1
|
Zhang Y, Yu Y, Xue J, Yu W, Zhou X, Jin M, Liu P, Wang T, Gao Z, Feng C. Lianhua qingke alleviates cigarette smoke induced cellular senescence in COPD mice by regulating the Sp1/SIRT1/HIF-1α pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119831. [PMID: 40250635 DOI: 10.1016/j.jep.2025.119831] [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: 01/23/2025] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lianhua Qingke (LHQK) has been utilized as a complementary therapy for respiratory diseases like tracheobronchitis and acute exacerbations of COPD in China. However, its therapeutic efficacy and underlying mechanisms for COPD remain elusive. AIM OF THE STUDY This study aimed to elucidate the mechanisms underlying the effects of LHQK on COPD, focusing on its anti-senescence properties. MATERIALS AND METHODS The therapeutic effects of LHQK were assessed by chronic cigarette smoke exposure induced COPD mice model. Lung function, histopathology investigation, cytokines detection and bio-molecular analysis were conducted to assess the impact of LHQK on pulmonary inflammation, mucin secretion, and cellular senescence of cigarette smoke (CS)-induced COPD mice. RESULTS A comprehensive analysis identified a total of 41 compounds as the key compounds of LHQK. Oral administration of LHQK markedly reversed the decline in pulmonary function, suppressed inflammation and mucus secretion, mitigated emphysema, and histopathology damage in lungs of COPD mice. In addition, LHQK attenuated secretory phenotype associated with cellular senescence in pulmonary and circulatory, and reduced the senescence-associated markers levels, such as SA-β-gal, miR-125a-5p, p21, p27 and p53. Network pharmacology and molecular assays indicated that LHQK enhanced Sp1 and SIRT1 expression, resulting to repression of HIF-1α, finally alleviating cellular senescence in COPD mice. CONCLUSIONS LHQK demonstrates potential as a complementary therapy for COPD, attenuating CS-triggered emphysema and pulmonary inflammation by targeting cellular senescence processes and modulation of Sp1/SIRT1/HIF-1α pathway.
Collapse
Affiliation(s)
- Yixin Zhang
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| | - Yan Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Jianbo Xue
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Xianqiang Zhou
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| | - Mengtong Jin
- Linfen Clinical Medicine Research Center, Linfen 041000, China; Linfen Central Hospital, Linfen 041000, China.
| | - Peng Liu
- Linfen Clinical Medicine Research Center, Linfen 041000, China; Linfen Central Hospital, Linfen 041000, China.
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China.
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China; Institute of Chest and Lung Diseases, Shanxi Medical University, Taiyuan 030001, China; Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing 100101, China; Shanxi Provincial Clinical Medical Research Center for Respiratory Diseases (COPD), Linfen 041000, China.
| | - Cuiling Feng
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
2
|
Li K, Gu X, Zhu Y, Guan N, Wang J, Wang L. Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomes Attenuates Experimental Periodontitis in Mice Partly by Delivering miRNAs. Int J Nanomedicine 2025; 20:2879-2899. [PMID: 40078652 PMCID: PMC11900796 DOI: 10.2147/ijn.s502192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Periodontitis is the most common non-communicable disease in humans. The main challenge in the treatment of periodontitis is to effectively control periodontal inflammation and promote tissue repair. Human umbilical cord mesenchymal stem cells-derived exosomes (hucMSCs-exo) have been reported to modulate inflammatory responses and promote tissue repairment mainly through miRNAs in several diseases. However, the effect of hucMSCs-exo on periodontitis remains unknown. In this study, we hypothesized that hucMSCs-exo could inhibit bone destruction in periodontitis mice. Methods In this study, we constructed and characterized the exo@H drug delivery platform. Lipopolysaccharide was used to construct an inflammatory microenvironment in vitro to detect MC3T3-E1 cells proliferation and bone regeneration capacity. Ligation induced to construct an experimental periodontitis mouse model. The distance of the cement-enamel junction (CEJ) to the alveolar bone crest (ABC) was measured for bone resorption evaluation. Hematoxylin-eosin (H&E) staining and Tartrate resistant acid phosphatase (TRAP) staining were used to observe periodontal tissue changes. MicroRNA (miRNA) sequencing was used to detect differential genes and for bioinformatics analysis. Real-time quantitative polymerase chain reaction (qRT-PCR). WB assay and dual luciferase assay were used to further validate the screened differentially expressed miRNAs and the targeted binding relationship with the corresponding target genes. Results We found that lyophilized hucMSCs-exo promoted the proliferation and osteogenic differentiation of MC3T3-E1 cells, and showed more significant proliferative and osteogenic differentiation abilities in combination with the hydrogel (P < 0.05). Using periodontitis mice, bone resorption evaluation revealed a significant reduction in alveolar bone resorption in the exo@H group compared to the hydrogel group (P < 0.01), and exo@H was able to reduce the inflammatory response of periodontal tissues and the number of osteoclasts on the surface of the alveolar bone compared to the hydrogel group. Moreover, 59 miRNAs were upregulated, such as let-7f-5p and miR-203-3p, which positively targeted IL-13 and Nit2, respectively. Discussion These results suggest that exo@H provides protection against periodontitis partly by delivering miRNAs to periodontal tissue. Our results confirm the feasibility of the exo@H delivery platform we constructed and the effectiveness of its use for periodontitis treatment, and this study provides a promising approach for the treatment of periodontitis via miRNA.
Collapse
Affiliation(s)
- Ke Li
- Department of Periodontics and Mucosa, The second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Xiaoli Gu
- Department of Periodontics and Mucosa, The second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Yanan Zhu
- Department of Periodontics and Mucosa, The second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Ning Guan
- Key Laboratory of Brain and Spinal Cord Injury Research, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Jinlei Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| | - Linyuan Wang
- Department of Periodontics and Mucosa, The second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People’s Republic of China
| |
Collapse
|
3
|
Wei Y, Ni W, Zhao L, Gao Y, Zhou B, Feng Q, Ma Y, Wang L. Phillygenin Inhibits PI3K-Akt-mTOR Signalling Pathway to Prevent bleomycin-Induced Idiopathic Pulmonary Fibrosis in Mice. Clin Exp Pharmacol Physiol 2025; 52:e70017. [PMID: 39746665 DOI: 10.1111/1440-1681.70017] [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: 08/15/2024] [Revised: 11/15/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease characterised by irreversible lung structure and function. Phillygenin (PHI) is a lignan extracted from Forsythiae fructus with the activities of anti-inflammatory and antioxidant. This study aimed to explore the protective effect of PHI on IPF. The mouse model of IPF was established by bleomycin (BLM), and then treated with PHI. After 15 days of administration, the lung index was calculated. H&E staining, Masson staining and immunohistochemical methods were used to detect the effect of PHI on pulmonary fibrosis. MDA and SOD were tested to evaluate the effect of PHI on lung tissue oxidative stress. Western blot was used to detect the effect of PHI on the expressions of α-SMA, p-smad2, TGF- β1, Nrf2, HO-1 and NQO-1. Network pharmacology was used to identify the key signalling pathways for PHI to improve IPF, and Western blot was used to validate the result. The results showed that PHI prevented mice from BLM-induced IPF, manifested by reducing lung index, improving lung tissue pathological damage, inhibiting collagen deposition and expression of fibrosis markers including α-SMA, collagen1, p-smad2 and TGF-β1. PHI inhibited oxidative stress by upregulating the expressions of Nrf2, HO-1 and NQO-1. Network pharmacology revealed that PI3K-Akt-mTOR signalling pathway was the underlying target of PHI for IPF. Molecular docking indicated strong binding of PHI with PIK3CA, AKT1 and RELA. Western blot validated that PHI downregulated the PI3K-Akt-mTOR signalling pathway and stimulated autophagy. This study indicated that PHI prevented BLM-induced pulmonary fibrosis by inhibiting PI3K-Akt-mTOR signalling pathway.
