1
|
Fang X, Mo C, Zheng L, Gao F, Xue FS, Zheng X. Transfusion-Related Acute Lung Injury: from Mechanistic Insights to Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413364. [PMID: 39836498 PMCID: PMC11923913 DOI: 10.1002/advs.202413364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/08/2024] [Indexed: 01/23/2025]
Abstract
Transfusion-related acute lung injury (TRALI) is a potentially lethal complication of blood transfusions, characterized by the rapid onset of pulmonary edema and hypoxemia within six hours post-transfusion. As one of the primary causes of transfusion-related mortality, TRALI carries a significant mortality rate of 6-12%. However, effective treatment strategies for TRALI are currently lacking, underscoring the urgent need for a comprehensive and in-depth understanding of its pathogenesis. This comprehensive review provides an updated and detailed analysis of the current landscape of TRALI, including its clinical presentation, pathogenetic hypotheses, animal models, cellular mechanisms, signaling pathways, and potential therapeutic targets. By highlighting the critical roles of these pathways and therapies, this review offers valuable insights to inform the development of preventative and therapeutic strategies and to guide future research efforts aimed at addressing this life-threatening condition.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Zheng
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fei Gao
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fu-Shan Xue
- Department of Anesthesiology/Critical Care Medicine, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University & Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical Medicine, Fujian Provincial Co-constructed Laboratory of "Belt and Road,", Fuzhou, Fujian, China
| |
Collapse
|
2
|
Yao L, Chen Z, Gan S, Fu C, Xie Z, Zhang H, Yang C, Fu L, Liu Y, Luo M, Kuang D, Cao J, Hu G, Li S, Yang L, Li L, Chen X, Tang H. Soluble E-cadherin contributes to inflammation in acute lung injury via VEGF/VEGFR2 signaling. Cell Commun Signal 2025; 23:113. [PMID: 40011876 DOI: 10.1186/s12964-025-02110-5] [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: 11/25/2024] [Accepted: 02/16/2025] [Indexed: 02/28/2025] Open
Abstract
As a gatekeeper of the airway epithelial cells, E-cadherin is not only a critical component for the maintenance of epithelial integrity, but also engaged in pathological processes through the release of a soluble form (sE-cadherin). This study was aimed to investigate the role of sE-cadherin in ALI/ARDS. Serum samples from patients with ARDS and healthy volunteers were collected for the detection of sE-cadherin. An LPS-induced mouse model was induced to analyze the expression of sE-cadherin, and a neutralizing antibody against sE-cadherin (DECMA-1) was given to the LPS-exposed mice. The effects of recombinant sE-cadherin were tested both in vitro and in vivo, and VEGFR2 inhibition was used to explore a possible mechanism for sE-cadherin-induced pulmonary inflammation. We observed an increased level of sE-cadherin in ARDS patients as well as in LPS-exposed mice. In vivo treatment of DECMA-1 significantly attenuated LPS-induced inflammation. In vitro, exogenous sE-cadherin can dramatically upregulate the expression of VEGF in THP1-derived macrophages and human primary macrophages. In addition, intratracheal instillation of recombinant sE-cadherin leads to significant increased infiltration of neutrophils as well as overproduction of IL-6 and IL1β, which could be attenuated by inhibition of VEGF/VEGFR2 signaling. While blockade of the VEGF/VEGFR2 pathway inhibited pulmonary inflammatory responses in LPS-exposed mice. Taken together, our data demonstrated that sE-cadherin contributes to lung inflammation in ALI/ARDS, which is related to activation of the VEGF/VEGFR2 pathway.
Collapse
Affiliation(s)
- Lihong Yao
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zemin Chen
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Sudan Gan
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chunlai Fu
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Zhefan Xie
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Hailing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changyun Yang
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lin Fu
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ye Liu
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Ming Luo
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Daibin Kuang
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Jingjing Cao
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Guodong Hu
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Shiyue Li
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liping Yang
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lingfeng Li
- Department of Emergency Medicine, Department of Respiratory and Critical Care Medicine, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Haixiong Tang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Xu S, Zhu T, Mou H, Tan S, Leong JM. Weakened Airway Epithelial Junctions and Enhanced Neutrophil Elastase Release Contribute to Age-Dependent Bacteremia Risk Following Pneumococcal Pneumonia. Aging Cell 2025:e14474. [PMID: 39778043 DOI: 10.1111/acel.14474] [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: 10/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Streptococcus pneumoniae (Sp; pneumococcus), the most common agent of community-acquired pneumonia, can spread systemically, particularly in the elderly, highlighting the need for adjunctive therapies. The airway epithelial barrier defends against bacteremia and is dependent upon apical junctional complex (AJC) proteins such as E-cadherin. After mouse lung challenge, pneumolysin (PLY), a key Sp virulence factor, stimulates epithelial secretion of an inflammatory eicosanoid, triggering the infiltration of polymorphonuclear leukocytes (PMNs) that secrete high levels of neutrophil elastase (NE), thus promoting epithelial damage and systemic infection. Here, pulmonary E-cadherin staining of intratracheally (i.t.) inoculated mice revealed PLY-mediated disruption of AJC independently of PMNs. Apical infection of air-liquid interface (ALI) respiratory epithelial monolayers similarly showed that PLY disrupts AJCs. This epithelial damage promoted PMN transmigration and bacterial apical-to-basolateral translocation, and pharmacologically fortifying epithelial barrier function diminished both barrier breach in vitro and bacteremia in vivo. E-cadherin staining after Sp i.t. inoculation of > 20-month-old mice, or apical infection of ALI monolayers derived from these mice, revealed an age-associated vulnerability to PLY-mediated AJC disruption, which in turn enhanced PMN migration and bacteremia. In addition, we found that PMNs from aged mice secrete increased levels of tissue-damaging NE. Simultaneous pharmacological inhibition of tissue-destructive NE and fortification of pulmonary epithelial barrier function was required to reduce the level of Sp bacteremia in aged mice to that of young mice. This work underscores the importance of fully characterizing the multifactorial sources of age-associated susceptibility in devising adjunctive therapies to mitigate invasive pneumococcal disease in the elderly.
Collapse
Affiliation(s)
- Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Tianmou Zhu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
- Stuart B Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University, Boston, MA, USA
| |
Collapse
|
4
|
Liu X, Su S, Xia L, Lei X, Zou S, Zhou L, Yang R, Li K, Lin P, Li Y. Lysophosphatidylcholine 14:0 Alleviates Lipopolysaccharide-Induced Acute Lung Injury via Protecting Alveolar Epithelial Barrier by Activation of Nrf2/HO-1 Pathway. J Inflamm Res 2024; 17:10533-10546. [PMID: 39659750 PMCID: PMC11630720 DOI: 10.2147/jir.s495227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
Background Acute lung injury (ALI) is characterized by diffuse alveolar injury and acute non-cardiac pulmonary edema, with high morbidity and mortality. Lysophosphatidylcholine 14:0 (LPC14:0) has anti-inflammatory and anti-oxidative effects in sepsis and bacteremia. We hypothesized that LPC14:0 could be a potential treatment for ALI. Therefore, the effects of LPC14:0 on lung epithelial cells and the underlying mechanism on ALI were investigated. Methods Lipopolysaccharide (LPS) was instilled intratracheally in vivo while the Murine Lung Epithelial-12 was stimulated by tert-butyl hydroperoxide (t-BHP) in vitro to induce the ALI model. In vivo, lung injury was evaluated by histopathological changes and pulmonary edema was assessed by wet/dry ratio. Evans blue infiltration in lung tissue, total protein content, total cell counts and inflammatory factors in bronchoalveolar lavage fluid were evaluated for alveolar permeability. In vitro, cell viability and cell death rate were assessed by cell counting kit-8 and Calcein-AM/PI stain respectively. The expression of ZO-1, Occludin, Nrf2, and HO-1 was evaluated by Western blot. Results LPC14:0 attenuated the LPS-stimulated lung injury and oxidative stress in vivo, and alleviated the t-BHP-induced cell damage in vitro. Moreover, LPC14:0 significantly inhibited the degradation of the tight junction proteins and activated the Nrf2/HO-1 signaling pathway both in vivo and in vitro. Mechanistically, ML385, the Nrf2 inhibitor, inhibited the protective effects of LPC14:0 on barrier function in vitro. Conclusion This study first demonstrated that LPC14:0 mitigated LPS-induced ALI and the destruction of tight junctions, at least in part through up-regulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Xiling Liu
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Shanshan Su
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Lijing Xia
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Xiong Lei
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Shangpu Zou
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Liwen Zhou
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Ruobing Yang
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Kai Li
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Pengcheng Lin
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Yuping Li
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| |
Collapse
|
5
|
Zhou L, Lin Y, Zhou T, Xue Y, Bellusci S, Shen M, Chen C, Chen C. Evidence that a Novel Chalcone Derivative, Compound 27, Acts on the Epithelium Via the PI3K/AKT/Nrf2-Keap1 Signaling Pathway, to Mitigate LPS-Induced Acute Lung Injury in Mice. Inflammation 2024:10.1007/s10753-024-02051-0. [PMID: 38789816 DOI: 10.1007/s10753-024-02051-0] [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: 02/26/2024] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024]
Abstract
Acute lung injury (ALI) is a highly heterogeneous clinical syndrome and an important cause of mortality in critically ill patients, with limited treatment options currently available. Chalcone, an essential secondary metabolite found in edible or medicinal plants, exhibits good antioxidant activity and simple structure for easy synthesis. In our study, we synthesized a novel chalcone derivative, compound 27 (C27). We hypothesized that C27 could be a potential treatment for acute respiratory distress syndrome (ARDS). Therefore, the protective effects of C27 on lung epithelial cells during ALI and the underlying molecular mechanisms were investigated. In vivo, Intratracheal instillation of LPS (10 mg/kg) was used to induce acute lung injury in mice. In vitro, the bronchial epithelial cell line (Beas-2b) was treated with 30 μM tert-butyl hydroperoxide (t-BHP) to simulate oxidative stress. Our findings demonstrate that pretreatment with C27 reduces LPS-induced oxidative destruction and cellular apoptosis in lung tissues of mice. Furthermore, it significantly attenuates t-BHP-induced cellular reactive oxygen species (ROS) generation, mitochondrial damage, and apoptosis in vitro. Mechanistically, the signaling pathway involving Nrf2-Keap1 and the downstream antioxidative proteins were activated by C27 in vivo. Additionally, PI3K inhibitor LY294002 and Nrf2 inhibitor ML385 abolished the effect of C27 in vitro, indicating that the protective effect of C27 is mediated via the PI3K/AKT/Nrf2-Keap1 pathway. Our study provides evidence that C27 protects against LPS-induced ALI by mitigating oxidative stress via activation of the PI3K/AKT/Nrf2-Keap1 signaling pathway. Therefore, we hypothesize that C27 represents a viable alternative for ALI therapy.
Collapse
Affiliation(s)
- Liqin Zhou
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuting Lin
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tengfei Zhou
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Yincong Xue
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Saverio Bellusci
- Department of Internal Medicine, German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the, Justus-Liebig University Giessen , 35392, Giessen, Germany
| | - Mengya Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Chengshui Chen
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| | - Chaolei Chen
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
6
|
Chen Z, Tang H, Gan S, Yang C, Li S, Li J, Yao L. Ferroptosis mediates airway epithelial E-cadherin dysfunction in LPS-induced acute lung injury. Pulm Pharmacol Ther 2024; 84:102284. [PMID: 38154519 DOI: 10.1016/j.pupt.2023.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Loss of E-cadherin in the airway epithelial cells is a critical contributor to the development of ALI/ARDS. Yet the underlying mechanisms are largely unknown. Increasing evidences have revealed the significance of ferroptosis in the pathophysiological process of ALI/ARDS. The aim of this study was to investigate the role of ferroptosis in dysregulation of airway epithelial E-cadherin in ALI/ARDS. METHODS BALB/c mice were subjected to intratracheal instillation of lipopolysaccharide (LPS) to establish an ALI model. Two inhibitors of ferroptosis, liproxstatin-1 (Lip-1, at the dose of 10 mg/kg and 30 mg/kg) and ferrostatin-1 (Fer-1, at the dose of 1 mg/kg and 5 mg/kg), were respectively given to the mice through intraperitoneal injection after LPS challenge. The expression of ferroptotic markers, full-length E-cadherin and soluble E-cadherin (sE-cadherin) were both detected. RESULTS LPS exposure dramatically down-regulated pulmonary expression of E-cadherin in mice, with profound loss of membrane E-cadherin in the airway epithelial cells and increased secretion of sE-cadherin in the airway lumen. At the same time, we found that the mitochondrial of airway epithelial cells in LPS-exposed mice exhibited significant morphological alterations that are hallmark features of ferroptosis, with smaller volume and increased membrane density. Other makers of ferroptosis were also detected, including increased cytoplasmic levels of iron and lipid peroxidates (MDA), as well as decreased GPX4 expression. 30 mg/kg of Lip-1 not only showed potent protective effects against the LPS-induced injury, inflammation, edema of the lung in those mice, but also rescued airway epithelial E-cadherin expression and decreased the release of sE-cadherin through inhibiting ferroptosis. While no noticeable changes induced by LPS were observed in mice treated with Lip-1 at 10 mg/kg nor Fer-1 at 1 mg/kg or 5 mg/kg. CONCLUSIONS Taken together, these data demonstrated that ferroptosis mediates airway epithelial E-cadherin dysfunction in LPS-induced ALI.
