1
|
King EM, Zhao Y, Moore CM, Steinhart B, Anderson KC, Vestal B, Moore PK, McManus SA, Evans CM, Mould KJ, Redente EF, McCubbrey AL, Janssen WJ. Gpnmb and Spp1 mark a conserved macrophage injury response masking fibrosis-specific programming in the lung. JCI Insight 2024; 9:e182700. [PMID: 39509324 DOI: 10.1172/jci.insight.182700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Macrophages are required for healthy repair of the lungs following injury, but they are also implicated in driving dysregulated repair with fibrosis. How these 2 distinct outcomes of lung injury are mediated by different macrophage subsets is unknown. To assess this, single-cell RNA-Seq was performed on lung macrophages isolated from mice treated with LPS or bleomycin. Macrophages were categorized based on anatomic location (airspace versus interstitium), developmental origin (embryonic versus recruited monocyte derived), time after inflammatory challenge, and injury model. Analysis of the integrated dataset revealed that macrophage subset clustering was driven by macrophage origin and tissue compartment rather than injury model. Gpnmb-expressing recruited macrophages that were enriched for genes typically associated with fibrosis were present in both injury models. Analogous GPNMB-expressing macrophages were identified in datasets from both fibrotic and nonfibrotic lung disease in humans. We conclude that this subset represents a conserved response to tissue injury and is not sufficient to drive fibrosis. Beyond this conserved response, we identified that recruited macrophages failed to gain resident-like programming during fibrotic repair. Overall, fibrotic versus nonfibrotic tissue repair is dictated by dynamic shifts in macrophage subset programming and persistence of recruited macrophages.
Collapse
Affiliation(s)
- Emily M King
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Yifan Zhao
- Center for Genes, Environment, and Health, and
| | | | | | | | | | - Peter K Moore
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Shannon A McManus
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Christopher M Evans
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kara J Mould
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Elizabeth F Redente
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Alexandra L McCubbrey
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - William J Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
2
|
Yu H, Liu S, Wang S, Gu X. The involvement of HDAC3 in the pathogenesis of lung injury and pulmonary fibrosis. Front Immunol 2024; 15:1392145. [PMID: 39391308 PMCID: PMC11464298 DOI: 10.3389/fimmu.2024.1392145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Acute lung injury (ALI) and its severe counterpart, acute respiratory distress syndrome (ARDS), are critical respiratory conditions with high mortality rates due primarily to acute and intense pulmonary inflammation. Despite significant research advances, effective pharmacological treatments for ALI and ARDS remain unavailable, highlighting an urgent need for therapeutic innovation. Notably, idiopathic pulmonary fibrosis (IPF) is a chronic, progressive disease characterized by the irreversible progression of fibrosis, which is initiated by repeated damage to the alveolar epithelium and leads to excessive extracellular matrix deposition. This condition is further complicated by dysregulated tissue repair and fibroblast dysfunction, exacerbating tissue remodeling processes and promoting progression to terminal pulmonary fibrosis. Similar to that noted for ALI and ARDS, treatment options for IPF are currently limited, with no specific drug therapy providing a cure. Histone deacetylase 3 (HDAC3), a notable member of the HDAC family with four splice variants (HD3α, -β, -γ, and -δ), plays multiple roles. HDAC3 regulates gene transcription through histone acetylation and adjusts nonhistone proteins posttranslationally, affecting certain mitochondrial and cytoplasmic proteins. Given its unique structure, HDAC3 impacts various physiological processes, such as inflammation, apoptosis, mitochondrial homeostasis, and macrophage polarization. This article explores the intricate role of HDAC3 in ALI/ARDS and IPF and evaluates its therapeutic potential the treatment of these severe pulmonary conditions.