Collapse
Affiliation(s)
- Yongjia Wei
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Wenting Ni
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lizhi Zhao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Yanhong Gao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Bing Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Qun Feng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Yun Ma
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Limin Wang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
4
|
Han X, Zhang Y, Zhang F, Li X, Meng Y, Huo J, Chen M, Liu F, Wang W, Wang N. Network pharmacology and phytochemical composition combined with validation in vivo and in vitro reveal the mechanism of platycodonis radix ameliorating PM2.5-induced acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118829. [PMID: 39278295 DOI: 10.1016/j.jep.2024.118829] [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: 06/02/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Platycodonis radix (PR), the root of Platycodon grandiflorus (Jacq.) A. DC., is a traditional Chinese medicine recognized for its dual role as both a medicinal and dietary substance, exhibiting significant anti-inflammatory properties. It is frequently utilized in the treatment of lung diseases. However, the molecular mechanisms by which PR exerts its effects in the treatment of acute lung injury (ALI) remain unclear. AIM OF THE STUDY This study presents a novel strategy that integrates network pharmacology, molecular docking, untargeted metabolomics analysis and experimental validation to investigate the molecular mechanisms through which PR treats ALI. MATERIALS AND METHOD Initially, the bioactive components of PR, along with its targets and pathways in the treatment of ALI, were identified using network pharmacology. Following this, preliminary validation was conducted through molecular docking. The active ingredients in the aqueous extract of PR were characterized using HPLC-MS. Finally, in vivo and in vitro experiments were performed to further validate the findings from the network pharmacology. RESULTS A total of 14 bioactive components and 156 effective targets were identified using the TCMSP, DisGeNET, Genecard, OMIM databases and Venny 2.1.0. Protein-protein interaction (PPI) analysis revealed 22 core targets including TP53, AKT1, STAT3 and JUN. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses indicated that these targets primarily participate in the regulation of cellular apoptosis, lung cancer and inflammatory pathways. Molecular docking demonstrated that four bioactive components exhibited strong affinities with their respective docking targets. LC-MS analysis confirmed that the aqueous extract of PR contained 87 components, including two active ingredients identified through network pharmacology and molecular docking. Preliminary validation was conducted in mice with ALI induced by acute PM2.5 exposure, revealing that the aqueous extract of PR reduced inflammatory factor levels in bronchoalveolar lavage fluid, enhanced antioxidant capacity in lung tissue, and decreased lung cell apoptosis in PM2.5-exposed mice. Notably, PR alleviated PM2.5-induced ALI through the STAT3, JUN, and AKT1 signaling pathways. Similarly, the results of in vitro intervention experiments further confirmed that the aqueous extract of PR protected pulmonary epithelial cells against PM2.5 exposure through activating AKT1 sinalling pathway, and inhibiting STAT3 and JUN signalling pathways. CONCLUSION This study identifies the active components of PR and elucidates the molecular mechanisms by which PR alleviates ALI, specifically by inhibiting the phosphorylation levels of STAT3 and c-JUN, or by activating the phosphorylation level of AKT1. These results provide a foundational basis for the application of PR in the treatment or prevention of lung injuries induced by particulate matter.
Collapse
Affiliation(s)
- Xianlei Han
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yue Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Fan Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xiumei Li
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yanli Meng
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, 150036, China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, 150036, China
| | - Mian Chen
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Postdoctoral Scientific Research Workstation, Jinan, 2501011, China
| | - Fei Liu
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Postdoctoral Scientific Research Workstation, Jinan, 2501011, China
| | - Weiming Wang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, 150036, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
5
|
Sadu Murari LS, Kunkel S, Shetty A, Bents A, Bhandary A, Rivera-Mulia JC. p63: A Master Regulator at the Crossroads Between Development, Senescence, Aging, and Cancer. Cells 2025; 14:43. [PMID: 39791744 PMCID: PMC11719615 DOI: 10.3390/cells14010043] [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: 10/31/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
The p63 protein is a master regulatory transcription factor that plays crucial roles in cell differentiation, adult tissue homeostasis, and chromatin remodeling, and its dysregulation is associated with genetic disorders, physiological and premature aging, and cancer. The effects of p63 are carried out by two main isoforms that regulate cell proliferation and senescence. p63 also controls the epigenome by regulating interactions with histone modulators, such as the histone acetyltransferase p300, deacetylase HDAC1/2, and DNA methyltransferases. miRNA-p63 interactions are also critical regulators in the context of cancer metastasis. This review aims to elaborate on the diverse roles of p63, focusing on disease, development, and the mechanisms controlling genome organization and function.
Collapse
Affiliation(s)
- Lakshana Sruthi Sadu Murari
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sam Kunkel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
| | - Anala Shetty
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Addison Bents
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
| | - Aayush Bhandary
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
| | - Juan Carlos Rivera-Mulia
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (L.S.S.M.); (S.K.); (A.B.); (A.B.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Hou Z, Yang F, Zhang Q, Wang Y, Liu J, Liang F. Targeting the PI3K/AKT signaling pathway with PNU120596 protects against LPS-induced acute lung injury. J Pharm Pharmacol 2024; 76:1508-1520. [PMID: 39288376 DOI: 10.1093/jpp/rgae076] [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: 11/13/2023] [Accepted: 06/04/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES This study investigated the potential therapeutic benefits of PNU120596, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (α7nAChR), in mitigating acute lung injury (ALI) induced by lipopolysaccharide (LPS) in a mouse model. Specifically, we sought to examine the impact of PNU120596 on the PI3K/AKT signaling pathway in the context of ALI. METHODS ALI was induced in mice by LPS administration, and the protective effects of PNU120596 were assessed. Lung injury, lung function, and the inflammatory response were evaluated. Additionally, the activation of the PI3K/AKT signaling pathway was examined, along with the levels of inflammatory factors and oxidative stress markers. KEY FINDINGS PNU120596 significantly ameliorated LPS-induced lung injury, improved lung function, and reduced the inflammatory response in the mouse model of ALI. Furthermore, we observed that PNU120596 inhibited the activation of the PI3K/AKT signaling pathway, which was associated with decreased levels of inflammatory factors and oxidative stress markers. CONCLUSIONS PNU120596 exhibits promising therapeutic potential for the treatment of acute lung injury, potentially by targeting the PI3K/AKT signaling pathway. These findings suggest that modulation of the α7 nicotinic acetylcholine receptor with PNU120596 may offer a viable strategy for the management of ALI, warranting further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Zixin Hou
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
- Department of Anesthesiology, General Hospital, Hunan University of Medicine, Huaihua 418000, PR China
| | - Qiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Yuxia Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Junwen Liu
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| |
Collapse
|
7
|
Fan W, Gui B, Zhou X, Li L, Chen H. A narrative review on lung injury: mechanisms, biomarkers, and monitoring. Crit Care 2024; 28:352. [PMID: 39482752 PMCID: PMC11526606 DOI: 10.1186/s13054-024-05149-x] [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: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Lung injury is closely associated with the heterogeneity, severity, mortality, and prognosis of various respiratory diseases. Effective monitoring of lung injury is crucial for the optimal management and improved outcomes of patients with lung diseases. This review describes acute and chronic respiratory diseases characterized by significant lung injury and current clinical tools for assessing lung health. Furthermore, we summarized the mechanisms of lung cell death observed in these diseases and highlighted recently identified biomarkers in the plasma indicative of injury to specific cell types and scaffold structure in the lung. Last, we propose an artificial intelligence-driven lung injury monitoring model to assess disease severity, and predict mortality and prognosis, aiming to achieve precision and personalized medicine.
Collapse
Affiliation(s)
- Wenping Fan
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China
| | - Biyu Gui
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
| | - Xiaolei Zhou
- Department of Pulmonary Medicine, Chest Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Li Li
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| |
Collapse
|
8
|
Ma Y, Hou Y, Han Y, Liu Y, Han N, Yin Y, Wang X, Jin P, He Z, Sun J, Hao Y, Guo J, Wang T, Feng W, Qi H, Jia Z. Ameliorating lipopolysaccharide induced acute lung injury with Lianhua Qingke: focus on pulmonary endothelial barrier protection. J Thorac Dis 2024; 16:6899-6917. [PMID: 39552861 PMCID: PMC11565356 DOI: 10.21037/jtd-24-700] [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: 04/29/2024] [Accepted: 08/16/2024] [Indexed: 11/19/2024]
Abstract
Background Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) has long posed challenges in clinical practice, lacking established preventive and therapeutic approaches. Lianhua Qingke (LHQK), a patented traditional Chinese medicine (TCM), has been found to have anti-inflammatory effects for ameliorating ALI/ARDS induced by lipopolysaccharide (LPS). This study aimed to investigate the effects and potential mechanisms of LHQK on endothelial protection in LPS-induced ALI/ARDS in vivo and in LPS-induced human pulmonary microvascular endothelial cells (HPMECs) injury in vitro. Methods In the animal experiment, we induced an ALI/ARDS model by intratracheal injection of LPS (5 mg/mL). LHQK (3.7 g/kg/d for low dose and 7.4 g/kg/d for high dose) or dexamethasone (DEX) (5 mg/kg/d) was administered to mice 3 days prior to LPS treatment. In the in vitro experiments, HPMECs were pretreated with LHQK at concentrations of 125 and 250 µg/mL for 2 hours before being stimulated with LPS (10 µg/mL). We employed lung function test, measurement of lung index, hematoxylin and eosin (H&E) staining, bronchoalveolar lavage fluid (BALF) cell counts, and inflammatory cytokine levels to assess the therapeutic effect of LHQK. Additionally, the extravasation assay of fluorescein isothiocyanate-dextran (FITC-dextran) dye and the transmembrane electrical resistance (TEER) assay were used to evaluate endothelial barrier. Barrier integrity and relevant protein validation were assessed using immunofluorescence (IF) and Western blot analyses. Furthermore, network pharmacology analysis and cellular level screening were employed to predict and screen the active ingredients of LHQK. Results Compared to the LPS group, LHQK significantly improved lung function, mitigated lung pathological injuries, reduced inflammatory cells and inflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6] levels in BALF, and inhibited the expression of vascular cell adhesion molecule-1 (VCAM-1), attenuated LPS-induced pulmonary oedema and FITC-dextran permeability, and enhanced the expression of vascular endothelial-cadherin (VE-cadherin) and occludin. In vitro, LHQK attenuated LPS-induced HPMECs injury by elevating TEER values and enhancing VE-cadherin and occludin protein levels. Finally, network pharmacology analysis and cellular level validation identified potential active ingredients of LHQK. Conclusions In summary, LHQK can mitigate LPS-induced inflammatory infiltration, pulmonary edema, and pulmonary vascular endothelial barrier dysfunction in the context of ALI/ARDS. This is achieved by decreasing the levels of VCAM-1, and increasing the expression levels of barrier-associated junctions, such as VE-cadherin and occludin. Consequently, LHQK exhibits promising therapeutic potential in preventing the progression of ALI/ARDS.