Collapse
Affiliation(s)
- Zemin Chen
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixiong Tang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sudan Gan
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changyun Yang
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyue Li
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lihong Yao
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Zhang H, Feng Y, Si Y, Lu C, Wang J, Wang S, Li L, Xie W, Yue Z, Yong J, Dai S, Zhang L, Li X. Shank3 ameliorates neuronal injury after cerebral ischemia/reperfusion via inhibiting oxidative stress and inflammation. Redox Biol 2024; 69:102983. [PMID: 38064762 PMCID: PMC10755590 DOI: 10.1016/j.redox.2023.102983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/01/2024] Open
Abstract
Shank3, a key molecule related to the development and deterioration of autism, has recently been found to downregulate in the murine brain after ischemia/reperfusion (I/R). Despite this discovery, however, its effects on neuronal injury and the mechanism underlying the effects remain to be clarified. To address this, in this study, based on genetically modified mice models, we revealed that the expression of Shank3 showed a time-dependent change in murine hippocampal neurons after I/R, and that conditional knockout (cko) of Shank3 in neurons resulted in aggravated neuronal injuries. The protective effects of Shank3 against oxidative stress and inflammation after I/R were achieved through direct binding STIM1 and subsequent proteasome-mediated degradation of STIM1. The STIM1 downregulation induced the phosphorylation of downstream Nrf2 Ser40, which subsequently translocated to the nucleus, and further increased the expression of antioxidant genes such as NQO1 and HO-1 in HT22 cells. In vivo, the study has further confirmed that double knockout of Shank3 and Stim1 alleviated oxidative stress and inflammation after I/R in Shank3cko mice. In conclusion, the present study has demonstrated that Shank3 interacts with STIM1 and inhibits post-I/R neuronal oxidative stress and inflammatory response via the Nrf2 pathway. This interaction can potentially contribute to the development of a promising method for I/R treatment.
Collapse
Affiliation(s)
- Hongchen Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Feng
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanfang Si
- Department of Ophthalmology, The Eighth Medical Center, Affiliated to the Senior Department of Ophthalmology, The Third Medical Center, Chinese People's Liberation Army General Hospital, Beijing, 100091, China
| | - Chuanhao Lu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiquan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenyu Xie
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheming Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Yong
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
8
|
Huang Y, Dong S, Li X, Shi J, Zhang Y, Liu S, Zhang Y, Yu J. VNS-mediated α7nAChR signaling promotes SPM synthesis via regulation of netrin-1 expression during LPS-induced ALI. FASEB J 2024; 38:e9664. [PMID: 38038805 DOI: 10.1096/fj.202301623r] [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: 08/09/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023]
Abstract
The α7 nicotinic acetylcholine receptor (α7nAChR) plays a crucial role in the cholinergic anti-inflammatory pathway (CAP) during sepsis-associated acute lung injury (ALI). Increasing evidence suggests that specialized pro-resolving mediators (SPMs) are important in resolving α7nAChR-mediated ALI resolution. Our study aims to elucidate the pivotal role of α7nAChR in the CAP during LPS-associated acute lung injury (ALI). By employing vagus nerve stimulation (VNS), we identified α7nAChR as the key CAP subunit in ALI mice, effectively reducing lung permeability and the release of inflammatory cytokines. We further investigated the alterations in SPMs regulated by α7nAChR, revealing a predominant synthesis of lipoxin A4 (LXA4). The significance of α7nAChR-netrin-1 pathway in governing SPM synthesis was confirmed through the use of netrin-1 knockout mice and siRNA-transfected macrophages. Additionally, our evaluation identified a synchronous alteration of LXA4 synthesis in the α7nAChR-netrin-1 pathway accompanied by 5-lipoxygenase (5-LOX), thereby confirming an ameliorative effect of LXA4 on lung injury and macrophage inflammatory response. Concurrently, inhibiting the function of LXA4 annulled the lung-protective effect of VNS. As a result, our findings reveal a novel anti-inflammatory pathway wherein VNS modulates netrin-1 expression via α7nAChR, ultimately leading to LXA4 synthesis and subsequent lung protection.
Collapse
Affiliation(s)
- Yan Huang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shasha Liu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Ye Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
9
|
Jin Z, Shao Z, Yang S, Guo A, Han Y, Wu Y, Zhao Y, Wu Y, Shen J, Zhang M, Zhan X, Diao W, Ying S, Zhang C, Li W, Shen H, Chen Z, Yan F. Airway epithelial cGAS inhibits LPS-induced acute lung injury through CREB signaling. Cell Death Dis 2023; 14:844. [PMID: 38114479 PMCID: PMC10730695 DOI: 10.1038/s41419-023-06364-0] [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: 07/08/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
Increased levels of cytosolic DNA in lung tissues play an important role in acute lung injury. However, the detailed mechanisms involved remain elusive. Here, we found that cyclic GMP-AMP synthase (cGAS, a cytosolic DNA sensor) expression was increased in airway epithelium in response to increased cytosolic DNA. Conditional deletion of airway epithelial cGAS exacerbated acute lung injury in mice, cGAS knockdown augmented LPS-induced production of interleukin (IL)-6 and IL-8. Mechanically, deletion of cGAS augmented expression of phosphorylated CREB (cAMP response element-binding protein), and cGAS directly interacted with CREB via its C-terminal domain. Furthermore, CREB knockdown rescued the LPS-induced excessive inflammatory response caused by cGAS deletion. Our study demonstrates that airway epithelial cGAS plays a protective role in acute lung injury and confirms a non-canonical cGAS-CREB pathway that regulates the inflammatory responses in airway epithelium to mediate LPS-induced acute lung injury.
Collapse
Affiliation(s)
- Zhangchu Jin
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Zhehua Shao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Shiyi Yang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Anyi Guo
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yinling Han
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yinfang Wu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yun Zhao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yanping Wu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Jiaxin Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Min Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xueqin Zhan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310009, Zhejiang, China
| | - Wenqi Diao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Songmin Ying
- International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
- Department of Pharmacology and Department of Respiratory and Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Chao Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Department of Anatomy, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Huahao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
- State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China.
| | - Zhihua Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| | - Fugui Yan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
10
|
Jiao Y, Zhang T, Liu M, Zhou L, Qi M, Xie X, Shi X, Gu X, Ma Z. Exosomal PGE2 from M2 macrophages inhibits neutrophil recruitment and NET formation through lipid mediator class switching in sepsis. J Biomed Sci 2023; 30:62. [PMID: 37533081 PMCID: PMC10394797 DOI: 10.1186/s12929-023-00957-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Excess polymorphonuclear neutrophil (PMN) recruitment or excessive neutrophil extracellular trap (NET) formation can lead to the development of multiple organ dysfunction during sepsis. M2 macrophage-derived exosomes (M2-Exos) have exhibited anti-inflammatory activities in some inflammatory diseases to mediate organ functional protection, but their role in treating sepsis-related acute lung injury (ALI) remains unclear. In this study, we sought to investigate whether M2-Exos could prevent potentially deleterious inflammatory effects during sepsis-related ALI by modulating abnormal PMN behaviours. METHODS C57BL/6 wild-type mice were subjected to a caecal ligation and puncture (CLP) mouse model to mimic sepsis in vivo, and M2-Exos were administered intraperitoneally 1 h after CLP. H&E staining, immunofluorescence and immunohistochemistry were conducted to investigate lung tissue injury, PMN infiltration and NET formation in the lung. We further demonstrated the role of M2-Exos on PMN function and explored the potential mechanisms through an in vitro coculture experiment using PMNs isolated from both healthy volunteers and septic patients. RESULTS Here, we report that M2-Exos inhibited PMN migration and NET formation, alleviated lung injury and reduced mortality in a sepsis mouse model. In vitro, M2-Exos significantly decreased PMN migration and NET formation capacity, leading to lipid mediator class switching from proinflammatory leukotriene B4 (LTB4) to anti-inflammatory lipoxin A4 (LXA4) by upregulating 15-lipoxygenase (15-LO) expression in PMNs. Treatment with LXA4 receptor antagonist attenuated the effect of M2-Exos on PMNs and lung injury. Mechanistically, prostaglandin E2 (PGE2) enriched in M2-Exos was necessary to increase 15-LO expression in PMNs by functioning on the EP4 receptor, upregulate LXA4 production to downregulate chemokine (C-X-C motif) receptor 2 (CXCR2) and reactive oxygen species (ROS) expressions, and finally inhibit PMN function. CONCLUSIONS Our findings reveal a previously unknown role of M2-Exos in regulating PMN migration and NET formation through lipid mediator class switching, thus highlighting the potential application of M2-Exos in controlling PMN-mediated tissue injury in patients with sepsis.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Ti Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mei Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Luyang Zhou
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Mengzhi Qi
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xin Xie
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xueyin Shi
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
11
|
Icariin Alleviates Escherichia coli Lipopolysaccharide-Mediated Endometritis in Mice by Inhibiting Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:ijms231810219. [PMID: 36142129 PMCID: PMC9499631 DOI: 10.3390/ijms231810219] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Icariin (ICA) is a naturally occurring phytochemical agent primarily extracted from Epimedium Brevicornum Maxim (Family Berberidaceae) with a broad spectrum of bioactivities. Endometritis is a uterine disease that causes enormous losses in the dairy industry worldwide. In this study, anti-inflammatory and anti-oxidant properties of ICA were investigated against lipopolysaccharide (LPS)-induced endometritis in mice to investigate possible underlying molecular mechanisms. Sixty heathy female Kunming mice were randomly assigned to four groups (n = 15), namely control, LPS, LPS + ICA, and ICA groups. The endometritis was induced by intrauterine infusion of 50 µL of LPS (1 mg/mL). After 24 h of onset of LPS-induced endometritis, ICA groups were injected thrice by ICA intraperitoneally six hours apart. Histopathological examination, enzyme linked immunosorbent assay (ELISA), real time quantitative polymerase chain reaction (RT-qPCR), western blotting, and immunohistochemistry were used in this study. Histological alterations revealed that ICA markedly mitigated uterine tissue injury caused by LPS. The results showed that the ICA inhibited the production of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) and boosted the production of anti-inflammatory cytokines (IL-10). Additionally, ICA modulated the expression of malondialdehyde (MDA), reactive oxygen species (ROS), superoxide dismutase 1 (SOD1), catalase (CAT), and glutathione peroxidase 1 (Gpx1) induced by LPS. The administration of ICA significantly (p < 0.05) improved the mRNA and protein expression of Toll-like receptor (TLR) 4. The western blotting and ELISA finding revealed that the ICA repressed LPS-triggered NF-κB pathway activation. Moreover, ICA improved the antioxidant defense system via activation of the Nrf2 pathway. The results revealed that ICA up-regulated the mRNA and protein expression of Nuclear erythroid-2-related factor (Nrf2), NAD(P)H: quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1), and glutamate-cysteine ligase catalytic subunit (GCLC) under LPS exposure. Conclusively, our findings strongly suggested that ICA protects endometritis caused by LPS by suppressing TLR4-associated NF-κB and Nrf2 pathways. Altogether, these innovative findings may pave the way for future studies into the therapeutic application of ICA to protect humans and animals against endometritis.
