barrier dysfunction and asthma exacerbation in vitro and in vivo experiments. In conclusion, the molecular mechanism by which PS-NPs aggravates asthma in mice was elucidated, which helps to improve the understanding of the health effects of PS-NPs and lays a theoretical foundation for addressing the health risks posed by PS-NPs.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Wen He
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
| | - Rui Feng
- Shanghai Key Laboratory of Intelligent Information Processing, School of Computer Science, Fudan University, Shanghai 200433, China
| | - Yingwen Wang
- Department of Nursing, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Lijuan Liu
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yuan Yuan
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Jiajia Dai
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200032, China.
| | - Xiaobo Zhang
- Department of Respiratory Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China; Center for Pediatric Clinical Quality Control of Shanghai, Shanghai 201102, China.
| |
Collapse
|
2
|
Sun ZC, Liao R, Xian C, Lin R, Wang L, Fang Y, Zhang Z, Liu Y, Wu J. Natural pachypodol integrated, lung targeted and inhaled lipid nanomedicine ameliorates acute lung injury via anti-inflammation and repairing lung barrier. J Control Release 2024; 375:300-315. [PMID: 39265826 DOI: 10.1016/j.jconrel.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a high-mortality disease caused by multiple disorders such as COVID-19, influenza, and sepsis. Current therapies mainly rely on the inhalation of nitric oxide or injection of pharmaceutical drugs (e.g., glucocorticoids); however, their toxicity, side effects, or administration routes limit their clinical application. In this study, pachypodol (Pac), a hydrophobic flavonol with anti-inflammatory effects, was extracted from Pogostemon cablin Benth and intercalated in liposomes (Pac@liposome, Pac-lipo) to improve its solubility, biodistribution, and bioavailability, aiming at enhanced ALI/ARDS therapy. Nanosized Pac-lipo was confirmed to have stable physical properties, good biodistribution, and reliable biocompatibility. In vitro tests proved that Pac-lipo has anti-inflammatory property and protective effects on endothelial and epithelial barriers in lipopolysaccharide (LPS)-induced macrophages and endothelial cells, respectively. Further, the roles of Pac-lipo were validated on treating LPS-induced ALI in mice. Pac-lipo showed better effects than did Pac alone on relieving ALI phenotypes: It significantly attenuated lung index, improved pulmonary functions, inhibited cytokine expression such as TNF-α, IL-6, IL-1β, and iNOS in lung tissues, alleviated lung injury shown by HE staining, reduced protein content and total cell number in bronchoalveolar lavage fluid, and repaired lung epithelial and vascular endothelial barriers. As regards the underlying mechanisms, RNA sequencing results showed that the effects of the drugs were associated with numerous immune- and inflammation-related signaling pathways. Molecular docking and western blotting demonstrated that Pac-lipo inhibited the activation of the TLR4-MyD88-NF-κB/MAPK signaling pathway. Taken together, for the first time, our new drug (Pac-lipo) ameliorates ALI via inhibition of TLR4-MyD88-NF-κB/MAPK pathway-mediated inflammation and disruption of lung barrier. These findings may provide a promising strategy for ALI treatment in the clinic.
Collapse
Affiliation(s)
- Zhi-Chao Sun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Ran Liao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Caihong Xian
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Ran Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Liying Wang
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifei Fang
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhongde Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China.
| | - Yuntao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China.
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; Division of Life Science, The Hong Kong University of Science and Technology, 999077, Hong Kong SAR.
| |
Collapse
|
3
|
Lu M, Guo XW, Zhang FF, Wu DH, Xie D, Luo FQ. Dexmedetomidine ameliorates diabetic intestinal injury by promoting the polarization of M2 macrophages through the MMP23B pathway. World J Diabetes 2024; 15:1962-1978. [PMID: 39280187 PMCID: PMC11372634 DOI: 10.4239/wjd.v15.i9.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Diabetes is often associated with gastrointestinal dysfunctions, which can lead to hypoglycemia. Dexmedetomidine (DEX) is a commonly used sedative in perioperative diabetic patients and may affect gastrointestinal function. AIM To investigate whether sedative doses of DEX alleviate diabetes-caused intestinal dysfunction. METHODS Sedation/anesthesia scores and vital signs of streptozotocin (STZ)-induced diabetic mice under DEX sedation were observed. Diabetic mice were divided into saline and DEX groups. After injecting sedatives intraperitoneally, tight junctions (TJs) and apoptotic levels were evaluated 24 hours later to assess the intestinal barrier function. The role of DEX was validated using Villin-MMP23B flox/flox mice with intestinal epithelial deletion. In vitro, high glucose and hyperosmolarity were used to culture Caco-2 monolayer cells with STZ inter-vention. Immunofluorescence techniques were used to monitor the barrier and mitochondrial functions. RESULTS MMP23B protein levels in the intestinal tissue of STZ-induced diabetic mice were significantly higher than those in the intestinal tissue of control mice, with the DEX group displaying decreased MMP23B levels. Diabetes-mediated TJ dis-ruption, increased intestinal mucosal permeability, and systemic inflammation in wild-type mice might be reversed by DEX. In Caco-2 cells, MMP23B was associated with increased reactive oxygen species accumulation, mitochondrial membrane potential depolarization, and TJ disruption. CONCLUSION DEX reduces MMP23B, which may potentially contribute to STZ-induced intestinal barrier dysfunction, affecting TJ modification through mitochondrial dysfunction.