Collapse
Affiliation(s)
- Yan Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yunlong Hou
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Yu Han
- Department of Pharmacy, Hebei Children’s Hospital, Shijiazhuang, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Yujie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Xiaoqi Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Peipei Jin
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhuo He
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jiemeng Sun
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuanjie Hao
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Jing Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Wei Feng
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Zhenhua Jia
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
- Department of Respiratory, Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
Liu C, He Y, Zhou K, Wang H, Zhou M, Sun J, Lu Y, Huang Y, Wang Y, Liu T, Li Y. Mitigation of allergic asthma in mice: A compound mixture comprising luteolin, arbutin, and marmesin from Gerbera Piloselloides Herba by suppression of PI3K/Akt pathway. Heliyon 2024; 10:e37632. [PMID: 39381113 PMCID: PMC11456855 DOI: 10.1016/j.heliyon.2024.e37632] [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: 05/09/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Background Gerberae Piloselloidis Herba (GPH) exhibits notable efficacy in alleviating allergic asthma. Previous studies in our research have identified a mixture of luteolin, arbutin, and marmesin as effective components of GPH in treating allergic asthma. However, the underlying mechanism remains unclear. This study aims to elucidate the molecular mechanism of these active components. Method Using an ovalbumin (OVA)-induced allergic asthma mouse model, various treatment groups were administered, including GPH, the active component mixture (termed "Mixture") containing luteolin, arbutin, and marmesin, and a positive drug (dexamethasone, DEX). Relevant indices were assessed, including behavioral characteristics, inflammatory cell counts, cytokine levels, histopathological examination of lung tissue, apoptosis, and expression of key proteins such as Caspase-3, Bax, Bcl-2, PI3K, p-PI3K, Akt, and p-Akt. The effect of the Mixture on the PI3K/Akt signaling pathway was further verified using the PI3K inhibitor LY294002. Results The Mixture significantly alleviated asthma symptoms, decreased IgE levels, cytokine levels (IL-4, IL-5, IL-13 and TNF-α), and the number of inflammatory cells in serum or bronchoalveolar lavage fluid (BALF), leading to the alleviation of lung pathological lesions. Additionally, the Mixture reduced the expression of Bax and Caspase-3 while increasing Bcl-2 expression, resulting in mitigated apoptosis in lung tissue. Furthermore, there appeared a decrease in the levels of PI3K and p-PI3K, as well as the ratio of p-Akt to Akt in the Mixture group, indicating the suppression of PI3K and Akt phosphorylation. Interestingly, the effects of the Mixture were comparable to those of GPH, LY294002, or the combination of LY294002 with the Mixture. Conclusion The study confirms that the Mixture containing luteolin, arbutin, and marmesin indeed alleviates allergic asthma induced by OVA in mice by suppressing the PI3K/Akt signaling pathway. These findings highlight the potential of the GPH-derived Mixture as a novel therapeutic for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Chunhua Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Yu He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China
| | - Kun Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China
| | - Hong Wang
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Meng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Jia Sun
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Yuan Lu
- Computer Education and Information Technology Center, Guizhou Medical University,Guiyang, 561113, China
| | - Yong Huang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Yonglin Wang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Ting Liu
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Yongjun Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
10
|
Su Y, Lucas R, Fulton DJ, Verin AD. Mechanisms of pulmonary endothelial barrier dysfunction in acute lung injury and acute respiratory distress syndrome. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:80-87. [PMID: 39006829 PMCID: PMC11242916 DOI: 10.1016/j.pccm.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 07/16/2024]
Abstract
Endothelial cells (ECs) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. Pulmonary endothelial barrier integrity is maintained through coordinated cellular processes involving receptors, signaling molecules, junctional complexes, and protein-regulated cytoskeletal reorganization. In acute lung injury (ALI) or its more severe form acute respiratory distress syndrome (ARDS), the loss of endothelial barrier integrity secondary to endothelial dysfunction caused by severe pulmonary inflammation and/or infection leads to pulmonary edema and hypoxemia. Pro-inflammatory agonists such as histamine, thrombin, bradykinin, interleukin 1β, tumor necrosis factor α, vascular endothelial growth factor, angiopoietin-2, and platelet-activating factor, as well as bacterial toxins and reactive oxygen species, cause dynamic changes in cytoskeletal structure, adherens junction disorganization, and detachment of vascular endothelial cadherin (VE-cadherin) from the actin cytoskeleton, leading to an increase in endothelial permeability. Endothelial interactions with leukocytes, platelets, and coagulation enhance the inflammatory response. Moreover, inflammatory infiltration and the associated generation of pro-inflammatory cytokines during infection cause EC death, resulting in further compromise of the structural integrity of lung endothelial barrier. Despite the use of potent antibiotics and aggressive intensive care support, the mortality of ALI is still high, because the mechanisms of pulmonary EC barrier disruption are not fully understood. In this review, we summarized recent advances in the studies of endothelial cytoskeletal reorganization, inter-endothelial junctions, endothelial inflammation, EC death, and endothelial repair in ALI and ARDS, intending to shed some light on the potential diagnostic and therapeutic targets in the clinical management of the disease.
Collapse
Affiliation(s)
- Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J.R. Fulton
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alexander D. Verin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
11
|
Rogulska O, Vackova I, Prazak S, Turnovcova K, Kubinova S, Bacakova L, Jendelova P, Petrenko Y. Storage conditions affect the composition of the lyophilized secretome of multipotent mesenchymal stromal cells. Sci Rep 2024; 14:10243. [PMID: 38702388 PMCID: PMC11068735 DOI: 10.1038/s41598-024-60787-z] [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: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/06/2024] Open
Abstract
The widespread use of multipotent mesenchymal stromal cell-derived secretome (MSC-sec) requires optimal preservation methods. Lyophilization offers benefits like concentrating the secretome, reducing the storage volume, and making storage conditions more flexible. This study evaluated the influence of storage duration and temperature on lyophilized MSC-sec. The conditioned medium from Wharton's jelly MSCs was stored at - 80 °C or lyophilized with or without trehalose. Lyophilized formulations were kept at - 80 °C, - 20 °C, 4 °C, or room temperature (RT) for 3 and 30 months. After storage and reconstitution, the levels of growth factors and cytokines were assessed using multiplex assay. The storage of lyophilized MSC-sec at - 80 °C ensured biomolecule preservation for 3 and 30 months. Following 3 month storage at 4 °C and RT, a notable decrease occurred in BDNF, bNGF, and sVCAM-1 levels. Prolonged 30 month storage at the same temperatures significantly reduced BDNF, bNGF, VEGF-A, IL-6, and sVCAM-1, while storage at - 20 °C decreased BDNF, bNGF, and VEGF- A levels. Trehalose supplementation of MSC-sec improved the outcome during storage at 4 °C and RT. Proper storage conditions were crucial for the preservation of lyophilized MSC-sec composition. Short-term storage at various temperatures maintained over 60% of the studied growth factors and cytokines; long-term preservation was only adequate at -80 °C.
Collapse
Affiliation(s)
- Olena Rogulska
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
| | - Irena Vackova
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic.
| | - Simon Prazak
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
| | - Karolina Turnovcova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
| | - Sarka Kubinova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Na Slovance 1999/2, 18221, Prague, Czech Republic
| | - Lucie Bacakova
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
| | - Pavla Jendelova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic
| | - Yuriy Petrenko
- Laboratory of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic.
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14200, Prague, Czech Republic.
| |
Collapse
|
12
|
Han L, Zhu W, Qi H, He L, Wang Q, Shen J, Song Y, Shen Y, Zhu Q, Zhou J. The cuproptosis-related gene glutaminase promotes alveolar macrophage copper ion accumulation in chronic obstructive pulmonary disease. Int Immunopharmacol 2024; 129:111585. [PMID: 38325045 DOI: 10.1016/j.intimp.2024.111585] [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: 08/24/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
Cuproptosis, a novel mode of cell death, is strongly associated with a variety of diseases. However, the contribution of cuproptosis to the onset or progression of chronic obstructive pulmonary disease (COPD), the third most common chronic cause of mortality, is not yet clear. To investigate the potential role of cuproptosis in COPD, raw datasets from multiple public clinical COPD databases (including RNA-seq, phenotype, and lung function data) were used. For further validation, mice exposed to cigarette smoke for three months were used as in vivo models, and iBMDMs (immortalized bone marrow-derived macrophages) and RAW264.7 cells stimulated with cigarette smoke extract were used as in vitro models. For the first time, the expression of the cuproptosis-related gene glutaminase (GLS) was found to be decreased in COPD, and the low expression of GLS was significantly associated with the grade of pulmonary function. In vivo experiments confirmed the decreased expression of GLS in COPD, particularly in alveolar macrophages. Furthermore, in vitro studies revealed that copper ions accumulated in alveolar macrophages, leading to a substantially decreased amount of cell activity of macrophages when stimulated with cigarette extract. In summary, we demonstrate the high potential of GLS as an avenue for diagnosis and therapy in COPD.