Collapse
|
12
|
Fang CN, Tan HQ, Song AB, Jiang N, Liu QR, Song T. NGF/TrkA promotes the vitality, migration and adhesion of bone marrow stromal cells in hypoxia by regulating the Nrf2 pathway. Metab Brain Dis 2022; 37:2017-2026. [PMID: 35579787 DOI: 10.1007/s11011-022-00974-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Bone marrow stromal cells (BMSCs) transplantation is a treatment strategy for ischemic stroke (IS) with great potential. However, the vitality, migration and adhesion of BMSCs are greatly impaired due to the harsh environment of the ischemic area, which affects the therapeutic effects. Herein, we aimed to investigate the roles of nerve growth factor (NGF) in regulating cell behaviors of BMSCs in IS. METHODS The mRNA and protein expressions were assessed using qRT-PCR and western blot, respectively. To simulate ischemic-like conditions in vitro, Brain microvascular (bEnd.3) cells were exposed to oxygen and glucose deprivation (OGD). Cell viability and cell proliferation were evaluated by MTT assay and BrdU assay, respectively. Transwell migration and cell adhesion assays were carried out to determine cell migration and adhesion of BMSCs, respectively, coupled with flow cytometry to evaluate cell apoptosis of bEnd.3 cells. Finally, angiogenesis assay was performed to assess the angiogenesis ability of bEnd.3 cells. RESULTS NGF overexpression resulted in increased cell vitality, adhesion and migration of BMSCs, while NGF knockdown presented the opposite effects. We subsequently discovered that TrkA was a receptor for NGF, and TrkA knockdown significantly inhibited the cell viability, migration and adhesion of BMSCs. Besides, Nrf2 was confirmed as the downstream target of NGF/TrkA to promote the viability, adhesion and migration of BMSC cells. Finally, NGF-silenced BMSCs could not effectively restore the OGD-induced brain microvascular cell damage. CONCLUSIONS NGF/TrkA promoted the viability, migration and adhesion of BMSCs in IS via activating Nrf2 pathway.
Collapse
Affiliation(s)
- Cui-Ni Fang
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China
| | - Hai-Qun Tan
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China
| | - Ao-Bo Song
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China
| | - Ni Jiang
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China
| | - Qian-Rong Liu
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China
| | - Tao Song
- Department of Rehabilitation, Hunan Provincial People's Hospital (the first-affiliated Hospital of Hunan normal University), No.89, Guhan Road, Furong District, 410000, Changsha, Hunan Province, P.R. China.
| |
Collapse
|
13
|
Pro- and anti-inflammatory bioactive lipids imbalance contributes to the pathobiology of autoimmune diseases. Eur J Clin Nutr 2022:10.1038/s41430-022-01173-8. [PMID: 35701524 DOI: 10.1038/s41430-022-01173-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/22/2022] [Accepted: 05/26/2022] [Indexed: 12/27/2022]
Abstract
Autoimmune diseases are driven by TH17 cells that secrete pro-inflammatory cytokines, especially IL-17. Under normal physiological conditions, autoreactive T cells are suppressed by TGF-β and IL-10 secreted by microglia and dendritic cells. When this balance is upset due to injury, infection and other causes, leukocyte recruitment and macrophage activation occurs resulting in secretion of pro-inflammatory IL-6, TNF-α, IL-17 and PGE2, LTs (leukotrienes) accompanied by a deficiency of anti-inflammatory LXA4, resolvins, protecting, and maresins. PGE2 facilitates TH1 cell differentiation and promotes immune-mediated inflammation through TH17 expansion. There is evidence to suggest that autoimmune diseases can be suppressed by anti-inflammatory bioactive lipids LXA4, resolvins, protecting, and maresins. These results imply that systemic and/or local application of LXA4, resolvins, protecting, and maresins and administration of their precursors AA/EPA/DHA could form a potential therapeutic approach in the prevention and treatment of autoimmune diseases.
Collapse
|
14
|
Shokry AA, El-Shiekh RA, Kamel G, Bakr AF, Ramadan A. Bioactive phenolics fraction of Hedera helix L. (Common Ivy Leaf) standardized extract ameliorates LPS-induced acute lung injury in the mouse model through the inhibition of proinflammatory cytokines and oxidative stress. Heliyon 2022; 8:e09477. [PMID: 35647334 PMCID: PMC9130539 DOI: 10.1016/j.heliyon.2022.e09477] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/06/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022] Open
Abstract
Hedera helix L. (family Araliaceae) is classified as a conventional plant used as a medicinal product in the cure and prevention of upper respiratory tract inflammation and infection due to its secretolytic and broncholytic effects. Our research was conducted to authenticate the anti-inflammatory effect of ivy leaves extract in the prevention of acute lung injury (ALI) caused by intranasal administration of lipopolysaccharides (LPS). In-vitro antimicrobial, anti-inflammatory, and anti-oxidant were evaluated, in addition to the in-vivo acute lung inflammation model induced by LPS in mice. The animals were divided into seven groups randomly (each group containing 10 mice): control negative (saline only), control positive (LPS group), standard (Dexamethasone 2 mg/kg), ethanolic ivy leaves extract (EIE, 100 mg/kg), ethanolic ivy leaves extract (EIE, 200 mg/kg), saponin rich fraction (SRF, 100 mg/kg) and phenolic rich fraction (PRF, 100 mg/kg). Right lungs were homogenized to determine the levels of SOD, MDA, catalase, IL-10, TNF-α, NO, IL-1β, IL-6, PGE2, and MPO. Left lungs were excised for histopathology and histomorphometry. Immunohistochemistry of Cox-2 and TNF-α levels were measured. Additionally, Western blotting was used to determine the levels of phosphorylated MAPK. Also, the ethanolic extract was also standardized through HPLC analysis for its content of rutin.The data showed that the oral supplementation with EIE, 200 mg/kg significantly (P < 0.05) decreased the pro-inflammatory mediators, and oxidative stress biomarkers induced by LPS. Interestingly, the phenolics showed promising activity, therefore they are responsible for the action. In conclusion, the standardized ivy leaf extract could be advised for acute lung injury for its antimicrobial, anti-oxidant, and anti-inflammatory activities. Ivy leaf is a traditional perennial edible herb used as an anti-inflammatory agent for respiratory disorders. The plant significantly reduced the serum oxidative stress biomarkers and inflammatory cytokines in the in-vivo acute lung inflammation model induced by LPS. Also, it had antimicrobial activity. Phenolics not saponins are responsible for the activity of the plant.
Collapse
Affiliation(s)
- Aya A. Shokry
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
- Corresponding author.
| | - Riham A. El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Gehan Kamel
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Alaa F. Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Amer Ramadan
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
- Corresponding author.
| |
Collapse
|
15
|
Telocytes reduce oxidative stress by downregulating DUOX2 expression in inflamed lungs of mice. Acta Biochim Biophys Sin (Shanghai) 2022; 54:574-582. [PMID: 35607956 PMCID: PMC9828416 DOI: 10.3724/abbs.2022017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Telocytes (TCs), a novel type of interstitial cells, have been found to participate in tissue protection and repair. In this study, we investigated the antioxidative effects of TCs in inflamed lungs of mice. Acute respiratory distress syndrome (ARDS) mice were used as models of inflamed lungs of mice. Gene sequencing was used to screen the differentially expressed miRNAs in TCs after lipopolysaccharide (LPS) stimulation. AntagomiR-146a-5p-pretreated TCs were first injected into mice, and antioxidant activity of TCs was estimated. TCs, RAW264.7 cells, and MLE-12 cells were collected for the detection of expressions of NOX1-4, DUOX1-2, SOD1-3, GPX1-2, CAT, Nrf2, miR-146a-5p, and miR-21a-3p after LPS stimulation. Silencing miRNAs were delivered to examine the involved signaling pathways. Oxidative stress was examined by measuring malondialdehyde (MDA) levels. We found that microRNA-146a-5p and microRNA-21a-3p were upregulated in TCs after LPS stimulation. ARDS mice that were preinfused with TCs had lower lung tissue injury scores, lung wet-dry ratios, white blood cell counts in alveolar lavage fluid and lower MDA concentrations in lung tissue. However, in antagomiR-146a-5p-pretreated ARDS mice, the infusion of TCs caused no corresponding changes. After LPS stimulation, DUOX2 and MDA concentrations were downregulated in TCs, while DUOX2 was restored by antagomiR-146a-5p in TCs. Dual-luciferase reporter assay confirmed that CREB1 was downregulated by miR-146a-5p, while DUOX2 was downregulated by CREB1, which was confirmed by treating TCs with a specific CREB1 inhibitor. This study demonstrates that LPS stimulation upregulates miR-146a-5p in TCs, which downregulates the CREB1/DUOX2 pathway, resulting in a decrease in oxidative stress in cultured TCs. TCs reduce LPS-induced oxidative stress by decreasing DUOX2 in inflamed lungs of mice.
Collapse
|
16
|
Rubusoside relieves lipopolysaccharide-induced acute lung injury via modulating inflammatory responses: in vitro and in vivo studies. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-021-02246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Xiao J, Yang Q, Zhang Y, Xu H, Ye Y, Li L, Yang Y, Jin S. Maresin conjugates in tissue regeneration-1 suppresses ferroptosis in septic acute kidney injury. Cell Biosci 2021; 11:221. [PMID: 34961563 PMCID: PMC8711186 DOI: 10.1186/s13578-021-00734-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Background Ferroptosis is unique among different types of regulated cell death and closely related to organ injury. Whether ferroptosis occurs in sepsis-associated acute kidney injury (SA-AKI) is not clear. Nuclear factor-erythroid-2-related factor 2 (Nrf2) is crucial to the regulation of ferroptosis. We and others have shown that Maresin conjugates in tissue regeneration 1 (MCTR1) or other members of specialized pro-resolving mediators (SPMs) can actively regulate inflammation resolution and protect organs against injury in inflammatory diseases by activating the Nrf2 signaling. The aim of this study was to determine whether ferroptosis occurs in SA-AKI. Furthermore, we investigated the potential role and mechanism of MCTR1 in the regulation of ferroptosis in SA-AKI, which mainly focus on the Nrf2 signaling. Results We demonstrated for the first time that ferroptosis is present in SA-AKI. Moreover, MCTR1 effectively suppressed ferroptosis in SA-AKI. Meanwhile, MCTR1 upregulated the expression of Nrf2 in the kidney of septic mice. Nrf2 inhibitor ML-385 reversed MCTR1-regulated ferroptosis and AKI, implying that Nrf2 is involved in the inhibitory effects of MCTR1 on ferroptosis in SA-AKI. Further, MCTR1 inhibited ferroptosis and elevated the expression of Nrf2 in LPS-induced HK-2 cells. However, Nrf2 siRNA offset the effect of MCTR1 on ferroptosis. Finally, we observed that MCTR1 ameliorates multi-organ injury and improves survival in animal models of sepsis. Conclusions These data demonstrate that MCTR1 suppresses ferroptosis in SA-AKI through the Nrf2 signaling. Our study enriches the pathophysiological mechanism of SA-AKI and provides new therapeutic ideas and potential intervention targets for SA-AKI. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00734-x.
Collapse
Affiliation(s)
- Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qian Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ye'an Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Haoran Xu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yang Ye
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Linchao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
18
|
Essential Fatty Acids and Their Metabolites in the Pathobiology of Inflammation and Its Resolution. Biomolecules 2021; 11:biom11121873. [PMID: 34944517 PMCID: PMC8699107 DOI: 10.3390/biom11121873] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022] Open
Abstract
Arachidonic acid (AA) metabolism is critical in the initiation and resolution of inflammation. Prostaglandin E2 (PGE2) and leukotriene B4/D4/E4 (LTB4/LD4/LTE4), derived from AA, are involved in the initiation of inflammation and regulation of immune response, hematopoiesis, and M1 (pro-inflammatory) macrophage facilitation. Paradoxically, PGE2 suppresses interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) production and triggers the production of lipoxin A4 (LXA4) from AA to initiate inflammation resolution process and augment regeneration of tissues. LXA4 suppresses PGE2 and LTs' synthesis and action and facilitates M2 macrophage generation to resolve inflammation. AA inactivates enveloped viruses including SARS-CoV-2. Macrophages, NK cells, T cells, and other immunocytes release AA and other bioactive lipids to produce their anti-microbial actions. AA, PGE2, and LXA4 have cytoprotective actions, regulate nitric oxide generation, and are critical to maintain cell shape and control cell motility and phagocytosis, and inflammation, immunity, and anti-microbial actions. Hence, it is proposed that AA plays a crucial role in the pathobiology of ischemia/reperfusion injury, sepsis, COVID-19, and other critical illnesses, implying that its (AA) administration may be of significant benefit in the prevention and amelioration of these diseases.