Collapse
Affiliation(s)
| | | | | | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of
China Medical University, Shenyang, China
| |
Collapse
|
3
|
Guo J, Chen X, Wang C, Ruan F, Xiong Y, Wang L, Abdel-Razek O, Meng Q, Shahbazov R, Cooney RN, Wang G. LIRAGLUTIDE ALLEVIATES ACUTE LUNG INJURY AND MORTALITY IN PNEUMONIA-INDUCED SEPSIS THROUGH REGULATING SURFACTANT PROTEIN EXPRESSION AND SECRETION. Shock 2024; 61:601-610. [PMID: 38150354 PMCID: PMC11009087 DOI: 10.1097/shk.0000000000002285] [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] [Indexed: 12/29/2023]
Abstract
ABSTRACT Glucagon-like peptide 1 (GLP-1) analogs are used to treat type 2 diabetes, and they can regulate insulin secretion, energy homeostasis, inflammation, and immune cell function. This study sought to determine whether the GLP-1 analog liraglutide exerts a beneficial action in an acute lung injury model of pneumonia-induced sepsis. Methods: Wild-type FVB/NJ mice (n = 114) were infected by intratracheal injection with Pseudomonas aeruginosa Xen5 (4 × 10 4 CFU/mouse) or an equal volume (50 μL) of saline (control) with or without a subcutaneous injection of liraglutide (2 mg/kg, 30 min after infection). Mice were killed 24 h after infection. Lung tissues and BALF were analyzed. In separate experiments, the dynamic growth of bacteria and animal mortality was monitored using in vivo imaging system within 48 h after infection. In addition, primary lung alveolar type II cells isolated from mice were used to study the mechanism of liraglutide action. Result: Liraglutide improved survival ( P < 0.05), decreased bacterial loads in vivo , and reduced lung injury scores ( P < 0.01) in septic mice. Liraglutide-treated mice showed decreased levels of inflammatory cells ( P < 0.01) and proinflammatory cytokines (TNF-α and IL-6) ( P < 0.01) in the lung compared with septic controls. Liraglutide significantly increased pulmonary surfactant proteins (SP-A and SP-B) expression/secretion ( P < 0.01) and phospholipid secretion ( P < 0.01) in vivo . Primary alveolar type II cells pretreated with liraglutide improved SP-A and SP-B expression after LPS exposure ( P < 0.01). Conclusion: Liraglutide attenuates mortality and lung inflammation/injury in pneumonia-induced sepsis. The increased surfactant expression/secretion and anti-inflammatory effects of liraglutide represent potential mechanisms by GLP-1 agonists potentiate host defense and maintain alveolar respiratory function in acute lung injury.
Collapse
Affiliation(s)
- Junping Guo
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Rainbowfish Rehabilitation & Nursing School, Hangzhou Vocational & Technical College, Hangzhou 310018, China
| | - Xinghua Chen
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Nephrology, Wuhan University, Renmin Hospital, Wuhan 430060, China
| | - Cole Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Feng Ruan
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Yunhe Xiong
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Lijun Wang
- Department of Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Osama Abdel-Razek
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Qinghe Meng
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Rauf Shahbazov
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Robert N Cooney
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
4
|
Simões JS, Rodrigues RF, Zavan B, Emídio RMP, Soncini R, Boralli VB. Endotoxin-Induced Sepsis on Ceftriaxone-Treated Rats' Ventilatory Mechanics and Pharmacokinetics. Antibiotics (Basel) 2024; 13:83. [PMID: 38247642 PMCID: PMC10812549 DOI: 10.3390/antibiotics13010083] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Sepsis can trigger acute respiratory distress syndrome (ARDS), which can lead to a series of physiological changes, modifying the effectiveness of therapy and culminating in death. For all experiments, male Wistar rats (200-250 g) were split into the following groups: control and sepsis-induced by endotoxin lipopolysaccharide (LPS); the control group received only intraperitoneal saline or saline + CEF while the treated groups received ceftriaxone (CEF) (100 mg/kg) IP; previously or not with sepsis induction by LPS (1 mg/kg) IP. We evaluated respiratory mechanics, and alveolar bronchial lavage was collected for nitrite and vascular endothelial growth factor (VEGF) quantification and cell evaluation. For pharmacokinetic evaluation, two groups received ceftriaxone, one already exposed to LPS. Respiratory mechanics shows a decrease in total airway resistance, dissipation of viscous energy, and elastance of lung tissues in all sepsis-induced groups compared to the control group. VEGF and NOx values were higher in sepsis animals compared to the control group, and ceftriaxone was able to reduce both parameters. The pharmacokinetic parameters for ceftriaxone, such as bioavailability, absorption, and terminal half-life, were smaller in the sepsis-induced group than in the control group since clearance was higher in septic animals. Despite the pharmacokinetic changes, ceftriaxone showed a reduction in resistance in the airways. In addition, CEF lowers nitrite levels in the lungs and acts on their adverse effects, reflecting pharmacological therapy of the disease.