Collapse
Affiliation(s)
- Linxiao Han
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China
| | - Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050091, Hebei, China
| | - Ludan He
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China
| | - Jie Shen
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Fudan University, Shanghai 200540, China; Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Fudan University, Shanghai 200540, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 200540, China
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China
| | - Yao Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Shanghai 201399, China.
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai 200032, China; Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050091, Hebei, China; Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Fudan University, Shanghai 200540, China; Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Fudan University, Shanghai 200540, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 200540, China.
| |
Collapse
|
13
|
Sun X, Xiang H, Liu Z, Xiao H, Li X, Gong W, Pan L, Zhao L, Yao J, Sun C, Zhang G. Jingfang Granules () alleviates bleomycin-induced acute lung injury through regulating PI3K/Akt/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116946. [PMID: 37482261 DOI: 10.1016/j.jep.2023.116946] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury is a kind of clinical emergency severe syndrome which might trigger acute respiratory distress syndrome. Jingfang Granules () is a traditional Chinese medicine which has been proven to improve acute lung injury induced by bleomycin through inhibiting recruitment and overactive of inflammation. However, the potential mechanisms are still not well evaluated. AIM OF STUDY The aim of this study was to evaluate the protective function of Jingfang Granules on bleomycin caused acute lung injury and further discuss the potential pharmacological mechanisms. MATERIALS AND METHODS C57BL/6J mice were intratracheal injected bleomycin to induce model with acute lung injury. The protective impact of Jingfang Granules on acute lung injury and lung fibrosis triggered by bleomycin were evaluated through detecting mice body weight, lung appearance, lung index, and histopathology. The potential pharmacological mechanism of Jingfang Granules in treating acute lung injury was further elucidated by the methods of network pharmacology, proteomics, metabolomics, as well as western blot. Additionally, the network pharmacology analysis and molecular docking technology were integrated to investigate the targets of Jingfang Granules improving acute lung injury. RESULTS Our results indicated that Jingfang Granules effectively protected mice from acute lung injury induced by bleomycin, which was confirmed by higher body weight, lower pulmonary edema and lung index, and improved pathology and fibrosis of lung tissue compared to model group. Proteomics, western blot, and metabolomics were integrated and the results confirmed that Jingfang Granules regulated the Glycolysis/Gluconogenesis and Pyruvate metabolism through downregulating the PI3K/Akt/mTOR signaling pathway. The network pharmacology analysis and molecular docking technology results showed that the targets of Jingfang Granules for treating acute lung injury were enriched in the PI3K/Akt signaling pathway, which included 7 target proteins such as MAPK1, MAPK3, JAK2, HRAS, EGFR, PIK3R1, and PIK3CA. CONCLUSION This study indicates that Jingfang Granules displays a markedly protective effect on acute lung injury caused by bleomycin through downregulating PI3K/Akt/mTOR signaling pathway, which in turn regulates Glycolysis/Gluconogenesis and Pyruvate metabolism.
Collapse
Affiliation(s)
- Xingxu Sun
- School of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, 510006, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Haixin Xiang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
| | - Zhong Liu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - He Xiao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Xin Li
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Wenqiao Gong
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Lizhi Zhao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guimin Zhang
- School of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, 510006, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| |
Collapse
|
14
|
Peng W, Song Y, Zhu G, Zeng Y, Cai H, Lu C, Abuduxukuer Z, Song X, Gao X, Ye L, Wang J, Jin M. FGF10 attenuates allergic airway inflammation in asthma by inhibiting PI3K/AKT/NF-κB pathway. Cell Signal 2024; 113:110964. [PMID: 37956773 DOI: 10.1016/j.cellsig.2023.110964] [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: 07/27/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND The effect of fibroblast growth factor 10 (Fgf10) against allergic asthma has remained unclear, despite its importance in lung development and homeostasis maintenance. The purpose of this study was to investigate the protective effect and potential mechanism of Fgf10 on asthma. METHOD House Dust Mite (HDM)-induced asthma mice were administered recombinant Fgf10 intranasally during activation. Flow cytometry and ELISA were performed to determine type of inflammatory cells and type 2 cytokines levels in bronchoalveolar lavage fluid (BALF). Hematoxylin and eosin (H&E) and periodic acid - Schiff (PAS) staining of lung sections were conducted to evaluate histopathological assessment. Transcriptome profiling was analyzed using RNA-seq, followed by bioinformatics and network analyses to investigate the potential mechanisms of Fgf10 in asthma. RT-qPCR was also used to search for and validate differentially expressed genes in human Peripheral Blood Mononuclear Cells (PBMCs). RESULTS Exogenous administration of Fgf10 alleviated HDM-induced inflammation and mucus secretion in lung tissues of mice. Fgf10 also significantly inhibited the accumulation of eosinophils and type 2 cytokines (IL-4, IL-5, and IL-13) in BALF. The PI3K/AKT/NF-κB pathway may mediate the suppressive impact of Fgf10 on the asthma inflammation. Through RNA-seq analysis, the intersection of 71 differentially expressed genes (DEGs) was found between HDM challenge and Fgf10 treatment. GO and KEGG enrichment analyses indicated a strong correlation between the DEGs and different immune response. Immune infiltration analysis predicted the differential infiltration of five types of immune cells, such as NK cells, dendritic cells, monocytes and M1 macrophages. PPI analysis determined hub genes such as Irf7, Rsad2, Isg15 and Rtp4. Interestingly, above genes were consistently altered in human PBMCs in asthmatic patients. CONCLUSION Asthma airway inflammation could be attenuated by Fgf10 in this study, suggesting that it could be a potential therapeutic target.
Collapse
Affiliation(s)
- Wenjun Peng
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yansha Song
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guiping Zhu
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingying Zeng
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Cai
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chong Lu
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zilinuer Abuduxukuer
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xixi Song
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ling Ye
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Meiling Jin
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
15
|
Hao Y, Wang T, Hou Y, Wang X, Yin Y, Liu Y, Han N, Ma Y, Li Z, Wei Y, Feng W, Jia Z, Qi H. Therapeutic potential of Lianhua Qingke in airway mucus hypersecretion of acute exacerbation of chronic obstructive pulmonary disease. Chin Med 2023; 18:145. [PMID: 37924136 PMCID: PMC10623880 DOI: 10.1186/s13020-023-00851-4] [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: 06/25/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Lianhua Qingke (LHQK) is an effective traditional Chinese medicine used for treating acute tracheobronchitis. In this study, we evaluated the effectiveness of LHQK in managing airway mucus hypersecretion in the acute exacerbation of chronic obstructive pulmonary disease (AECOPD). METHODS The AECOPD model was established by subjecting male Wistar rats to 12 weeks of cigarette smoke (CS) exposure (80 cigarettes/day, 5 days/week for 12 weeks) and intratracheal lipopolysaccharide (LPS) exposure (200 μg, on days 1, 14, and 84). The rats were divided into six groups: control (room air exposure), model (CS + LPS exposure), LHQK (LHQK-L, LHQK-M, and LHQK-H), and a positive control group (Ambroxol). H&E staining, and AB-PAS staining were used to evaluate lung tissue pathology, inflammatory responses, and goblet cell hyperplasia. RT-qPCR, immunohistochemistry, immunofluorescence and ELISA were utilized to analyze the transcription, expression and secretion of proteins related to mucus production in vivo and in the human airway epithelial cell line NCI-H292 in vitro. To predict and screen the active ingredients of LHQK, network pharmacology analysis and NF-κB reporter system analysis were employed. RESULTS LHQK treatment could ameliorate AECOPD-triggered pulmonary structure damage, inflammatory cell infiltration, and pro-inflammatory cytokine production. AB-PAS and immunofluorescence staining with CCSP and Muc5ac antibodies showed that LHQK reduced goblet cell hyperplasia, probably by inhibiting the transdifferentiation of Club cells into goblet cells. RT-qPCR and immunohistochemistry of Muc5ac and APQ5 showed that LHQK modulated mucus homeostasis by suppressing Muc5ac transcription and hypersecretion in vivo and in vitro, and maintaining the balance between Muc5ac and AQP5 expression. Network pharmacology analysis and NF-κB luciferase reporter system analysis provided insights into the active ingredients of LHQK that may help control airway mucus hypersecretion and regulate inflammation. CONCLUSION LHQK demonstrated therapeutic effects in AECOPD by reducing inflammation, suppressing goblet cell hyperplasia, preventing Club cell transdifferentiation, reducing Muc5ac hypersecretion, and modulating airway mucus homeostasis. These findings support the clinical use of LHQK as a potential treatment for AECOPD.
Collapse
Affiliation(s)
- Yuanjie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Yunlong Hou
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Xiaoqi Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Yujie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yan Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yaru Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Wei Feng
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Zhenhua Jia
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
- Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, 050091, Hebei, China.