Collapse
|
19
|
Xiao R, Luo S, Zhang T, Lv Y, Wang T, Zhang J, Su Y, Ruiz M, Dupuis J, Zhu L, Hu Q. Peptide Blocking Self-Polymerization of Extracellular Calcium-Sensing Receptor Attenuates Hypoxia-Induced Pulmonary Hypertension. Hypertension 2021; 78:1605-1616. [PMID: 34565182 DOI: 10.1161/hypertensionaha.120.16712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rui Xiao
- From the Department of Pathophysiology, School of Basic Medicine (R.X., S.L., T.Z., L.Z., Q.H.).,Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.)
| | - Shengquan Luo
- From the Department of Pathophysiology, School of Basic Medicine (R.X., S.L., T.Z., L.Z., Q.H.).,Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.)
| | - Ting Zhang
- From the Department of Pathophysiology, School of Basic Medicine (R.X., S.L., T.Z., L.Z., Q.H.).,Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.)
| | - Yankai Lv
- Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.).,Department of Pathology (Y.L.), Tongji Hospital
| | - Tao Wang
- Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.).,Department of Respiratory and Critical Care Medicine (T.W.), Tongji Hospital
| | | | - Yuan Su
- Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.).,Department of Respiratory and Critical Care Medicine, Union Hospital (Y.S.)
| | - Matthieu Ruiz
- Tongji Medical College, Huazhong University of Science and Technology (HUST), China; Department of Nutrition, Université de Montréal, Canada (M.R.).,Montreal Heart Institute, Canada (M.R., J.D.)
| | - Jocelyn Dupuis
- Montreal Heart Institute, Canada (M.R., J.D.).,Department of medicine, Université de Montréal, Canada (J.D.)
| | - Liping Zhu
- From the Department of Pathophysiology, School of Basic Medicine (R.X., S.L., T.Z., L.Z., Q.H.).,Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.)
| | - Qinghua Hu
- From the Department of Pathophysiology, School of Basic Medicine (R.X., S.L., T.Z., L.Z., Q.H.).,Key Laboratory of Pulmonary Diseases of Ministry of Health (R.X., S.L., T.Z., Y.L., T.W., J.Z., Y.S., L.Z., Q.H.)
| |
Collapse
|
20
|
Sharma A, Chakraborty A, Jaganathan BG. Review of the potential of mesenchymal stem cells for the treatment of infectious diseases. World J Stem Cells 2021; 13:568-593. [PMID: 34249228 PMCID: PMC8246252 DOI: 10.4252/wjsc.v13.i6.568] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The therapeutic value of mesenchymal stem cells (MSCs) for the treatment of infectious diseases and the repair of disease-induced tissue damage has been explored extensively. MSCs inhibit inflammation, reduce pathogen load and tissue damage encountered during infectious diseases through the secretion of antimicrobial factors for pathogen clearance and they phagocytose certain bacteria themselves. MSCs dampen tissue damage during infection by downregulating the levels of pro-inflammatory cytokines, and inhibiting the excessive recruitment of neutrophils and proliferation of T cells at the site of injury. MSCs aid in the regeneration of damaged tissue by differentiating into the damaged cell types or by releasing paracrine factors that direct tissue regeneration, differentiation, and wound healing. In this review, we discuss in detail the various mechanisms by which MSCs help combat pathogens, tissue damage associated with infectious diseases, and challenges in utilizing MSCs for therapy.
Collapse
Affiliation(s)
- Amit Sharma
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Anuja Chakraborty
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Bithiah Grace Jaganathan
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
21
|
Yao L, Tang Y, Chen J, Li J, Wang H, Lu M, Gao L, Liu F, Chang P, Liu X, Tang H. Impaired airway epithelial barrier integrity was mediated by PI3Kδ in a mouse model of lipopolysaccharide-induced acute lung injury. Int Immunopharmacol 2021; 95:107570. [PMID: 33773208 DOI: 10.1016/j.intimp.2021.107570] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 01/03/2023]
Abstract
Cell-cell junctions are critical for the maintenance of cellular as well as tissue polarity and integrity. Dysfunction of airway epithelial barrier has been shown to be involved in the pathogenesis of acute lung injury (ALI). Yet the role of phosphatidylinositol 3-kinase delta (PI3Kδ) in dysregulation of airway epithelial barrier integrity in ALI has not been addressed. Mice were subjected to intratracheal instillation of lipopolysaccharide (LPS) to generate a ALI model. Two pharmacological inhibitors of PI3Kδ, IC87114 and AMG319, were respectively given to the mice. Expression of p110δ and its downstream substrate phospho-AKT (Ser473) was increased in LPS-exposed lungs. These increases were inhibited by IC87114 or AMG319. LPS led to pronounced lung injury that was accompanied by significant airway neutrophil recruitment and bronchial epithelial morphological alterations 72 h after exposure. We also found compromised expression of adherens junction protein E-cadherin and tight junction protein claudin-2 in the airway epithelial cells. Treatment with either IC87114 or AMG319 not only attenuated LPS-induced edema, lung injury and neutrophilc inflammation, reduced total protein concentration and IL-6, TNF-α secretion in BALF, but also restored epithelial E-cadherin and claudin-2 expression. In summary, our results showed that LPS can induce a delayed effect on airway epithelial barrier integrity that is mediated by PI3Kδ in a mouse model of ALI.
Collapse
Affiliation(s)
- Lihong Yao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ying Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Lu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lijuan Gao
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xingxing Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Berberine attenuates the inflammatory response by activating the Keap1/Nrf2 signaling pathway in bovine endometrial epithelial cells. Int Immunopharmacol 2021; 96:107738. [PMID: 33984720 DOI: 10.1016/j.intimp.2021.107738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 11/23/2022]
Abstract
Endometritis is a common inflammatory disease that disturbs the rapid development of dairy farming. In the present study, we investigated the anti-inflammatory effects of berberine on the LPS-induced inflammatory response in bovine endometrial epithelial cells (bEECs) and the participation of the Keap1/Nrf2 signaling pathway in this process. Berberine treatment significantly reduced the LPS-induced expression levels of CRP, IL-1β, IL-6, and TNF-α in bEECs. The Nrf2 signaling pathway in these cells was also activated by berberine. We further evaluated the effects of Nrf2 activators and inhibitors on the downregulation of proinflammatory cytokines. The activator of Nrf2 significantly inhibited the production of these proinflammatory cytokines that was induced by LPS. However, an inhibitor of Nrf2 only partially inhibited the anti-inflammatory effects of berberine on the LPS-induced inflammatory response in bEECs. In conclusion, our findings suggest that berberine exerts anti-inflammatory effects partially by activating the Keap1/Nrf2 signaling pathway.
Collapse
|
23
|
Gerardi G, Cavia-Saiz M, Rivero-Pérez MD, González-SanJosé ML, Muñiz P. The protective effects of wine pomace products on the vascular endothelial barrier function. Food Funct 2021; 11:7878-7891. [PMID: 32812564 DOI: 10.1039/d0fo01199a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endothelial dysfunction is associated with cardiovascular diseases and involves a chronic inflammatory process that together with oxidative stress increases the permeability of the vascular endothelium. The aim of this study was to evaluate the role of red and white wine pomace products (rWPPs and wWPPs) in the maintenance of endothelial integrity in hyperglycemia of EA.hy926 endothelial cells. EA.hy926 endothelial cells exposed to hyperglycemia were treated with the in vitro digested fractions of rWPPs and wWPPs. A Real Time Cellular Analysis (RTCA) system was used to evaluate the endothelial monolayer integrity after INF-γ stimulation of pre-treated endothelial cells with the digested fractions. The changes in cell viability, NO, ROS and NOX4 were recorded and actin cytoskeleton and E-cadherin junctions were evaluated by immunofluorescence. All digested fractions prevent the hyperglycemic actions in the cell viability and NO/ROS balance. The inflammatory mediator INF-γ and hyperglycemia caused a decrease in RTCA adhesion of the EA.hy926 endothelial cell monolayer. Pre-treatment with all digested fractions enhanced the EA.hy926 endothelial monolayer integrity and maintained actin cytoskeleton and E-cadherin junctions. These in vitro studies elucidate that the anti-hyperglycemic and anti-inflammatory actions of wine pomace products involve a decrease in ROS production and the stabilization of junction proteins via modulation of VE-cadherin and actin cytoskeleton suggesting a potential prevention of endothelial damage by these natural products.
Collapse
Affiliation(s)
- Gisela Gerardi
- Department of Biotechnology and Food Science, Faculty of Sciences, University of Burgos, Plaza Misael Bañuelos, 09001, Burgos, Spain.
| | - Mónica Cavia-Saiz
- Department of Biotechnology and Food Science, Faculty of Sciences, University of Burgos, Plaza Misael Bañuelos, 09001, Burgos, Spain.
| | - María D Rivero-Pérez
- Department of Biotechnology and Food Science, Faculty of Sciences, University of Burgos, Plaza Misael Bañuelos, 09001, Burgos, Spain.
| | - María L González-SanJosé
- Department of Biotechnology and Food Science, Faculty of Sciences, University of Burgos, Plaza Misael Bañuelos, 09001, Burgos, Spain.
| | - Pilar Muñiz
- Department of Biotechnology and Food Science, Faculty of Sciences, University of Burgos, Plaza Misael Bañuelos, 09001, Burgos, Spain.
| |
Collapse
|
24
|
Ai M, Lin S, Zhang M, Wu T, Yang N, Li Y, Li L. Cirsilineol attenuates LPS-induced inflammation in both in vivo and in vitro models via inhibiting TLR-4/NFkB/IKK signaling pathway. J Biochem Mol Toxicol 2021; 35:e22799. [PMID: 33949057 DOI: 10.1002/jbt.22799] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/25/2021] [Accepted: 04/22/2021] [Indexed: 01/03/2023]
Abstract
The anti-inflammatory activity of cirsilineol in in vivo condition was assessed by measuring the relative organ weight, lung dry/wet weight ratio, protein concentration, and infiltration of inflammatory cells in bronchoalveolar lavage fluid. We estimated the myeloperoxidase activity and levels of cytokines, chemokines, and inflammatory markers to analyze the efficacy of cirsilineol against lipopolysaccharide (LPS)-induced lung inflammation. Furthermore, we quantified the gene expression of NFkB/IKK signaling molecules in cirsilineol-treated and untreated acute lung injury mice to confirm the anti-inflammatory property of cirsilineol. The lung histology was assessed with hematoxylin and eosin staining. Apart from in vivo experiments, in vitro tests with LPS-stimulated RAW 264.7 macrophages were also performed. Cell viability assay was performed in the presence and absence of LPS in RAW 264.7 macrophages to determine the cytotoxic effect of cirsilineol against macrophages. Reverse-transcription polymerase chain reaction (RT-PCR) analysis was done to analyze the gene expression of inflammatory markers in LPS-treated RAW 264.7 macrophages to prove that cirsilineol effectively inhibits inflammation in vitro. The results of our study prove that cirsilineol effectively inhibits inflammation in both in vivo and in vitro conditions. RT-PCR analysis results of NFkB/IKK signaling molecules clearly illustrate that cirsilineol inhibited the expression of NFkB/IKK signaling protein and thereby prevented inflammation in in vivo condition, and it is further confirmed with the results of inflammatory protein expression in vitro model. The lung histopathological studies authentically confirm that cirsilineol potentially prevented the mice from LPS-induced lung inflammation.