Collapse
Affiliation(s)
- Juliana Savioli Simões
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| | - Rafaela Figueiredo Rodrigues
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| | - Bruno Zavan
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Ricardo Murilo Pereira Emídio
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Roseli Soncini
- Insituto de Ciências da Natureza, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (B.Z.); (R.M.P.E.); (R.S.)
| | - Vanessa Bergamin Boralli
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas 371300-001, Brazil; (J.S.S.); (R.F.R.)
| |
Collapse
|
5
|
Tang Y, Zheng F, Bao X, Zheng Y, Hu X, Lou S, Zhao H, Cui S. Discovery of Highly Selective and Orally Bioavailable PI3Kδ Inhibitors with Anti-Inflammatory Activity for Treatment of Acute Lung Injury. J Med Chem 2023; 66:11905-11926. [PMID: 37606563 DOI: 10.1021/acs.jmedchem.3c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
PI3Kδ is a promising target for the treatment of inflammatory disease; however, the application of PI3Kδ inhibitors in acute respiratory inflammatory diseases is rarely investigated. In this study, through scaffold hopping design, we report a new series of 1H-pyrazolo[3,4-d]pyrimidin-4-amine-tethered 3-methyl-1-aryl-1H-indazoles as highly selective and potent PI3Kδ inhibitors with significant anti-inflammatory activities for treatment of acute lung injury (ALI). There were 29 compounds designed, prepared, and subjected to PI3Kδ inhibitory activity evaluation and anti-inflammatory activity evaluation in macrophages. (S)-29 was identified as a candidate with high PI3Kδ inhibitory activity, isoform selectivity, and high oral bioavailability. The in vivo administration of (S)-29 at 10 mg/kg dosage could significantly ameliorate histopathological changes and attenuate lung inflammation in lung tissues of LPS-challenged mice. Molecular docking demonstrated the success of scaffold hopping design. Overall, (S)-29 is a potent PI3Kδ inhibitor which might be a promising candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Yongmei Tang
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fanli Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Xiaodong Bao
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanan Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Xueping Hu
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao 266237, China
| | - Siyue Lou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Huajun Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Wu X, Jiang Y, Li R, Xia Y, Li F, Zhao M, Li G, Tan X. Ficolin B secreted by alveolar macrophage exosomes exacerbates bleomycin-induced lung injury via ferroptosis through the cGAS-STING signaling pathway. Cell Death Dis 2023; 14:577. [PMID: 37648705 PMCID: PMC10468535 DOI: 10.1038/s41419-023-06104-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023]
Abstract
Pathogenesis exploration and timely intervention of lung injury is quite necessary as it has harmed human health worldwide for years. Ficolin B (Fcn B) is a recognition molecule that can recognize a variety of ligands and play an important role in mediating the cell cycle, immune response, and tissue homeostasis in the lung. However, the role of Fcn B in bleomycin (BLM)-induced lung injury is obscure. This study aims to investigate the sources of Fcn B and its mechanism in BLM-induced lung injury. WT, Fcna-/-, and Fcnb-/- mice were selected to construct the BLM-induced lung injury model. Lung epithelial cells were utilized to construct the BLM-induced cell model. Exosomes that were secreted from alveolar macrophages (AMs) were applied for intervention by transporting Fcn B. Clinical data suggested M-ficolin (homologous of Fcn B) was raised in plasma of interstitial lung disease (ILD) patients. In the mouse model, macrophage-derived Fcn B aggravated BLM-induced lung injury and fibrosis. Fcn B further promoted the development of autophagy and ferroptosis. Remarkably, cell experiment results revealed that Fcn B transported by BLM-induced AMs exosomes accelerated autophagy and ferroptosis in lung epithelial cells through the activation of the cGAS-STING pathway. In contrast, the application of 3-Methyladenine (3-MA) reversed the promotion effect of Fcn B from BLM-induced AMs exosomes on lung epithelial cell damage by inhibiting autophagy-dependent ferroptosis. Meanwhile, in the BLM-induced mice model, the intervention of Fcn B secreted from BLM-induced AMs exosomes facilitated lung injury and fibrosis via ferroptosis. In summary, this study demonstrated that Fcn B transported by exosomes from AMs exacerbated BLM-induced lung injury by promoting lung epithelial cells ferroptosis through the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Xu Wu
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Yixia Jiang
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Rong Li
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yezhou Xia
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Feifan Li
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Meiyun Zhao
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guoqing Li
- Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
- The Key Laboratory of Molecular Diagnosis and Precision Medicine in Hengyang, Hengyang, Hunan, China.