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| |
Collapse
|
16
|
Li Y, Giovannini S, Wang T, Fang J, Li P, Shao C, Wang Y, Shi Y, Candi E, Melino G, Bernassola F. p63: a crucial player in epithelial stemness regulation. Oncogene 2023; 42:3371-3384. [PMID: 37848625 PMCID: PMC10638092 DOI: 10.1038/s41388-023-02859-4] [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: 07/31/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Epithelial tissue homeostasis is closely associated with the self-renewal and differentiation behaviors of epithelial stem cells (ESCs). p63, a well-known marker of ESCs, is an indispensable factor for their biological activities during epithelial development. The diversity of p63 isoforms expressed in distinct tissues allows this transcription factor to have a wide array of effects. p63 coordinates the transcription of genes involved in cell survival, stem cell self-renewal, migration, differentiation, and epithelial-to-mesenchymal transition. Through the regulation of these biological processes, p63 contributes to, not only normal epithelial development, but also epithelium-derived cancer pathogenesis. In this review, we provide an overview of the role of p63 in epithelial stemness regulation, including self-renewal, differentiation, proliferation, and senescence. We describe the differential expression of TAp63 and ΔNp63 isoforms and their distinct functional activities in normal epithelial tissues and in epithelium-derived tumors. Furthermore, we summarize the signaling cascades modulating the TAp63 and ΔNp63 isoforms as well as their downstream pathways in stemness regulation.
Collapse
Affiliation(s)
- Yanan Li
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Sara Giovannini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Tingting Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Peishan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Ying Wang
- Shanghai Institute of Nutrition and Health, Shanghai, 200031, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China.
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
17
|
Tan J, Zhuo Z, Wang X, Zhang Y, Qian Y, Lin F. Secretome of EMSCs neutralizes LPS‑induced acute lung injury via aerosol administration. Int J Mol Med 2023; 52:104. [PMID: 37772372 PMCID: PMC10558219 DOI: 10.3892/ijmm.2023.5307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Ectodermal mesenchymal stem cells (EMSCs) are cells harvested from the stem cell niche (nasal mucosa) with high therapeutic potential. To the best of our knowledge, however, the anti‑inflammatory properties of these neural crest‑derived EMSCs have been rarely reported. The present study aimed to explore the effects of aerosolized EMSC‑Secretome (EMSC‑Sec) and clarify underlying mechanisms in treating acute lung injury (ALI). EMSCs were isolated by adherent method and identified by immunofluorescence staining. EMSC‑Sec was collected and evaluated using western blotting, BCA and ELISA tests. Then, mouse lung epithelial cells (MLE‑12) were used to mimic inflammatory stimulation with lipopolysaccharide (LPS). After developing an ALI model through intraperitoneal injection of LPS, mice were treated with an EMSC‑Sec spray. The lung in each group underwent an observation and measurement to preliminarily assess the extent of damage. H&E staining, immunohistochemical staining, immunofluorescence and western‑blotting were utilized to further access the impacts of EMSC‑Sec. The results showed that EMSC‑Sec had great anti‑inflammatory potential and was highly successful in vitro and in vivo. EMSC‑Sec mitigated LPS‑induced ALI with low inflammatory cell inflation and mild damage. EMSC‑Sec could regulate inflammation via the NF‑κB(p50/p65)/NLRP3 pathway. Overall, the present study demonstrated that EMSC‑Sec regulated inflammation, hoping to provide a novel strategy for ALI treatment.
Collapse
Affiliation(s)
- Jianing Tan
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Ziliang Zhuo
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Xiuyu Wang
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Yanshuang Zhang
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Yucheng Qian
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Fangfang Lin
- Department of Oncology, The First People's Hospital of Zhenjiang, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| |
Collapse
|
18
|
Tang S, Liang Y, Wang M, Lei J, Peng Y, Tao Q, Ming T, Yang W, Zhang C, Guo J, Xu H. Qinhuo Shanggan oral solution resolves acute lung injury by down-regulating TLR4/NF- κB signaling cascade and inhibiting NLRP3 inflammasome activation. Front Immunol 2023; 14:1285550. [PMID: 37954597 PMCID: PMC10634205 DOI: 10.3389/fimmu.2023.1285550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Acute lung injury (ALI) is a common condition, particularly in the COVID-19 pandemic, which is distinguished by sudden onset of respiratory insufficiency with tachypnea, oxygen-refractory cyanosis, reduced lung compliance and diffuse infiltration of pulmonary alveoli. It is well-established that increasing activity of toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling axis and the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation are associated with the pathogenesis of ALI. Since ALI poses a huge challenge to human health, it is urgent to tackle this affliction with therapeutic intervention. Qinhuo Shanggan oral solution (QHSG), a traditional Chinese herbal formula, is clinically used for effective medication of various lung diseases including ALI, with the action mechanism obscure. In the present study, with the rat model of lipopolysaccharide (LPS)-induced ALI, QHSG was unveiled to ameliorate ALI by alleviating the pathological features, reversing the alteration in white blood cell profile and impeding the production of inflammatory cytokines through down-regulation of TLR4/NF-κB signaling cascade and inhibition of NLRP3 inflammasome activation. In LPS-stimulated RAW264.7 mouse macrophages, QHSG was discovered to hinder the generation of inflammatory cytokines by lessening TLR4/NF-κB signaling pathway activity and weakening NLRP3 inflammasome activation. Taken together, QHSG may resolve acute lung injury, attributed to its anti-inflammation and immunoregulation by attenuation of TLR4/NF-κB signaling cascade and inhibition of NLRP3 inflammasome activation. Our findings provide a novel insight into the action mechanism of QHSG and lay a mechanistic foundation for therapeutic intervention in acute lung injury with QHSG in clinical practice.
Collapse
Affiliation(s)
- Shun Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanjing Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Minmin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiarong Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhui Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenyu Yang
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmaceutical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
19
|
Ge R, Wang F, Peng Z. Advances in Biomarkers for Diagnosis and Treatment of ARDS. Diagnostics (Basel) 2023; 13:3296. [PMID: 37958192 PMCID: PMC10649435 DOI: 10.3390/diagnostics13213296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/10/2023] [Accepted: 05/18/2023] [Indexed: 11/15/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and fatal disease, characterized by lung inflammation, edema, poor oxygenation, and the need for mechanical ventilation, or even extracorporeal membrane oxygenation if the patient is unresponsive to routine treatment. In this review, we aim to explore advances in biomarkers for the diagnosis and treatment of ARDS. In viewing the distinct characteristics of each biomarker, we classified the biomarkers into the following six categories: inflammatory, alveolar epithelial injury, endothelial injury, coagulation/fibrinolysis, extracellular matrix turnover, and oxidative stress biomarkers. In addition, we discussed the potential role of machine learning in identifying and utilizing these biomarkers and reviewed its clinical application. Despite the tremendous progress in biomarker research, there remain nonnegligible gaps between biomarker discovery and clinical utility. The challenges and future directions in ARDS research concern investigators as well as clinicians, underscoring the essentiality of continued investigation to improve diagnosis and treatment.
Collapse
Affiliation(s)
- Ruiqi Ge
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China;
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Fengyun Wang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China;
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China;
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| |
Collapse
|
20
|
Peng W, Qi H, Zhu W, Tong L, Rouzi A, Wu Y, Han L, He L, Yan Y, Pan T, Liu J, Wang Q, Jia Z, Song Y, Zhu Q, Zhou J. Lianhua Qingke ameliorates lipopolysaccharide-induced lung injury by inhibiting neutrophil extracellular traps formation and pyroptosis. Pulm Circ 2023; 13:e12295. [PMID: 37808899 PMCID: PMC10557103 DOI: 10.1002/pul2.12295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023] Open
Abstract
LHQK is a patented Traditional Chinese Medicine (TCM) which is clinically used for acute tracheobronchitis, cough, and other respiratory diseases. Recent studies have proved that LHQK exhibits excellent clinical efficacy in the treatment of acute lung injury (ALI). However, the corresponding mechanisms remain largely unexplored. In this study, we investigated the effects and the underlying mechanisms of LHQK on lipopolysaccharide (LPS)-induced ALI in mice. The pathological examination, inflammatory cytokines assessments, and mucus secretion evaluation indicated that administration of LHQK ameliorated LPS-induced lung injury, and suppressed the secretion of Muc5AC and pro-inflammatory cytokines (IL-6, TNF-α, and IL-1β) in plasma and BALF. Furthermore, the results of cell-free DNA level showed that LHQK significantly inhibited LPS-induced NETs formation. Western blot revealed that LHQK effectively inhibited LPS-triggered pyroptosis in the lung. In addition, RNA-Seq data analysis, relatively bioinformatic analysis, and network pharmacology analysis revealed that LHQK and relative components may play multiple protective functions in LPS-induced ALI/acute respiratory distress syndrome (ARDS) by regulating multiple targets directly or indirectly related to NETs and pyroptosis. In conclusion, LHQK can effectively attenuate lung injury and reduce lung inflammation by inhibiting LPS-induced NETs formation and pyroptosis, which may be regulated directly or indirectly by active compounds of LHQK.