Collapse
Affiliation(s)
- Meimei Ai
- Department of Emergency, First Affiliated Hospital of Air Force Military Medical University (Xijing Hospital), Xian, Shaanxi, China
| | - Shusheng Lin
- Department of Intensive Care Unit, Huaian Hospital of Huai'an City, Huai'an, Jiangsu, China
| | - Meixia Zhang
- Department of Nursing, First Affiliated Hospital of Air Force Military Medical University (Xijing Hospital), Xian, Shaanxi, China
| | - Tieli Wu
- Pneumology Department, Jilin Medical College Affiliated Hospital, Jilin, China
| | - Nan Yang
- Day Observation Ward, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Li
- Department of Health Examination, Peace Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Li Li
- Department of Emergency, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China.,Institute of Sepsis, Beijing University of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Yao P, Zhang Z, Cao J. Isorhapontigenin alleviates lipopolysaccharide-induced acute lung injury via modulating Nrf2 signaling. Respir Physiol Neurobiol 2021; 289:103667. [PMID: 33798789 DOI: 10.1016/j.resp.2021.103667] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022]
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2) is involved in mitigating various oxidative stress- and inflammation-induced diseases, including acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Isorhapontigenin (ISO), from the Chinese herb Gnetum cleistostachyum, exhibits antioxidant and anti-inflammatory properties. In this study, we explored the protective effects of ISO in ALI and its underlying molecular mechanisms. ISO significantly mitigated ALI by reducing the lung wet/dry weight ratio, protein concentration in the bronchoalveolar lavage fluid (BALF), and the levels of myeloperoxidase and malondialdehyde. ISO also improved the superoxide dismutase and glutathione activity in vivo. Moreover, ISO effectively ameliorated the changes in IL-1β, IL-6, and TNF-α concentrations in BALF, prevented IκB degradation, and inhibited the phosphorylation of NF-κB p65 subunit in lung tissues; furthermore, it enhanced the nuclear translocation of Nrf2 and inhibited IL-1β, IL-6, TNF-α, iNOS, COX-2, and ROS production in lipopolysaccharide-treated RAW264.7 cells. The protective effects of ISO in ALI were significantly reversed in ML385-treated RAW264.7 cells and the mouse model, indicating its role in Nrf2-activation. In conclusion, ISO effectively ameliorated lipopolysaccharide-induced ALI by reducing inflammation and oxidative stress, primarily through activation of Nrf2 signaling.
Collapse
Affiliation(s)
- Peiyu Yao
- Department of Respiratory and Critical Care, Tianjin Medical University General Hospital, Tianjin, 300052, China; Department of Emergency, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Zhuo Zhang
- Department of Emergency, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Jie Cao
- Department of Respiratory and Critical Care, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
26
|
Veres B, Eros K, Antus C, Kalman N, Fonai F, Jakus PB, Boros E, Hegedus Z, Nagy I, Tretter L, Gallyas F, Sumegi B. Cyclophilin D-dependent mitochondrial permeability transition amplifies inflammatory reprogramming in endotoxemia. FEBS Open Bio 2021; 11:684-704. [PMID: 33471430 PMCID: PMC7931201 DOI: 10.1002/2211-5463.13091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 01/05/2023] Open
Abstract
Microorganisms or LPS (lipopolysaccharide), an outer membrane component of Gram-negative bacteria, can induce a systemic inflammatory response that leads to sepsis, multiple organ dysfunction, and mortality. Here, we investigated the role of cyclophilin D (CypD)-dependent mitochondrial permeability transition (mPT) in the immunosuppressive phase of LPS-induced endotoxic shock. The liver plays an important role in immunity and organ dysfunction; therefore, we used liver RNA sequencing (RNA-seq) data, Ingenuity® Pathway Analysis (IPA ® ) to investigate the complex role of mPT formation in inflammatory reprogramming and disease progression. LPS induced significant changes in the expression of 2844 genes, affecting 179 pathways related to mitochondrial dysfunction, defective oxidative phosphorylation, nitric oxide (NO) and reactive oxygen species (ROS) accumulation, nuclear factor, erythroid 2 like 2 (Nrf2), Toll-like receptors (TLRs), and tumor necrosis factor α receptor (TNFR)-mediated processes in wild-type mice. The disruption of CypD reduced LPS-induced alterations in gene expression and pathways involving TNFRs and TLRs, in addition to improving survival and attenuating oxidative liver damage and the related NO- and ROS-producing pathways. CypD deficiency diminished the suppressive effect of LPS on mitochondrial function, nuclear- and mitochondrial-encoded genes, and mitochondrial DNA (mtDNA) quantity, which could be critical in improving survival. Our data propose that CypD-dependent mPT is an amplifier in inflammatory reprogramming and promotes disease progression. The mortality in human sepsis and shock is associated with mitochondrial dysfunction. Prevention of mPT by CypD disruption reduces inflammatory reprogramming, mitochondrial dysfunction, and lethality; therefore, CypD can be a novel drug target in endotoxic shock and related inflammatory diseases.
Collapse
Affiliation(s)
- Balazs Veres
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Krisztian Eros
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| | - Csenge Antus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Nikoletta Kalman
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Fruzsina Fonai
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Peter Balazs Jakus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Eva Boros
- Institute of BiochemistryBiological Research CentreSzegedHungary
| | - Zoltan Hegedus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- Institute of BiophysicsBiological Research CentreSzegedHungary
| | - Istvan Nagy
- Institute of BiochemistryBiological Research CentreSzegedHungary
- SeqOmics Biotechnology LtdMorahalomHungary
| | - Laszlo Tretter
- Department of Medical BiochemistrySemmelweis UniversityBudapestHungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| |
Collapse
|
27
|
Das UN. "Cell Membrane Theory of Senescence" and the Role of Bioactive Lipids in Aging, and Aging Associated Diseases and Their Therapeutic Implications. Biomolecules 2021; 11:biom11020241. [PMID: 33567774 PMCID: PMC7914625 DOI: 10.3390/biom11020241] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Lipids are an essential constituent of the cell membrane of which polyunsaturated fatty acids (PUFAs) are the most important component. Activation of phospholipase A2 (PLA2) induces the release of PUFAs from the cell membrane that form precursors to both pro- and ant-inflammatory bioactive lipids that participate in several cellular processes. PUFAs GLA (gamma-linolenic acid), DGLA (dihomo-GLA), AA (arachidonic acid), EPA (eicosapentaenoic acid) and DHA (docosahexaenoic acid) are derived from dietary linoleic acid (LA) and alpha-linolenic acid (ALA) by the action of desaturases whose activity declines with age. Consequently, aged cells are deficient in GLA, DGLA, AA, AA, EPA and DHA and their metabolites. LA, ALA, AA, EPA and DHA can also be obtained direct from diet and their deficiency (fatty acids) may indicate malnutrition and deficiency of several minerals, trace elements and vitamins some of which are also much needed co-factors for the normal activity of desaturases. In many instances (patients) the plasma and tissue levels of GLA, DGLA, AA, EPA and DHA are low (as seen in patients with hypertension, type 2 diabetes mellitus) but they do not have deficiency of other nutrients. Hence, it is reasonable to consider that the deficiency of GLA, DGLA, AA, EPA and DHA noted in these conditions are due to the decreased activity of desaturases and elongases. PUFAs stimulate SIRT1 through protein kinase A-dependent activation of SIRT1-PGC1α complex and thus, increase rates of fatty acid oxidation and prevent lipid dysregulation associated with aging. SIRT1 activation prevents aging. Of all the SIRTs, SIRT6 is critical for intermediary metabolism and genomic stability. SIRT6-deficient mice show shortened lifespan, defects in DNA repair and have a high incidence of cancer due to oncogene activation. SIRT6 overexpression lowers LDL and triglyceride level, improves glucose tolerance, and increases lifespan of mice in addition to its anti-inflammatory effects at the transcriptional level. PUFAs and their anti-inflammatory metabolites influence the activity of SIRT6 and other SIRTs and thus, bring about their actions on metabolism, inflammation, and genome maintenance. GLA, DGLA, AA, EPA and DHA and prostaglandin E2 (PGE2), lipoxin A4 (LXA4) (pro- and anti-inflammatory metabolites of AA respectively) activate/suppress various SIRTs (SIRt1 SIRT2, SIRT3, SIRT4, SIRT5, SIRT6), PPAR-γ, PARP, p53, SREBP1, intracellular cAMP content, PKA activity and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1-α). This implies that changes in the metabolism of bioactive lipids as a result of altered activities of desaturases, COX-2 and 5-, 12-, 15-LOX (cyclo-oxygenase and lipoxygenases respectively) may have a critical role in determining cell age and development of several aging associated diseases and genomic stability and gene and oncogene activation. Thus, methods designed to maintain homeostasis of bioactive lipids (GLA, DGLA, AA, EPA, DHA, PGE2, LXA4) may arrest aging process and associated metabolic abnormalities.
Collapse
Affiliation(s)
- Undurti N. Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA 98604, USA; ; Tel.: +508-904-5376
- BioScience Research Centre and Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India
- International Research Centre, Biotechnologies of the third Millennium, ITMO University, 191002 Saint-Petersburg, Russia
| |
Collapse
|
28
|
Croasdell Lucchini A, Gachanja NN, Rossi AG, Dorward DA, Lucas CD. Epithelial Cells and Inflammation in Pulmonary Wound Repair. Cells 2021; 10:339. [PMID: 33562816 PMCID: PMC7914803 DOI: 10.3390/cells10020339] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/15/2021] [Accepted: 01/30/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory diseases are frequently characterised by epithelial injury, airway inflammation, defective tissue repair, and airway remodelling. This may occur in a subacute or chronic context, such as asthma and chronic obstructive pulmonary disease, or occur acutely as in pathogen challenge and acute respiratory distress syndrome (ARDS). Despite the frequent challenge of lung homeostasis, not all pulmonary insults lead to disease. Traditionally thought of as a quiescent organ, emerging evidence highlights that the lung has significant capacity to respond to injury by repairing and replacing damaged cells. This occurs with the appropriate and timely resolution of inflammation and concurrent initiation of tissue repair programmes. Airway epithelial cells are key effectors in lung homeostasis and host defence; continual exposure to pathogens, toxins, and particulate matter challenge homeostasis, requiring robust defence and repair mechanisms. As such, the epithelium is critically involved in the return to homeostasis, orchestrating the resolution of inflammation and initiating tissue repair. This review examines the pivotal role of pulmonary airway epithelial cells in initiating and moderating tissue repair and restitution. We discuss emerging evidence of the interactions between airway epithelial cells and candidate stem or progenitor cells to initiate tissue repair as well as with cells of the innate and adaptive immune systems in driving successful tissue regeneration. Understanding the mechanisms of intercellular communication is rapidly increasing, and a major focus of this review includes the various mediators involved, including growth factors, extracellular vesicles, soluble lipid mediators, cytokines, and chemokines. Understanding these areas will ultimately identify potential cells, mediators, and interactions for therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | | | - Christopher D. Lucas
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh Bioquarter, Edinburgh EH16 4TJ, UK; (A.C.L.); (N.N.G.); (A.G.R.); (D.A.D.)
| |
Collapse
|
29
|
Shi Z, Wang Y, Ye W, Lin Z, Deng T, Zhang T, Zhao J, Tong Y, Shan Y, Chen G. The LipoxinA4 receptor agonist BML-111 ameliorates intestinal disruption following acute pancreatitis through the Nrf2-regulated antioxidant pathway. Free Radic Biol Med 2021; 163:379-391. [PMID: 33383086 DOI: 10.1016/j.freeradbiomed.2020.12.232] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/01/2020] [Accepted: 12/19/2020] [Indexed: 02/08/2023]
Abstract
Acute pancreatitis (AP) is characterized by excessive release of pro-inflammatory cytokines and provokes multiorgan dysfunction. Disruption of the intestinal epithelium often occurs during and following acute pancreatitis and may aggravate systemic organ injuries. Although it has been widely investigated, to date, there is no satisfactory clinical therapy to restore the inflammatory damage. BML-111 is an endogenous lipid mediator that is analogous to LipoxinA4. It has been shown that BML-111 has a stable and potent anti-inflammatory ability. However, it is unclear whether BML-111 is involved in the process of relieving acute pancreatitis and its induced intestinal barrier damage, and the underlying mechanism of this effect. Here, we demonstrated that BML-111 could enhance the expression of E-cadherin, alleviate apoptosis, and mitigate the accumulation of reactive oxygen species in intestinal epithelial cells, thereby contributing to the anti-inflammatory efficacy in vitro and in vivo. Mechanistically, BML-111 upregulates the expression of Nrf2, which is a key regulator of the antioxidant response, and activates its downstream HO-1/NQO-1 pathway to protect against oxidative stress-induced cell death and tissue injury, consequently ameliorating pancreatitis and intestinal epithelium injury. In Nrf2-deficient cell and Nrf2-knockout mouse models, the depletion of Nrf2 blocked BML-111-induced antioxidant effects and thus was unable to exert protective effects in tissue. Taken together, BML-111 attenuated AP-related intestinal injury via an Nrf2-dependent antioxidant mechanism. Targeting this pathway is a potential therapeutic approach for AP-related intestinal injury.