Collapse
Affiliation(s)
- Man Lu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Xiao-Wen Guo
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Fang-Fang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Dan-Hong Wu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Di Xie
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Feng-Qin Luo
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
4
|
Wu Z, Xi Q, Zhao Q, Zhu S. GDF11 OVEREXPRESSION ALLEVIATES SEPSIS-INDUCED LUNG MICROVASCULAR ENDOTHELIAL BARRIER DAMAGE BY ACTIVATING SIRT1/NOX4 SIGNALING TO INHIBIT FERROPTOSIS. Shock 2024; 62:245-254. [PMID: 38920138 DOI: 10.1097/shk.0000000000002391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
ABSTRACT Sepsis is a lethal clinical syndrome, and acute lung injury (ALI) is the earliest and most serious complication. We aimed to explore the role of growth differentiation factor 11 (GDF11) in sepsis-induced dysfunction of lung microvascular endothelial barrier in vivo and in vitro to elucidate its potential mechanism related to sirtuin 1 (SIRT1)/NADPH oxidase 4 (NOX4) signaling. Cecal ligation and puncture (CLP)-induced sepsis mice and lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cells (PMECs) were used in this study. Histopathological changes in lung tissues were tested by hematoxylin-eosin staining. Lung wet-to-dry weight ratio and inflammatory factors contents in bronchoalveolar lavage fluid were assessed. Evens blue index, trans-epithelial electrical resistance, and expression of zona occludens 1 (ZO-1), occludin-1, and claudin-1 were used to evaluate alveolar barrier integrity. Reactive oxygen species, lipid peroxidation, and ferroptosis markers were analyzed. Iron deposition in the lung tissues was assessed using Prussian blue staining. Intracellular Fe 2+ level was detected using FerroOrange staining. Additionally, expression of GDF11, SIRT1, and NOX4 was estimated with western blot. Then, EX527, a SIRT1 inhibitor, was employed to treat GDF11-overexpressed PMECs with LPS stimulation to clarify the regulatory mechanism. Results showed that GDF11 overexpression attenuated sepsis-induced pathological changes and inflammation and maintained alveolar barrier integrity. Moreover, GDF11 overexpression inhibited ferroptosis, upregulated SIRT1 expression and downregulated NOX4 expression. Additionally, EX527 treatment relieved the impacts of GDF11 overexpression on ferroptosis and destruction of integrity of human pulmonary microvascular endothelial cells exposed to LPS. Taken together, GDF11 overexpression could alleviate sepsis-induced lung microvascular endothelial barrier damage by activating SIRT1/NOX4 signaling to inhibit ferroptosis. Our findings potentially provide new molecular target for clinical therapy of ALI.
Collapse
Affiliation(s)
- Zhixiang Wu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | |
Collapse
|
5
|
Zhang Q, Zhang B, Yang F, Hu Y, Fan R, Wang M, Chen S. Forsythoside A regulates pulmonary fibrosis by inhibiting endothelial-to-mesenchymal transition and lung fibroblast proliferation via the PTPRB signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155715. [PMID: 38788399 DOI: 10.1016/j.phymed.2024.155715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/27/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an end-stage change in many interstitial lung diseases, whereas no proven effective anti-pulmonary fibrotic treatments. Forsythoside A (FA) derived from Forsythia suspensa (Thunb.) Vahl, has been found to possess lung-protective effect. However, studies on its anti-pulmonary fibrosis effect are limited and its mechanism of action remains unknown. PURPOSE This study aimed to explore the underlying mechanism of FA on PF. METHODS Male C57BL/6 mice were randomized into normal (CON), model (BLM), pirfenidone (PFD), low- and high-dose FA (FA-L, FA-H, respectively). Except for the CON group, which was injected with the same dose of saline, the model of PF was established by intratracheal instillation of BLM, during which the survival rate and body weight changes of the mice were measured. The lung histopathology was evaluated by Hematoxylin-eosin, Sirius red, and Masson staining. Transcriptome analysis was performed to screen for the differential genes associated with the role of FA in PF. Differential genes in normal and pulmonary fibrosis patients with the GSE2052 dataset were analyzed in the GEO database. The levels of CTGF, α-SMA, MMP-8 in lung and TNF-α in bronchoalveolar lavage fluid (BALF) were detected by ELISA. The levels of HYP in lungs were detected by digestion. The mRNA and protein levels of MMP-7, E-cadherin, CD31, α-SMA, TGF-β1, IL-6, β-catenin, ZO-1, PTPRB, E-cadherin, and vimentin in lungs were detected by RT-qPCR and Western blot. The expression of CD31, α-SMA, TGF-β1 and ZO-1 were detected by immunofluorescence. TGF-β1-stimulated HFL1 cells and human umbilical vein endothelial cells (HUVECs) were used in an attempt to explore the possible role of protein tyrosine phosphatase receptor type B (PTPRB) involved in FA-induced improvement of PF. RESULTS The results showed that FA could improve the survival rate and body weight of PF mice. FA could alleviate the symptoms of alveolar wall thickening, inflammatory cell infiltration, blue collagen fiber deposition, collagen fiber type Ⅰ and type Ⅲ in mice with PF. In addition, FA could reduce the levels of HYP, CTGF, α-SMA, TGF-β1, TNF-α, β-catenin and MMP8, and regulate the expression levels of CD31, ZO-1, PTPRB and E-cadherin in lung of mice with PF, inhibiting endothelial-to-mesenchymal transition (EndMT) and fibroblasts proliferation. In the GSE2052 dataset, the expression level of PTPRB is reduced in lung tissue from PF patients, and results from transcriptome sequencing indicate that PTPRB expression is also reduced in PF mice. In addition, the effect of FA on TGF-β1-induced HFL1 or HUVECs cells could be attenuated by the inhibitor of PTPRB, suggesting that the effect of FA on PF is related to PTPRB. CONCLUSION This study demonstrated that FA could ameliorate PF by inhibiting lung fibroblast proliferation and EndMT, and that PTPRB might be a target of FA to ameliorate PF, which provided evidence to support FA as a candidate phytochemical for PF.
Collapse
Affiliation(s)
- Qinqin Zhang
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China; Henan Key Laboratory of Chinese Medicine Resources and Chemistry, Zhengzhou 450046, Henan, China
| | - Beibei Zhang
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China; Henan Key Laboratory of Chinese Medicine Resources and Chemistry, Zhengzhou 450046, Henan, China; Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R., Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Fan Yang
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China; Henan Key Laboratory of Chinese Medicine Resources and Chemistry, Zhengzhou 450046, Henan, China
| | - Yingbo Hu
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Ruyi Fan
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Mengya Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Suiqing Chen
- Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China; Henan Key Laboratory of Chinese Medicine Resources and Chemistry, Zhengzhou 450046, Henan, China; Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R., Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China; Henan University of Chinese Medicine, Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, China.
| |
Collapse
|
6
|
Gao H, Yuan Z, Liang H, Liu Y. Integrating UPLC-Q-Orbitrap MS with serum pharmacochemistry network and experimental verification to explore the pharmacological mechanisms of Cynanchi stauntonii rhizoma et radix against sepsis-induced acute lung injury. Front Pharmacol 2024; 15:1261772. [PMID: 38584603 PMCID: PMC10995315 DOI: 10.3389/fphar.2024.1261772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/03/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction: Patients with sepsis are at an incremental risk of acute lung injury (ALI). Baiqian, also known as Cynanchi stauntonii rhizoma et radix (Csrer), has anti-inflammatory properties and is traditionally used to treat cough and phlegm. This study aimed to demonstrate the multicomponent, multitarget, and multi-pathway regulatory molecular mechanisms of Csrer in treating lipopolysaccharide (LPS)-induced ALI. Methods: The bioactive components of Csrer were identified by ultrahigh-performance liquid chromatography Q-Orbitrap mass spectrometry (UPLC-Q-Orbitrap MS). Active targets predicted from PharmMapper. DrugBank, OMIM, TTD, and GeneCards were used to identify potential targets related to ALI. Intersection genes were identified for Csrer against ALI. The PPI network was analysed to identify prime targets. GO and KEGG analyses were performed. A drug-compound-target-pathway-disease network was constructed. Molecular docking and simulations evaluated the binding free energy between key proteins and active compounds. The protective effect and mechanism of Csrer in ALI were verified using an ALI model in mice. Western blot, Immunohistochemistry and TUNEL staining evaluated the mechanisms of the pulmonary protective effects of Csrer. Results: Forty-six bioactive components, one hundred and ninety-two potential cross-targets against ALI and ten core genes were identified. According to GO and KEGG analyses, the PI3K-Akt, apoptosis and p53 pathways are predominantly involved in the "Csrer-ALI" network. According to molecular docking and dynamics simulations, ten key genes were firmly bound by the principal active components of Csrer. The "Csrer-ALI" network was revealed to be mediated by the p53-mediated apoptosis and inflammatory pathways in animal experiments. Conclusion: Csrer is a reliable source for ALI treatment based on its practical components, potential targets and pathways.
Collapse
Affiliation(s)
- Hejun Gao
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ziyi Yuan
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haoxuan Liang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|