Collapse
Affiliation(s)
- Wenjun Peng
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western MedicineHebeiShijiazhuangChina
| | - Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Lin Tong
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ainiwaer Rouzi
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Yuanyuan Wu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Linxiao Han
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ludan He
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Yu Yan
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ting Pan
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Jie Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Zhenhua Jia
- Hebei Academy of Integrated Traditional Chinese and Western MedicineHebeiShijiazhuangChina
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan UniversityFudan UniversityShanghaiChina
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan UniversityFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionFudan UniversityShanghaiChina
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan UniversityFudan UniversityShanghaiChina
| |
Collapse
|
21
|
Hua F, Cui E, Lv L, Wang B, Li L, Lu H, Chen N, Chen W. Fecal microbiota transplantation from HUC-MSC-treated mice alleviates acute lung injury in mice through anti-inflammation and gut microbiota modulation. Front Microbiol 2023; 14:1243102. [PMID: 37840733 PMCID: PMC10569429 DOI: 10.3389/fmicb.2023.1243102] [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: 06/20/2023] [Accepted: 08/29/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Acute lung injury (ALI) is a severe respiratory tract disorder facilitated by dysregulated inflammation, oxidative stress and intestinal ecosystem. Fecal microbiota transplantation (FMT) is a rapid method for gut microbiota (GM) reconstruction. Furthermore, our previous studies have confirmed that human umbilical cord mesenchymal stromal cells (HUC-MSCs) can alleviate ALI by improving GM composition. Therefore, we aimed to explore the efficacy and mechanism of FMT from HUC-MSCs-treated mice on ALI. Methods In brief, fresh feces from HUC-MSCs-treated mice were collected for FMT, and the mice were randomly assigned into NC, FMT, LPS, ABX-LPS, and ABX-LPS-FMT groups (n = 12/group). Subsequently, the mice were administrated with antibiotic mixtures to deplete GM, and given lipopolysaccharide and FMT to induce ALI and rebuild GM. Next, the therapeutic effect was evaluated by bronchoalveolar lavage fluid (BALF) and histopathology. Immune cells in peripheral blood and apoptosis in lung tissues were measured. Furthermore, oxidative stress- and inflammation-related parameter levels were tested in BALF, serum, lung and ileal tissues. The expressions of apoptosis-associated, TLR4/NF-κB pathway-associated, Nrf2/HO-1 pathway related and tightly linked proteins in the lung and ileal tissues were assessed. Moreover, 16S rRNA was conducted to assess GM composition and distribution. Results Our results revealed that FMT obviously improved the pathological damage of lung and ileum, recovered the immune system of peripheral blood, decreased the cell apoptosis of lung, and inhibited inflammation and oxidative stress in BALF, serum, lung and ileum tissues. Moreover, FMT also elevated ZO-1, claudin-1, and occludin protein expressions, activating the Nrf2/HO-1 pathway but hindering the TLR4/NF-κB pathway. Of note, the relative abundances of Bacteroides, Christensenella, Coprococcus, and Roseburia were decreased, while the relative abundances of Xenorhabdus, Sutterella, and Acinetobacter were increased in the ABX-LPS-FMT group. Conclusion FMT from HUC-MSCs-treated mice may alleviate ALI by inhibiting inflammation and reconstructing GM, additionally, we also found that the TLR4/NF-κB and Nrf2/HO-1 pathways may involve in the improvement of FMT on ALI, which offers novel insights for the functions and mechanisms of FMT from HUC-MSCs-treated mice on ALI.
Collapse
Affiliation(s)
- Feng Hua
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Enhai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Bin Wang
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Liqin Li
- Traditional Chinese Medicine Key Laboratory Cultivation Base of Zhejiang Province for the Development and Clinical Transformation of Immunomodulatory Drugs, Huzhou, China
| | - Huadong Lu
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Na Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Wenyan Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| |
Collapse
|
22
|
Joshi JM, Muttigi MS, Upadhya R, Seetharam RN. An overview of the current advances in the treatment of inflammatory diseases using mesenchymal stromal cell secretome. Immunopharmacol Immunotoxicol 2023:1-11. [PMID: 36786742 DOI: 10.1080/08923973.2023.2180388] [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: 02/15/2023]
Abstract
The growing interest in mesenchymal stromal cell (MSC) therapy has been leading to the utilization of its therapeutic properties in a variety of inflammatory diseases. The clinical translation of the related research from bench to bedside is cumbersome due to some obvious limitations of cell therapy. It is evident from the literature that the MSC secretome components mediate their wide range of functions. Cell-free therapy using MSC secretome is being considered as an emerging and promising area of biotherapeutics. The secretome mainly consists of bioactive factors, free nucleic acids, and extracellular vesicles. Constituents of the secretome are greatly influenced by the cell's microenvironment. The broad array of immunomodulatory properties of MSCs are now being employed to target inflammatory diseases. This review focuses on the emerging MSC secretome therapies for various inflammatory diseases. The mechanism of action of the various anti-inflammatory factors is discussed. The potential of MSC secretome as a viable anti-inflammatory therapy is deliberated.
Collapse
Affiliation(s)
- Jahnavy Madhukar Joshi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manjunatha S Muttigi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghavendra Upadhya
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raviraja N Seetharam
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
23
|
Chen C, Li L, Liu X, Zhang D, Liu Y, Li Y. 23-O-acetylshengmanol-3-O-α-L-arabinoside alleviates lipopolysaccharide-induced acute lung injury through inhibiting IκB/NF-κB and MAPK/AP-1 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115725. [PMID: 36115602 DOI: 10.1016/j.jep.2022.115725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/05/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cimicifuga foetida L. is a well-established traditional Chinese medicine with heat-clearing and detoxifying effects and has good therapeutic effect on oral mucosal ulcer and pharyngitis. The rhizome of this herb is rich in triterpenoid glycosides, including 23-O-acetylshengmanol-3-o-α-L-arabinoside (DA). AIM OF THE STUDY Whether and how DA attenuates acute lung injury (ALI) are unclear. Accordingly, we focused on its anti-inflammatory effects and underlying molecular mechanisms in lipopolysaccharide (LPS)-stimulated ALI mice and RAW264.7 cells. MATERIALS AND METHODS The model of ALI mice was established by exposed intratracheal instillation of LPS. Lung pathological changes were evaluated by hematoxylin and eosin staining. Pulmonary function was assessed by whole-body plethysmography. Total protein content in bronchoalveolar lavage fluid (BALF) was detected by bicinchoninic acid method. Wet/dry lung ratio was used to evaluate the degree of pulmonary edema in mice. The levels of pro-inflammatory mediators were measured using enzyme-linked immunosorbent assay. The relative expression of pro-inflammatory gene mRNA was examined by RT-qPCR. The expression of inflammatory-related proteins was detected by Western blot. RAW264.7 cells were used to test the anti-inflammatory effects of DA in vitro. Cytotoxicity was assessed using a MTT assay. Nitric oxide production was measured by Griess assay. The production and expression of inflammatory mediators and the protein levels of inflammatory signaling molecules in the NF-κB and MAPK pathways were measured. Furthermore, immunofluorescence staining was used to analyze the expression of p-IκBα, p-ERK, and p-p38 in lung macrophages and the nuclear translocation of NF-κB p65 and AP-1 in cells. RESULTS DA evidently alleviated histopathological changes and ameliorated pulmonary edema. Moreover, DA could reduce excessive inflammatory reaction in lung tissue as manifested by the reduction of proinflammatory mediators (IL-1β, IL-6, TNF-α, MCP-1, iNOS, and COX-2) in BALF, serum, and lung tissues. Further, DA inhibited the activation of the NLRP3/caspase-1 pathway in the lung. DA reduced the production and expression of the proinflammatory mediators above in RAW264.7 cells. Mechanistically, DA remarkably blocked the nuclear translocation of NF-κB p65, suppressed IκBα phosphorylation, and markedly reduced the nuclear translocation of AP-1 and the phosphorylation of ERK and p38. CONCLUSIONS The findings demonstrated that DA exerts anti-inflammatory effects in LPS-stimulated ALI mice and macrophages by downregulating the NLRP3/caspase-1 signaling pathway in lung tissue and the IκB/NF-κB and MAPKs/AP-1 pathways in macrophages, suggesting that DA may be promising in ALI treatment.
Collapse
Affiliation(s)
- Chunyan Chen
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaomei Liu
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Deqin Zhang
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yang Liu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
24
|
Cui E, Zhang L, Pan X, Zhang Q, Zhang L, Wu F, Chen N, Lv L, Chen W, Chen H, Lin A, Wang F, Liang J, Pan R. RNA-Sequencing approach for exploring the therapeutic effect of umbilical cord mesenchymal stem/stromal cells on lipopolysaccharide-induced acute lung injury. Front Immunol 2022; 13:1021102. [PMID: 36341363 PMCID: PMC9632738 DOI: 10.3389/fimmu.2022.1021102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/03/2022] [Indexed: 11/20/2022] Open
Abstract
Acute lung injury (ALI) is significantly associated with morbidity and mortality in patients with critical diseases. In recent years, studies have identified that mesenchymal stem/stromal cells (MSCs) ameliorate ALI and pulmonary fibrosis. However, the mechanism underlying this outcome in ALI has not yet been investigated. In this study, RNA sequencing technology was used to analyze the gene expression profile of lung tissue in lipopolysaccharide (LPS)-induced ALI rats following treatment with human umbilical cord MSC (HUCMSC). Differential expression analyses, gene ontology annotation, Kyoto Encyclopedia of Genes and Genomes enrichment, protein–protein interaction network identification, and hub gene analysis were also performed. HUCMSC treatment decreased inflammatory factor production and alveolar exudates, and attenuated lung damage in LPS-induced ALI rats. The RNA-Seq data indicated that HUCMSC treatment activated the IL-17, JAK-STAT, NF-κB, and TNF-α signaling pathways, increased oxygen transport, and decreased extracellular matrix organization. HUCMSC exert beneficial effects on ALI via these signaling pathways by reducing inflammation, inhibiting pulmonary fibrosis, and improving lung ventilation. Moreover, our study further revealed the hub genes (Tbx2, Nkx2-1, and Atf5) and signaling pathways involved in HUCMSC treatment, thus providing novel perspectives for future research into the molecular mechanisms underlying cell treatment of ALI. HUCMSC can regulate multiple genes and signaling pathways, which can prevent LPS-induced lung damage in an ALI rat model.