Collapse
Affiliation(s)
- Zhehao Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Wang
- Department of of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Wen Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zixia Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Tuo Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Tan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jungang Zhao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yifan Tong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yunfeng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
30
|
Xiao Q, Cui Y, Zhao Y, Liu L, Wang H, Yang L. Orientin relieves lipopolysaccharide-induced acute lung injury in mice: The involvement of its anti-inflammatory and anti-oxidant properties. Int Immunopharmacol 2021; 90:107189. [PMID: 33214095 DOI: 10.1016/j.intimp.2020.107189] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/14/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Oxidative stress and inflammatory responses are nearly involved in the pathogenesis of various diseases, including acute lung injury (ALI). Orientin (Ori), a flavonoid component extracted from natural plants, displayed anti-inflammatory and antioxidant properties in our previous studies. In the current study, we aimed to investigate the amelioration effect of Ori on lipopolysaccharide (LPS)-induced ALI, and we further explored the potential molecular mechanisms. The present results indicated that Ori effectively alleviated LPS-induced ALI by improving the histological changes of lung; decreasing the lung W/D ratio and protein levels, the release of inflammatory cells and cytokines into the bronchoalveolar lavage fluid (BALF); inhibiting nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and high mobility group box 1 (HMGB1) protein expression; reducing malondialdehyde (MDA) formation and reactive oxygen species (ROS) generation; and increasing the content of glutathione (GSH) and superoxide dismutase (SOD) contents. Moreover, Ori treatment not only significantly suppressed the LPS-induced nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome, and nuclear factor-kappa B (NF-κB) signaling pathway activation, but also obviously restored the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), NAD (P) H: quinone oxidoreductase (NQO1), glutamate-cysteine ligase catalytic (GCLC), and heme oxygenase 1 (HO-1) expression in the lung; all of which are reduced by LPS. Taken together, these data suggested that Ori plays an important role in the protection against ALI by suppressing inflammation and oxidative stress which may be strongly related to the suppression of NLRP3 inflammasome and NF-κB activation, as well as the upregulation of the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Qingfei Xiao
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yan Cui
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yongli Zhao
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
| | - Li Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
| | - Hongyue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Liming Yang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
31
|
Kang GJ, Kim EJ, Lee CH. Therapeutic Effects of Specialized Pro-Resolving Lipids Mediators on Cardiac Fibrosis via NRF2 Activation. Antioxidants (Basel) 2020; 9:antiox9121259. [PMID: 33321955 PMCID: PMC7764646 DOI: 10.3390/antiox9121259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Heart disease is the number one mortality disease in the world. In particular, cardiac fibrosis is considered as a major factor causing myocardial infarction and heart failure. In particular, oxidative stress is a major cause of heart fibrosis. In order to control such oxidative stress, the importance of nuclear factor erythropoietin 2 related factor 2 (NRF2) has recently been highlighted. In this review, we will discuss the activation of NRF2 by docosahexanoic acid (DHA), eicosapentaenoic acid (EPA), and the specialized pro-resolving lipid mediators (SPMs) derived from polyunsaturated lipids, including DHA and EPA. Additionally, we will discuss their effects on cardiac fibrosis via NRF2 activation.
Collapse
Affiliation(s)
- Gyeoung Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (G.J.K.); (E.J.K.)
| | - Eun Ji Kim
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (G.J.K.); (E.J.K.)
- College of Pharmacy, Dongguk University, Seoul 04620, Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Korea
- Correspondence: ; Tel.: +82-31-961-5213
| |
Collapse
|
32
|
Modulatory role of dietary polyunsaturated fatty acids in Nrf2-mediated redox homeostasis. Prog Lipid Res 2020; 80:101066. [DOI: 10.1016/j.plipres.2020.101066] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
|
33
|
Briottet M, Shum M, Urbach V. The Role of Specialized Pro-Resolving Mediators in Cystic Fibrosis Airways Disease. Front Pharmacol 2020; 11:1290. [PMID: 32982730 PMCID: PMC7493015 DOI: 10.3389/fphar.2020.01290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Cystic Fibrosis (CF) is a recessive genetic disease due to mutations of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene encoding the CFTR chloride channel. The ion transport abnormalities related to CFTR mutation generate a dehydrated airway surface liquid (ASL) layer, which is responsible for an altered mucociliary clearance, favors infections and persistent inflammation that lead to progressive lung destruction and respiratory failure. The inflammatory response is normally followed by an active resolution phase to return to tissue homeostasis, which involves specialized pro-resolving mediators (SPMs). SPMs promote resolution of inflammation, clearance of microbes, tissue regeneration and reduce pain, but do not evoke unwanted immunosuppression. The airways of CF patients showed a decreased production of SPMs even in the absence of pathogens. SPMs levels in the airway correlated with CF patients' lung function. The prognosis for CF has greatly improved but there remains a critical need for more effective treatments that prevent excessive inflammation, lung damage, and declining pulmonary function for all CF patients. This review aims to highlight the recent understanding of CF airway inflammation and the possible impact of SPMs on functions that are altered in CF airways.
Collapse
Affiliation(s)
| | | | - Valerie Urbach
- Institut national de la santé et de la recherche médicale (Inserm) U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| |
Collapse
|
34
|
Alcaraz MJ, Ferrándiz ML. Relevance of Nrf2 and heme oxygenase-1 in articular diseases. Free Radic Biol Med 2020; 157:83-93. [PMID: 31830562 DOI: 10.1016/j.freeradbiomed.2019.12.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/04/2019] [Accepted: 12/07/2019] [Indexed: 02/08/2023]
Abstract
Joint conditions pose an important public health problem as they are a leading cause of pain, functional limitation and physical disability. Oxidative stress is related to the pathogenesis of many chronic diseases affecting the joints such as rheumatoid arthritis and osteoarthritis. Cells have developed adaptive protection mechanisms to maintain homeostasis such as nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) which regulates the transcription of many genes involved in redox balance, detoxification, metabolism and inflammation. Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Ample evidence supports the notion that Nrf2 and HO-1 can confer protection against oxidative stress and inflammatory and immune responses in joint tissues. As a consequence, this pathway may control the activation and metabolism of articular cells to play a regulatory role in joint destruction thus offering new opportunities for better treatments. Further studies are necessary to identify improved strategies to regulate Nrf2 and HO-1 activation in order to enable the development of drugs with therapeutic applications in joint diseases.
Collapse
Affiliation(s)
- Maria José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100, Burjasot, Valencia, Spain.
| | - María Luisa Ferrándiz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100, Burjasot, Valencia, Spain
| |
Collapse
|
35
|
Jaén RI, Fernández-Velasco M, Terrón V, Sánchez-García S, Zaragoza C, Canales-Bueno N, Val-Blasco A, Vallejo-Cremades MT, Boscá L, Prieto P. BML-111 treatment prevents cardiac apoptosis and oxidative stress in a mouse model of autoimmune myocarditis. FASEB J 2020; 34:10531-10546. [PMID: 32543747 DOI: 10.1096/fj.202000611r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 02/05/2023]
Abstract
Myocarditis is an inflammation of the myocardium that can progress to a more severe phenotype of dilated cardiomyopathy (DCM). Three main harmful factors determine this progression: inflammation, cell death, and oxidative stress. Lipoxins and their derivatives are endogenous proresolving mediators that induce the resolution of the inflammatory process. This study aims to determine whether these mediators play a protective role in a murine model of experimental autoimmune myocarditis (EAM) by treating with the lipoxin A4 analog BML-111. We observed that EAM mice presented extensive infiltration areas that correlated with higher levels of inflammatory and cardiac damage markers. Both parameters were significantly reduced in BML-treated EAM mice. Consistently, cardiac dysfunction, hypertrophy, and emerging fibrosis detected in EAM mice was prevented by BML-111 treatment. At the molecular level, we demonstrated that treatment with BML-111 hampered apoptosis and oxidative stress induction by EAM. Moreover, both in vivo and in vitro studies revealed that these beneficial effects were mediated by activation of Nrf2 pathway through CaMKK2-AMPKα kinase pathway. Altogether, our data indicate that treatment with the lipoxin derivative BML-111 effectively alleviates EAM outcome and prevents cardiac dysfunction, thus, underscoring the therapeutic potential of lipoxins and their derivatives to treat myocarditis and other inflammatory cardiovascular diseases.
Collapse
Affiliation(s)
- Rafael I Jaén
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain
- CIBER de enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Fernández-Velasco
- CIBER de enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigación, Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Verónica Terrón
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain
- Instituto de Investigación, Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain
| | - Carlos Zaragoza
- CIBER de enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Servicio de cardiología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación sanitaria (IRYCIS)/Universidad Francisco de Vitoria, Madrid, Spain
| | | | - Almudena Val-Blasco
- Instituto de Investigación, Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - María Teresa Vallejo-Cremades
- Instituto de Investigación, Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Unidad de Imagen e inmunohistoquímica de la Fundación para la Investigación Biomédica del Hospital Universitario La Paz, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain
- CIBER de enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain
- CIBER de enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
36
|
Das UN. Bioactive Lipids as Mediators of the Beneficial Action(s) of Mesenchymal Stem Cells in COVID-19. Aging Dis 2020; 11:746-755. [PMID: 32765941 PMCID: PMC7390526 DOI: 10.14336/ad.2020.0521] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
It is proposed that the beneficial action of mesenchymal stem cells (MSCs) in COVID-19 and other inflammatory diseases could be attributed to their ability to secrete bioactive lipids (BALs) such as prostaglandin E2 (PGE2) and lipoxin A4 (LXA4) and other similar BALs. This implies that MSCs that have limited or low capacity to secrete BALs may be unable to bring about their beneficial actions. This proposal implies that pretreatment of MSCs with BALs enhance their physiological action or improve their (MSCs) anti-inflammatory and disease resolution capacity to a significant degree. Thus, the beneficial action of MSCs reported in the management of COVID-19 could be attributed to their ability to secrete BALs, especially PGE2 and LXA4. Since PGE2, LXA4 and their precursors AA (arachidonic acid), dihomo-gamma-linolenic acid (DGLA) and gamma-linolenic acid (GLA) inhibit the production of pro-inflammatory IL-6 and TNF-α, they could be employed to treat cytokine storm seen in COVID-19, immune check point inhibitory (ICI) therapy, sepsis and ARDS (acute respiratory disease). This is further supported by the observation that GLA, DGLA and AA inactivate enveloped viruses including COVID-19. Thus, infusions of appropriate amounts of GLA, DGLA, AA, PGE2 and LXA4 are of significant therapeutic benefit in COVID-19, ICI therapy and other inflammatory conditions including but not limited to sepsis. AA is the precursor of both PGE2 and LXA4 suggesting that AA is most suited for such preventive and therapeutic approach.
Collapse
Affiliation(s)
- Undurti N Das
- 1UND Life Sciences, Battle Ground, WA 98604, USA.,2BioScience Research Centre and Department of Medicine, Gayatri Vidya Parishad Medical College and Hospital, Visakhapatnam-530048, India
| |
Collapse
|
37
|
Chen Y, Evankovich JW, Lear TB, Tuncer F, Kennerdell JR, Camarco DP, Shishido MS, Liu Y, Chen BB. A small molecule NRF2 activator BC-1901S ameliorates inflammation through DCAF1/NRF2 axis. Redox Biol 2020; 32:101485. [PMID: 32171724 PMCID: PMC7068124 DOI: 10.1016/j.redox.2020.101485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/22/2022] Open
Abstract
NRF2 is a master regulator of cellular anti-oxidant and anti-inflammatory responses, and strategies to augment NRF2-dependent responses may beneficial in many diseases. Basal NRF2 protein level is constrained by constitutive KEAP1-mediated degradation, but in the presence of electrophiles, NRF2 ubiquitination is inhibited. Impeded NRF2 degradation increases NRF2 protein, resulting in up-regulation of anti-oxidant gene transcription, and decreased inflammation. KEAP1-independent mechanisms regulating NRF2 stability have also been reported. Here we employed an HTS approach and identified a small molecule, BC-1901S, that stabilized NRF2 and increased its activity. BC-1901S activated NRF2 by inhibiting NRF2 ubiquitination in a KEAP1-independent manner. It further increased NRF2-dependent anti-oxidant gene transcription, and exhibited anti-inflammatory effects in vitro and in vivo. Further, we identified a new NRF2-interacting partner, DDB1 and CUL4 Associated Factor 1 (DCAF1), an E3 ligase that targeted NRF2 for proteasomal degradation. Mechanistically, BC-1901S directly bound to DCAF1 and disrupted NRF2/DCAF1 interaction, thus activating NRF2. These findings provide new insights in NRF2 biology and NRF2 based anti-inflammatory therapy.
Collapse
Affiliation(s)
- Yanwen Chen
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - John W Evankovich
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Travis B Lear
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ferhan Tuncer
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | | | - Daniel P Camarco
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Morgan S Shishido
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yuan Liu
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Bill B Chen
- Aging Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
38
|
Gao Y, Sun J, Dong C, Zhao M, Hu Y, Jin F. Extracellular Vesicles Derived from Adipose Mesenchymal Stem Cells Alleviate PM2.5-Induced Lung Injury and Pulmonary Fibrosis. Med Sci Monit 2020; 26:e922782. [PMID: 32304204 PMCID: PMC7191958 DOI: 10.12659/msm.922782] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Exposure to PM2.5 (fine particulate matter ≤2.5 μm in aerodynamic diameter) in air increases the risk of lung injury and pulmonary fibrosis (PF). Extracellular vesicles (EVs) derived from adipose mesenchymal stem cells (ADSCs) have been identified as a potential treatment based on the proteins or RNAs delivery and immunomodulatory properties. Here, we assessed the protective effects and mechanisms of ADSCs-EVs on PM2.5-induced lung injury or PF. Material/Methods Rats (male, 6 weeks old) were exposed to PBS or PM2.5 (1.5 mg/kg/day) for 3 days a week for 4 weeks. ADSCs-EVs were extracted by ultracentrifugation. PBS and ADSCs-EVs were administrated through intratracheal instillation. After the end of exposure, the rats were anesthetized and killed. Lung tissues with different treatments were collected for Western blot analysis and HE, IHC, and IF staining analysis. Cells exposed to PM2.5 or “PM2.5+ADSCs-EVs” in vitro were also collected for further Western blotting, qRT-PCR, and IF staining evaluation. Results The results indicated that the initial response of lungs exposed to PM2.5 was lung injury with oxidative stress and inflammation. Long-term PM2.5 exposure resulted in obvious PF in rats. Treatment with ADSCs-EVs decreased PM2.5-induced apoptosis and necrosis in type II alveolar epithelial cells and alleviated lung injury and PF in rats. ADSCs-EVs suppressed reactive oxygen species (ROS) levels and inflammation induced by PM2.5. Furthermore, ADSCs-EVs inhibited TGF-βRI by transferring let-7d-5p and further mitigated PF. Conclusions Our results suggest that EVs derived from ADSCs can alleviate PM2.5-induced lung injury and PF.
Collapse
Affiliation(s)
- Yongheng Gao
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Jinbo Sun
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Chuan Dong
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Mingxuan Zhao
- Research Center of Clinical Pharmacology, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland)
| | - Ying Hu
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Faguang Jin
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
39
|
Zhang T, Hao H, Zhou ZQ, Zeng T, Zhang JM, Zhou XY. Lipoxin A4 inhibited the activation of hepatic stellate cells -T6 cells by modulating profibrotic cytokines and NF-κB signaling pathway. Prostaglandins Other Lipid Mediat 2020; 146:106380. [DOI: 10.1016/j.prostaglandins.2019.106380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022]
|
40
|
Lipoxin A4 Ameliorates Acute Pancreatitis-Associated Acute Lung Injury through the Antioxidative and Anti-Inflammatory Effects of the Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2197017. [PMID: 31781326 PMCID: PMC6875318 DOI: 10.1155/2019/2197017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/08/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023]
Abstract
Acute lung injury (ALI) is a critical event involved in the pathophysiological process of acute pancreatitis (AP). Many methods have been widely used for the treatment of AP-ALI, but few are useful during early inflammation. Lipoxin A4 (LXA4), a potent available anti-inflammatory and novel antioxidant mediator, has been extensively studied in AP-ALI, but its underlying mechanism as a protective mediator is not clear. This research was conducted to identify the possible targets and mechanisms involved in the anti-AP-ALI effect of LXA4. First, we confirmed that LXA4 strongly inhibited AP-ALI in mice. Next, using ELISA, PCR, and fluorescence detection to evaluate different parameters, LXA4 was shown to reduce the inflammatory cytokine production induced by AP and block reactive oxygen species (ROS) generation in vivo and in vitro. In addition, TNF-α treatment activated the nuclear factor E2-related factor 2 (Nrf2) signaling pathway and its downstream gene heme oxygenase-1 (HO-1) in human pulmonary microvascular endothelial cells (HPMECs), and LXA4 further promoted their expression. This study also provided evidence that LXA4 phosphorylates Ser40 and triggers its nuclear translocation to activate Nrf2. Moreover, when Nrf2-knockout (Nrf2−/−) mice and cells were used to further assess the effect of the Nrf2/HO-1 pathway, we found that Nrf2 expression knockdown partially eliminated the effect of LXA4 on the reductions in inflammatory factor levels while abrogating the inhibitory effect of LXA4 on the ROS generation stimulated by AP-ALI. Overall, LXA4 attenuated the resolution of AP-induced inflammation and ROS generation to mitigate ALI, perhaps by modulating the Nrf2/HO-1 pathway. These findings have laid a foundation for the treatment of AP-ALI.
Collapse
|
41
|
Ba F, Zhou X, Zhang Y, Wu C, Xu S, Wu L, Li J, Yin Y, Gu X. Lipoxin A4 ameliorates alveolar fluid clearance disturbance in lipopolysaccharide-induced lung injury via aquaporin 5 and MAPK signaling pathway. J Thorac Dis 2019; 11:3599-3608. [PMID: 31559067 DOI: 10.21037/jtd.2019.08.86] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background A characteristic of acute lung injury (ALI) is the inflammatory damage of alveolar fluid transport. Lipoxins are endogenous lipids involving in the resolution of inflammation. It is found that lipoxin A4 (LXA4) has the distinct properties to improve the anti-edema and pro-resolution function in inflammation. Since aquaporins (AQPs) have essential roles in the integrity of barrier function during fluid transport, especially AQP5 in the maintaining of the epithelium permeability, the current study is aimed to evaluate the potential role of LXA4 in regulating alveolar fluid clearance (AFC) during fluid transport and the corresponding change of AQP5 in the lung. Methods ALI was induced by the lipopolysaccharide (LPS) intraperitoneal injection, and LXA4 treatment was given 8 hours after LPS administration. We investigated changes in the capacity of AFC, pro-inflammatory cytokine concentrations in bronchoalveolar lavage fluid (BALF) and the severity of ALI. Then AQP5 expression in lung tissue and potential regulatory pathways in LPS-induced ALI was explored. Results LXA4 treatment was found to inhibit AFC capacity, inflammatory cytokine release, partially, alleviate ALI severity, and restored AQP5 expression partially. Additionally, we found that LXA4 played a protective role by the inhibition of the phosphorylation of p38 and JNK. Conclusions In summary, our results suggest that LXA4 plays a protective role in lipopolysaccharide-induced ALI by restoring AFC capacity and upregulating AQP5 expression and inhibiting the phosphorylation of p38 and JNK. These findings suggest potential new mechanism of LXA4 as anti-inflammation therapy for the impairment of alveolar fluid transport in ALI.
Collapse
Affiliation(s)
- Fang Ba
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaoming Zhou
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingqi Zhang
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Cen Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shenqian Xu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Liqin Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jiayang Li
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yan Yin
- Institute of Respiratory Disease, First Hospital of China Medical University, Shenyang 110004, China
| | - Xiu Gu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
42
|
Arachidonic Acid Metabolism and Kidney Inflammation. Int J Mol Sci 2019; 20:ijms20153683. [PMID: 31357612 PMCID: PMC6695795 DOI: 10.3390/ijms20153683] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/17/2022] Open
Abstract
As a major component of cell membrane lipids, Arachidonic acid (AA), being a major component of the cell membrane lipid content, is mainly metabolized by three kinds of enzymes: cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes. Based on these three metabolic pathways, AA could be converted into various metabolites that trigger different inflammatory responses. In the kidney, prostaglandins (PG), thromboxane (Tx), leukotrienes (LTs) and hydroxyeicosatetraenoic acids (HETEs) are the major metabolites generated from AA. An increased level of prostaglandins (PGs), TxA2 and leukotriene B4 (LTB4) results in inflammatory damage to the kidney. Moreover, the LTB4-leukotriene B4 receptor 1 (BLT1) axis participates in the acute kidney injury via mediating the recruitment of renal neutrophils. In addition, AA can regulate renal ion transport through 19-hydroxystilbenetetraenoic acid (19-HETE) and 20-HETE, both of which are produced by cytochrome P450 monooxygenase. Epoxyeicosatrienoic acids (EETs) generated by the CYP450 enzyme also plays a paramount role in the kidney damage during the inflammation process. For example, 14 and 15-EET mitigated ischemia/reperfusion-caused renal tubular epithelial cell damage. Many drug candidates that target the AA metabolism pathways are being developed to treat kidney inflammation. These observations support an extraordinary interest in a wide range of studies on drug interventions aiming to control AA metabolism and kidney inflammation.
Collapse
|
43
|
Zhang SD, Wang P, Zhang J, Wang W, Yao LP, Gu CB, Efferth T, Fu YJ. 2'O-galloylhyperin attenuates LPS-induced acute lung injury via up-regulation antioxidation and inhibition of inflammatory responses in vivo. Chem Biol Interact 2019; 304:20-27. [PMID: 30849337 DOI: 10.1016/j.cbi.2019.02.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 01/30/2019] [Accepted: 02/28/2019] [Indexed: 10/27/2022]
Abstract
2'O-galloylhyperin, an active flavonol glycoside compound with remarkable anti-immune activity, was isolated from Pyrola [P. incarnata Fisch.]. However, the evidence of anti-inflammatory activity in pulmonary diseases was still not convincing. The aim of the present study was (1) to investigate the effect of 2'O-galloylhyperin on LPS-induced acute lung injury in mice, and (2) to identify the mechanisms of attenuation of inflammatory responses. The results demonstrated that 2'O-galloylhyperin significantly reduced LPS-induced inflammation damage in a dose-dependent manner. After LPS challenge, treatment with 2'O-galloylhyperin reduced the production of pro-inflammatory cytokines and chemokines, and also improved LPS-induced lung histopathology changes. 2'O-galloylhyperin also increased the activities of antioxidant enzymes, including SOD and GSH-Px to maintain cellular redox homeostasis. Furthermore, 2'O-galloylhyperin inhibited translocation of nuclear factor (NF-κB) activation and suppressed phosphorylation of MAPK signaling pathway consisting of p38, ERK, JNK. In addition, 2'O-galloylhyperin enhanced heme oxygenase-1 (HO-1) expression to block LPS-induced inflammation via activating nuclear factor-crythroid 2-related factor (Nrf2). Moreover, 2'O-galloylhyperin induced adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation. 2'O-galloylhyperin attenuated LPS-induced acute lung injury by inhibiting the MAPK and NF-κB signaling pathways, presumably related to up-regulation of the AMPK and Nrf2 signaling pathways. Furthermore, 2'O-galloylhyperin is a potential protective antioxidant to protect lung tissues from the acute injury.
Collapse
Affiliation(s)
- Sun-Dong Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Peng Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Jing Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Wei Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Li-Ping Yao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Cheng-Bo Gu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China
| | - Thomas Efferth
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128, Mainz, Germany
| | - Yu-Jie Fu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin, 150040, China; College of Forestry, Beijing Forestry University, Beijing, 100083, China.
| |
Collapse
|
44
|
Duan J, Yang Z, Huang J, Zhu Y, Zhao H, Unwith S, Gao X, Lu K, Ning J. Inhibition of tyrosine kinases protects against lipopolysaccharide‐induced acute lung injury by preventing nuclear export of Nrf2. J Cell Biochem 2019; 120:12331-12339. [PMID: 30861161 DOI: 10.1002/jcb.28497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Jiaxiang Duan
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Zhen Yang
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Jian Huang
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Yuan Zhu
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine Chelsea & Westminster Hospital, Imperial College London London UK
| | - Sandeep Unwith
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine Chelsea & Westminster Hospital, Imperial College London London UK
| | - Xian Gao
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Kaizhi Lu
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| | - Jiaolin Ning
- Department of Anesthesia Southwest Hospital, Third Military Medical University Chongqing China
| |
Collapse
|
45
|
Krishnamoorthy N, Abdulnour REE, Walker KH, Engstrom BD, Levy BD. Specialized Proresolving Mediators in Innate and Adaptive Immune Responses in Airway Diseases. Physiol Rev 2018; 98:1335-1370. [PMID: 29717929 DOI: 10.1152/physrev.00026.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Airborne pathogens and environmental stimuli evoke immune responses in the lung. It is critical to health that these responses be controlled to prevent tissue damage and the compromise of organ function. Resolution of inflammation is a dynamic process that is coordinated by biochemical and cellular mechanisms. Recently, specialized proresolving mediators (SPMs) have been identified in resolution exudates. These molecules orchestrate anti-inflammatory and proresolving actions that are cell type specific. In this review, we highlight SPM biosynthesis, the influence of SPMs on the innate and adaptive immune responses in the lung, as well as recent insights from SPMs on inflammatory disease pathophysiology. Uncovering these mediators and cellular mechanisms for resolution is providing new windows into physiology and disease pathogenesis.