Collapse
Affiliation(s)
- Enhai Cui
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Luwen Zhang
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Qiang Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Ling Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Feifei Wu
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Na Chen
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lu Lv
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wenyan Chen
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Chen
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Feng Wang
- Department of Nephrology, Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinfeng Liang
- Department of Drug Evaluation, Zhejiang Center for Drug & Cosmetic Evaluation, Hangzhou, China
- *Correspondence: Ruolang Pan, ; Jinfeng Liang,
| | - Ruolang Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
- *Correspondence: Ruolang Pan, ; Jinfeng Liang,
| |
Collapse
|
25
|
Kaigorodov DG, Kaigorodova AD. The non-protein fraction of embryonic stem cell secretome has antibacterial effects against antibiotic-resistant strains of bacteria. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2022. [DOI: 10.15789/2220-7619-npf-1940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In recent years, the search for new antibacterial agents has been shown to be extremely important, as the burgeoning problem of antibiotic resistance and the toxicity of many antimicrobial compounds has forced scientists to turn their attention to alternatives. Searching stem cell secretomes, including the non-protein part, for new antimicrobials is a promising area of current research. We investigated the effect of the non-protein part of an embryonic stem cell secretome on various bacterial strains, including antibiotic-resistant ones. The non-protein fraction of the stem cell secretome was obtained by preparative high-performance liquid chromatography. Bactericidal activity was tested against eight museum strains and 206 clinical strains of bacteria by comparing the secretomes effects on growth of bacterial cultures. The museum strains showed some dose-dependent effects at concentrations of 25-100 g/ml. Against the clinical strains of Gram-negative microorganisms of different species, some bactericidal activity was shown at a concentration of 100 g/ml, but sensitivity of bacteria to the secretome fraction varied, with growth stimulation being detected in some strains. Application of higher concentrations of 100-1000 g/ml showed no dose-dependent effect. The clinical strains of E. coli and P. aeruginosa were shown to have reduced bactericidal activity after one day of incubation. Thus, this study has shown that the non-protein fraction of the embryonic stem cell secretome has bactericidal effects against some strains. However, more detailed studies are needed to identify the mechanism of action and to determine the most effective dose and frequency of administration.
Collapse
|
26
|
Wei S, Li J, Tang M, Zhang K, Gao X, Fang L, Liu W. STAT3 and p63 in the Regulation of Cancer Stemness. Front Genet 2022; 13:909251. [PMID: 36061200 PMCID: PMC9428145 DOI: 10.3389/fgene.2022.909251] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/13/2022] [Indexed: 11/28/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor with many important functions in normal and transformed cells. STAT3 regulatory activities are highly complex as they are involved in various signaling pathways in different cell types under different conditions. Biologically, STAT3 is a regulative factor for normal and cancer stem cells (CSCs). Tumor protein p63 (p63), a member of the p53 protein family, is involved in these biological processes and is also physically and functionally associated with STAT3. STAT3 activation occurs during various aspects of carcinogenesis, including regulation of CSCs properties. In combination with p63, STAT3 is a possible biological marker of CSCs and a major regulator of maintenance of stemness in CSCs. We summarized the STAT3 functions and regulation and its role in CSC properties and highlight how these are affected by its associations with p63.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Wang J, Luo F, Suo Y, Zheng Y, Chen K, You D, Liu Y. Safety, efficacy and biomarkers analysis of mesenchymal stromal cells therapy in ARDS: a systematic review and meta-analysis based on phase I and II RCTs. Stem Cell Res Ther 2022; 13:275. [PMID: 35752865 PMCID: PMC9233855 DOI: 10.1186/s13287-022-02956-3] [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: 04/05/2022] [Accepted: 06/08/2022] [Indexed: 11/11/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) therapy for acute respiratory distress syndrome (ARDS) is an emerging treatment, but most of the current trials of MSCs stay in the animal experimental stage, and the safety and efficacy of MSCs in clinical application are not clear. We aimed to analyze the safety, efficacy and biomarkers of mesenchymal stromal cells in the treatment of ARDS. Methods For this systematic review and meta-analysis, we searched PubMed, Embase, Cochrane Central Register of Controlled Trials, Web of science, CNKI, VIP and Wan Fang data, studies published between database inception and Mar 17, 2022. All randomized controlled trials (RCT) of stem cell interventions for ARDS were included, without language or date restrictions. We did separate meta-analyses for mortality, subjects with adverse events (AEs) and subjects with serious adverse events (SAEs). Since the trials data are dichotomous outcomes, the odds ratio (OR) is adopted for meta-analysis. The quality of the evidence was assessed with the Cochrane risk of bias tool. Findings In total, 5 trials involving 171 patients with ARDS were included in this meta-analysis. A total of 99 individuals were randomly assigned to receive MSCs treatment, and 72 were randomly assigned to receive placebo treatment. Treatment with MSCs appeared to increase the occurrence of adverse events, but this result was not statistically significant (OR, 1.58; 95%CI, 0.64–3.91; P = 0.32). The occurrence of serious adverse events was lower in the MSCs group than in the placebo group (OR, 0.57; 95%CI, 0.14–2.32; P = 0.43); there seems to be no significant difference between the two groups in terms of 28 days mortality (OR, 0.93; 95%CI, 0.45–1.89); oxygenation index and biomarkers showed a tendency to improve in treatment, but there was a lack of more statistically significant clinical evidence to support them. Interpretation Based on the current clinical trials, MSCs intervention has some safety for ARDS patients, but its effectiveness and predictive value of airspace biomarkers need to be determined by more large-scale, standard randomized controlled trials. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02956-3.
Collapse
Affiliation(s)
- Jianbao Wang
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Fenbin Luo
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Ye Suo
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Yuxin Zheng
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Kaikai Chen
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Deyuan You
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Yuqi Liu
- Department of Respiratory and Critical Care Medicine, Fujian Respiratory Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China.
| |
Collapse
|
28
|
Wang MY, Cui P, Xin HM. [Research advances of the roles of sphingosine-1-phosphate in acute lung injury]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:496-500. [PMID: 35599427 DOI: 10.3760/cma.j.cn501120-20210703-00234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Sphingosine-1-phosphate (S1P) is the main metabolite produced in the process of phospholipid metabolism, which can promote proliferation, migration, and apoptosis of cells, and maintain the barrier function of vascular endothelium. The latest researches showed that S1P can alleviate acute lung injury (ALI) and the inflammation caused by ALI, while the dosage of S1P is still needed to be considered. Mesenchymal stem cells (MSCs) have been a emerging therapy with potential therapeutic effects on ALI because of their characteristics of self-replication and multi-directional differentiation, and their advantages in hematopoiesis, immune regulation, and tissue repair. S1P can promote differentiation of MSCs and participate in immune regulation, while MSCs can regulate the homeostasis of S1P in the body. The synergistic effect of S1P and MSC provides a new treatment method for ALI. This article reviews the production and biological function of S1P, receptor and signal pathway of S1P, the therapeutic effects of S1P on ALI, and the research advances of S1P combined with MSCs in the treatment of ALI, aiming to provide theoretical references for the development of S1P targeted drugs in the treatment of ALI and the search for new combined treatment schemes for ALI.
Collapse
Affiliation(s)
- M Y Wang
- College of Life Sciences, Guangxi Normal University, Guilin 541002, China Lung Injury Group of Guangxi Key Laboratory of Metabolic Diseases, Department of Burns and Plastic Surgery, the 924th Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Guilin 541002, China
| | - P Cui
- Lung Injury Group of Guangxi Key Laboratory of Metabolic Diseases, Department of Burns and Plastic Surgery, the 924th Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Guilin 541002, China
| | - H M Xin
- Lung Injury Group of Guangxi Key Laboratory of Metabolic Diseases, Department of Burns and Plastic Surgery, the 924th Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Guilin 541002, China
| |
Collapse
|
29
|
Dai J, Zhang X, Zhang J, Yang W, Yang X, Bian H, Chen Z. Blockade of mIL‐6R alleviated lipopolysaccharide‐induced systemic inflammatory response syndrome by suppressing NF‐κB‐mediated Ccl2 expression and inflammasome activation. MedComm (Beijing) 2022; 3:e132. [PMID: 35548710 PMCID: PMC9075038 DOI: 10.1002/mco2.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is characterized by dysregulated cytokine release, immune responses and is associated with organ dysfunction. IL‐6R blockade indicates promising therapeutic effects in cytokine release storm but still remains unknown in SIRS. To address the issue, we generated the human il‐6r knock‐in mice and a defined epitope murine anti‐human membrane‐bound IL‐6R (mIL‐6R) mAb named h‐mIL‐6R mAb. We found that the h‐mIL‐6R and the commercial IL‐6R mAb Tocilizumab significantly improved the survival rate, reduced the levels of TNF‐α, IL‐6, IL‐1β, IFN‐γ, transaminases and blood urea nitrogen of LPS‐induced SIRS mice. Besides, the h‐mIL‐6R mAb could also dramatically reduce the levels of inflammatory cytokines in LPS‐treated THP‐1 cells in vitro. RNA‐seq analysis indicated that the h‐mIL‐6R mAb could regulate LPS‐induced activation of NF‐κB/Ccl2 and NOD‐like receptor signaling pathways. Furthermore, we found that the h‐mIL‐6R mAb could forwardly inhibit Ccl2 expression and NLRP3‐mediated pyroptosis by suppressing NF‐κB in combination with the NF‐κB inhibitor. Collectively, mIL‐6R mAbs suppressed NF‐κB/Ccl2 signaling and inflammasome activation. IL‐6R mAbs are potential alternative therapeutics for suppressing excessive cytokine release, over‐activated inflammatory responses and alleviating organ injuries in SIRS.
Collapse
Affiliation(s)
- Ji‐Min Dai
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
- Faculty of Hepato‐Biliary‐Pancreatic Surgery The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital Beijing P.R. China
| | - Xue‐Qin Zhang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Jia‐Jia Zhang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Wei‐Jie Yang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Xiang‐Min Yang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| |
Collapse
|
30
|
Inhaled Placental Mesenchymal Stromal Cell Secretome from Two- and Three-Dimensional Cell Cultures Promotes Survival and Regeneration in Acute Lung Injury Model in Mice. Int J Mol Sci 2022; 23:ijms23073417. [PMID: 35408778 PMCID: PMC8998959 DOI: 10.3390/ijms23073417] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is a common clinical problem, leading to significant morbidity and mortality, and no effective pharmacotherapy exists. The problem of ARDS causing mortality became more apparent during the COVID-19 pandemic. Biotherapeutic products containing multipotent mesenchymal stromal cell (MMSC) secretome may provide a new therapeutic paradigm for human healthcare due to their immunomodulating and regenerative abilities. The content and regenerative capacity of the secretome depends on cell origin and type of cultivation (two- or three-dimensional (2D/3D)). In this study, we investigated the proteomic profile of the secretome from 2D- and 3D-cultured placental MMSC and lung fibroblasts (LFBs) and the effect of inhalation of freeze-dried secretome on survival, lung inflammation, lung tissue regeneration, fibrin deposition in a lethal ALI model in mice. We found that three inhaled administrations of freeze-dried secretome from 2D- and 3D-cultured placental MMSC and LFB protected mice from death, restored the histological structure of damaged lungs, and decreased fibrin deposition. At the same time, 3D MMSC secretome exhibited a more pronounced trend in lung recovery than 2D MMSC and LFB-derived secretome in some measures. Taking together, these studies show that inhalation of cell secretome may also be considered as a potential therapy for the management of ARDS in patients suffering from severe pneumonia, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), however, their effectiveness requires further investigation.
Collapse
|
31
|
Kaya-Tilki E, Dikmen M. Neuroprotective effects of some epigenetic modifying drugs' on Chlamydia pneumoniae-induced neuroinflammation: A novel model. PLoS One 2021; 16:e0260633. [PMID: 34847172 PMCID: PMC8631675 DOI: 10.1371/journal.pone.0260633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/14/2021] [Indexed: 12/02/2022] Open
Abstract
Chlamydia pneumoniae (Cpn) is a gram-negative intracellular pathogen that causes a variety of pulmonary diseases, and there is growing evidence that it may play a role in Alzheimer's disease (AD) pathogenesis. Cpn can interact functionally with host histones, altering the host's epigenetic regulatory system by introducing bacterial products into the host tissue and inducing a persistent inflammatory response. Because Cpn is difficult to propagate, isolate, and detect, a modified LPS-like neuroinflammation model was established using lyophilized cell free supernatant (CFS) obtained from infected cell cultures, and the effects of CFS were compared to LPS. The neuroprotective effects of Trichostatin A (TSA), givinostat, and RG108, which are effective on epigenetic mechanisms, and the antibiotic rifampin, were studied in this newly introduced model and in the presence of amyloid beta (Aβ) 1-42. The neuroprotective effects of the drugs, as well as the effects of CFS and LPS, were evaluated in Aβ-induced neurotoxicity using a real-time cell analysis system, total ROS, and apoptotic impact. TSA, RG108, givinostat, and rifampin all demonstrated neuroprotective effects in both this novel model and Aβ-induced neurotoxicity. The findings are expected to provide early evidence on neuroprotective actions against Cpn-induced neuroinflammation and Aβ-induced neurotoxicity, which could represent a new treatment option for AD, for which there are currently few treatment options.
Collapse
Affiliation(s)
- Elif Kaya-Tilki
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
32
|
Lin X, Zhu Y, Le G. Tetramethylpyrazine Alleviates Tight Junction Disruption of Bronchial Mucosal Epithelial Cells Caused by Interleukin-17 via Inhibiting Nuclear Factor-κB-p65/Tumor Necrosis Factor-α Signaling Pathway. J Interferon Cytokine Res 2021; 41:415-424. [PMID: 34698528 DOI: 10.1089/jir.2021.0083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Bronchial mucosal epithelial dysregulation and barrier disruption are involved in the initiation and development of acute lung injury (ALI). Some inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-17 (IL-17) contribute to the pathological changes of ALI. However, the roles and relationship between TNF-α and IL-17 during the disruption of bronchial epithelial tight junction remain unclear. Tetramethylpyrazine (TMP) is confirmed to have beneficial functions in hemostasis, inflammation, and cell growth. Here, we demonstrated the protective effects of TMP on bronchial mucosal epithelial injury induced by IL-17. We showed that IL-17 stimulation in vitro markedly reduced occludin and zonula occludens-1 (ZO-1) expression in bronchial mucosal epithelial cells via the nuclear factor-κB-p65/TNF-α signaling pathway, including NF-κB-p65-triggered TNF-α gene transcription and expression. TMP obviously rescued IL-17-induced occludin and ZO-1 downregulation. Mechanically, TMP substantially suppressed NF-κB-p65 activation and NF-κB-p65-induced TNF-α production in bronchial mucosal epithelial cells caused by IL-17. Taken together, this study indicates that TMP has a protective effect on bronchial mucosal epithelial cell injury due to IL-17 induction by inhibiting the NF-κB-p65/TNF-α signaling pathway.
Collapse
Affiliation(s)
- Xin Lin
- Department of Anesthesiology, Medical College of Soochow University, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, P.R. China
| | - Yong Zhu
- Department of Anesthesiology, Kunshan Sixth People's Hospital, Kunshan, P.R. China
| | - Guohui Le
- Department of Anesthesiology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, P.R. China
| |
Collapse
|
33
|
Peng W, Wu Y, Zhang G, Zhu W, Chang M, Rouzi A, Jiang W, Tong L, Wang Q, Liu J, Song Y, Li H, Li K, Zhou J. GLIPR1 Protects Against Cigarette Smoke-Induced Airway Inflammation via PLAU/EGFR Signaling. Int J Chron Obstruct Pulmon Dis 2021; 16:2817-2832. [PMID: 34675506 PMCID: PMC8517531 DOI: 10.2147/copd.s328313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a major health problem associated with high mortality worldwide. Cigarette smoke (CS) exposure is the main cause of COPD. Glioma pathogenesis-related protein 1 (GLIPR1) plays a key role in cell growth, proliferation, and invasion; however, the role of GLIPR1 in COPD remains unclear. Methods To clarify the involvement of GLIPR1 in COPD pathogenesis, Glipr1 knockout (Glipr1-/-) mice were generated. Wild-type (WT) and Glipr1-/- mice were challenged with CS for 3 months. To illustrate how GLIPR1 regulates CS-induced airway damage, knockdown experiments targeting GLIPR1 and PLAU, as well as overexpression experiments of PLAU, were performed with human bronchial epithelial cells. Results Compared with WT mice, Glipr1-/- mice showed exacerbated CS-induced airway damage including lung inflammation, airway wall thickening, and alveolar destruction. After CS exposure, total proteins, total white cells, neutrophils, lymphocytes, IL-6, and matrix metalloproteinase-9 increased significantly in lung of Glipr1-/- mice than those in lung of WT mice. Furthermore, in vivo and in vitro experiments demonstrated that silencing of GLIPR1 inactivated PLAU/EGFR signaling and promoted caspase-1-dependent pyroptosis (a mode of inflammatory cell death) induced by CS and CS extract exposure, respectively. In vitro experiments further revealed the interaction between GLIPR1 and PLAU, and silencing of PLAU blocked EGFR signaling and promoted pyroptosis, while overexpression of PLAU activated EGFR signaling and reversed pyroptosis. Conclusion To conclude, GLIPR1 played a pivotal role in COPD pathogenesis and protected against CS-induced inflammatory response and airway damage, including cell pyroptosis, through the PLAU/EGFR signaling. Thus, GLIPR1 may play a potential role in COPD treatment.
Collapse
Affiliation(s)
- Wenjun Peng
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yuanyuan Wu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ge Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Meijia Chang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ainiwaer Rouzi
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Weipeng Jiang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Lin Tong
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yuanlin Song
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, 200540, People's Republic of China.,Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, People's Republic of China.,Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, Shanghai, 200032, People's Republic of China
| | - Huayin Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ka Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, 200540, People's Republic of China.,Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, People's Republic of China.,Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, Shanghai, 200032, People's Republic of China
| |
Collapse
|