Collapse
Affiliation(s)
- Nandini Krishnamoorthy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Raja-Elie E Abdulnour
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Katherine H Walker
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Braden D Engstrom
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
46
|
Liu H, Zhou K, Liao L, Zhang T, Yang M, Sun C. Lipoxin A4 receptor agonist BML-111 induces autophagy in alveolar macrophages and protects from acute lung injury by activating MAPK signaling. Respir Res 2018; 19:243. [PMID: 30518355 PMCID: PMC6282312 DOI: 10.1186/s12931-018-0937-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Background Acute lung injury (ALI) is a life-threatening lung disease where alveolar macrophages (AMs) play a central role both in the early phase to initiate inflammatory responses and in the late phase to promote tissue repair. In this study, we examined whether BML-111, a lipoxin A4 receptor agonist, could alter the phenotypes of AM and thus present prophylactic benefits for ALI. Methods In vitro, isolated AMs were treated with lipopolysaccharide (LPS) to induce ALI. In response to BML-111 pre-treatment, apoptosis and autophagy of AMs were examined by flow cytometry, and by measuring biomarkers for each process. The potential involvement of MAPK1 and mTOR signaling pathway was analyzed. In vivo, an LPS-induced septic ALI model was established in rats and the preventative significance of BML-111 was assessed. On the cellular and molecular levels, the pro-inflammatory cytokines TNF-α and IL-6 from bronchoalveolar lavage were measured by ELISA, and the autophagy in AMs examined using Western blot. Results BML-111 inhibited apoptosis and induced autophagy of AMs in response to ALI inducer, LPS. The enhancement of autophagy was mediated through the suppression of MAPK1 and MAPK8 signaling, but independent of mTOR signaling. In vivo, BML-111 pre-treatment significantly alleviated LPS-induced ALI, which was associated with the reduction of apoptosis, the dampened production of pro-inflammatory cytokines in the lung tissue, as well as the increase of autophagy of AMs. Conclusions This study reveals the prophylactic significance of BML-111 in ALI and the underlying mechanism: by targeting the MAPK signaling but not mTOR pathway, BML-111 stimulates autophagy in AMs, attenuates the LPS-induced cell apoptosis, and promotes the resolution of ALI.
Collapse
Affiliation(s)
- Huaizheng Liu
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Kefu Zhou
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Liangkan Liao
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Tianyi Zhang
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Mingshi Yang
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Chuanzheng Sun
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China.
| |
Collapse
|
47
|
Tian C, Zhang P, Yang J, Zhang Z, Wang H, Guo Y, Liu M. The protective effect of the flavonoid fraction of Abutilon theophrasti Medic. leaves on LPS-induced acute lung injury in mice via the NF-κB and MAPK signalling pathways. Biomed Pharmacother 2018; 109:1024-1031. [PMID: 30551352 DOI: 10.1016/j.biopha.2018.10.197] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 12/22/2022] Open
Abstract
Accompanied by the damages of epithelial and capillary endothelial cell, acute lung injury is diagnosed with the typical pathological symptoms in clinic, including diffusing of pulmonary interstitial, alveolar oedema and hypoxic respiratory insufficiency. Current study focused on the investigation the anti-inflammatory action and mechanisms of total flavonoids extract (TFE) from Abutilon theophrasti Medic. leaves on ALI mice induced by LPSs. Mice were administrated intragastrically with TFE at the concentrations of 0.25, 0.5, or 1.0 g/kg for 5 days, and on last day, nasal administration of LPSs for 6 h after 30 min for intragastric administration of TFE. Pretreatment with TFE not only reduced oxidative damage but also alleviated lung edema in ALI mice. Increased secretion of pro-inflammatory cytokines TNF-α, IL-1β and IL-6, caused by LPSs was reversed by TFE; on the contrary, the anti-inflammatory cytokine IL-10 was upregulated. The proteins expressions of pro-inflammatory mediators iNOS and COX-2 induced by LPSs, were down-regulated by TFE. Moreover, the activation of NF-κB and MAPK signalling pathways was inhibited by TFE in LPSs induced ALI mice. The results revealed that the anti-inflammatory mechanisms of TFE were via inhibition of NF-κB and MAPK activation. Combined, the results suggested that TFE might exert in vivo antioxidant and anti-inflammatory functions in LPSs stimulated mice, and will be potential in adjuvant treatment in oxidative stress and inflammation diseases.
Collapse
Affiliation(s)
- Chunlian Tian
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Peng Zhang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Jie Yang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Zehui Zhang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Hong Wang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Yuru Guo
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China
| | - Mingchun Liu
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe Dist. 110866, Shenyang Liaoning Prov., People's Republic of China.
| |
Collapse
|
48
|
Liang Y, Fan C, Yan X, Lu X, Jiang H, Di S, Ma Z, Feng Y, Zhang Z, Feng P, Feng X, Feng J, Jin F. Berberine ameliorates lipopolysaccharide‐induced acute lung injury via the
PERK
‐mediated
Nrf2/HO‐1
signaling axis. Phytother Res 2018; 33:130-148. [PMID: 30346043 DOI: 10.1002/ptr.6206] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/17/2018] [Accepted: 09/08/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Yuan Liang
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
- Department of Respiration Kunming General Hospital of the People's Liberation Army Kunming China
| | - Chongxi Fan
- Department of Biomedical Engineering The Fourth Military Medical University Xi'an China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Xi Lu
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Hua Jiang
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
- Department of Cardiothoracic Surgery The 97th Hospital of PLA Xuzhou China
| | - Zhengbin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Pan Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Faguang Jin
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| |
Collapse
|
49
|
Zhang JZ, Liu ZL, Zhang YX, Lin HJ, Zhang ZJ. Lipoxin A4 Ameliorates Lipopolysaccharide-Induced A549 Cell Injury through Upregulation of N-myc Downstream-Regulated Gene-1. Chin Med J (Engl) 2018; 131:1342-1348. [PMID: 29786049 PMCID: PMC5987507 DOI: 10.4103/0366-6999.232788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Lipoxin A4 (LXA4) can alleviate lipopolysaccharide (LPS)-induced acute lung injury (ALI) and acute respiratory distress syndrome through promoting epithelial sodium channel (ENaC) expression in lung epithelial cells. However, how LXA4 promote ENaC expression is still largely elusive. The present study aimed to explore genes and signaling pathway involved in regulating ENaC expression induced by LXA4. Methods: A549 cells were incubated with LPS and LXA4, or in combination, and analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) of ENaC-α/γ. Candidate genes affected by LXA4 were explored by transcriptome sequencing of A549 cells. The critical candidate gene was validated by qRT-PCR and Western blot analysis of A549 cells treated with LPS and LXA4 at different concentrations and time intervals. LXA4 receptor (ALX) inhibitor BOC-2 was used to test induction of candidate gene by LXA4. Candidate gene siRNA was adopted to analyze its influence on A549 viability and ENaC-α expression. Phosphoinositide 3-kinase (PI3K) inhibitor LY294002 was utilized to probe whether the PI3K signaling pathway was involved in LXA4 induction of candidate gene expression. Results: The A549 cell models of ALI were constructed and subjected to transcriptome sequencing. Among candidate genes, N-myc downstream-regulated gene-1 (NDRG1) was validated by real-time-PCR and Western blot. NDRG1 mRNA was elevated in a dose-dependent manner of LXA4, whereas BOC-2 antagonized NDRG1 expression induced by LXA4. NDRG1 siRNA suppressed viability of LPS-treated A549 cells (treatment vs. control, 0.605 ± 0.063 vs. 0.878 ± 0.083, P = 0.040) and ENaC-α expression (treatment vs. control, 0.458 ± 0.038 vs. 0.711 ± 0.035, P = 0.008). LY294002 inhibited NDRG1 (treatment vs. control, 0.459 ± 0.023 vs. 0.726 ± 0.020, P = 0.001) and ENaC-α (treatment vs. control, 0.236 ± 0.021 vs. 0.814 ± 0.025, P < 0.001) expressions and serum- and glucocorticoid-inducible kinase 1 phosphorylation (treatment vs. control, 0.442 ± 0.024 vs. 1.046 ± 0.082, P = 0.002), indicating the PI3K signaling pathway was involved in regulating NDRG1 expression induced by LXA4. Conclusion: Our research uncovered a critical role of NDRG1 in LXA4 alleviation of LPS-induced A549 cell injury through mediating PI3K signaling to restore ENaC expression.
Collapse
Affiliation(s)
- Jun-Zhi Zhang
- Department of Anesthesiology, 2nd Clinical Medical College of Jinan University; Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, China
| | - Zhan-Li Liu
- Department of Anesthesiology, 2nd Clinical Medical College of Jinan University; Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, China
| | - Yao-Xian Zhang
- Department of Anesthesiology, 2nd Clinical Medical College of Jinan University; Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, China
| | - Hai-Jiu Lin
- Research and Development Department, Shenzhen Acen Regenerative Medicine, Shenzhen, Guangdong 518122, China
| | - Zhong-Jun Zhang
- Department of Anesthesiology, 2nd Clinical Medical College of Jinan University; Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, China
| |
Collapse
|
50
|
Shi Z, Ye W, Zhang J, Zhang F, Yu D, Yu H, Chen B, Zhou M, Sun H. LipoxinA4 attenuates acute pancreatitis-associated acute lung injury by regulating AQP-5 and MMP-9 expression, anti-apoptosis and PKC/SSeCKS-mediated F-actin activation. Mol Immunol 2018; 103:78-88. [PMID: 30219663 DOI: 10.1016/j.molimm.2018.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 07/20/2018] [Accepted: 09/04/2018] [Indexed: 11/29/2022]
Abstract
An essential component of acute pancreatitis(AP)-induced acute lung injury(ALI) is the inflammation that is part of the body's systemic inflammatory response to a variety of systemic stimuli. Lipoxins(LXs) are considered important endogenous lipids that mediate the resolution of inflammation. In previous studies, we found that Lipoxin A4 (LXA4) reduced AP-induced pulmonary oedema and TNF-α production in lung. However, the underlying mechanism remains unclear. Due to the above studies, we investigated the aquaporin, matrix metalloprotein, apoptosis and PKC/SSeCKS signal pathway in cellular and animal models of AP-associated lung injury following LXA4 intervention. In this study, we first proved LXA4 could effectively promote F-actin reconstruction and regulate its expression in pulmonary microvascular endothelial cells both in vivo and vitro via suppressing PKC/SSeCKS signalling pathway. Next, we found that LXA4 attenuated cell growth inhibition and apoptosis in lung tissues of AP-ALI mice and HPMECs. Additionally, we demonstrated that LXA4 could regulate the expression of AQP-5 and MMP-9 to stabilize the permeability of pulmonary microvascular endothelial cell. In summary, our results suggest that the anti-inflammatory eff ;ects of LXA4 may be due to the inhibition of both the PKC/SSeCKS pathway and apoptosis to reduce alveolar fluid exudation and to the regulation of AQP-5 and MMP-9 expression to maintain the clearance of alveolar fluid. Thus, LXA4 is capable of exerting protective eff ;ects on AP-induced ALI.
Collapse
Affiliation(s)
- Zhehao Shi
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Wen Ye
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Jiecheng Zhang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Fan Zhang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Dinglai Yu
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Huajun Yu
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Bicheng Chen
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, People's Republic of China
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, People's Republic of China.
| | - Hongwei Sun
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|