- The Clinical Research Center for Gastric Cancer in Hunan Province, Hengyang, Hunan, China.
| | - Xiaowu Tan
- Pulmonary and Critical Care Medicine, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
7
|
Feng B, Feng X, Yu Y, Xu H, Ye Q, Hu R, Fang X, Gao F, Wu J, Pan Q, Yu J, Lang G, Li L, Cao H. Mesenchymal stem cells shift the pro-inflammatory phenotype of neutrophils to ameliorate acute lung injury. Stem Cell Res Ther 2023; 14:197. [PMID: 37553691 PMCID: PMC10408228 DOI: 10.1186/s13287-023-03438-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/31/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) treatment plays a major role in the management of acute lung injury (ALI), and neutrophils are the initial line of defense against ALI. However, the effect of MSCs on neutrophils in ALI remains mostly unknown. METHODS We investigated the characteristics of neutrophils in lung tissue of ALI mice induced by lipopolysaccharide after treatment with MSCs using single-cell RNA sequencing. Neutrophils separated from lung tissue in ALI were co-cultured with MSCs, and then samples were collected for reverse transcription-polymerase chain reaction and flow cytometry. RESULTS During inflammation, six clusters of neutrophils were identified, annotated as activated, aged, and circulatory neutrophils. Activated neutrophils had higher chemotaxis, reactive oxygen species (ROS) production, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase scores than aged neutrophils. Circulatory neutrophils occurred mainly in healthy tissue and were characterized by higher expression of Cxcr2 and Sell. Activated neutrophils tended to exhibit higher expression of Cxcl10 and Cd47, and lower expression of Cd24a, while aged neutrophils expressed a lower level of Cd47 and higher level of Cd24a. MSC treatment shifted activated neutrophils toward an aged neutrophil phenotype by upregulating the expression of CD24, thereby inhibiting inflammation by reducing chemotaxis, ROS production, and NADPH oxidase. CONCLUSION We identified the immunosuppressive effects of MSCs on the subtype distribution of neutrophils and provided new insight into the therapeutic mechanism of MSC treatment in ALI.
Collapse
Affiliation(s)
- Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Yingduo Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Haoying Xu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Qingqing Ye
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, 79 Qingchun Rd, Hangzhou, 310003, China
| | - Ruitian Hu
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC, 27708, USA
| | - Xinru Fang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Feiqiong Gao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Jian Wu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Guanjing Lang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou, 310003, China.
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China.
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, 79 Qingchun Rd, Hangzhou, 310003, China.
| |
Collapse
|
8
|
Bates JHT, Nieman GF, Kollisch-Singule M, Gaver DP. Ventilator-Induced Lung Injury as a Dynamic Balance Between Epithelial Cell Damage and Recovery. Ann Biomed Eng 2023; 51:1052-1062. [PMID: 37000319 DOI: 10.1007/s10439-023-03186-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/15/2023] [Indexed: 04/01/2023]
Abstract
Acute respiratory distress syndrome (ARDS) has a high mortality rate that is due in part to ventilator-induced lung injury (VILI). Nevertheless, the majority of patients eventually recover, which means that their innate reparative capacities eventually prevail. Since there are currently no medical therapies for ARDS, minimizing its mortality thus amounts to achieving an optimal balance between spontaneous tissue repair versus the generation of VILI. In order to understand this balance better, we developed a mathematical model of the onset and recovery of VILI that incorporates two hypotheses: (1) a novel multi-hit hypothesis of epithelial barrier failure, and (2) a previously articulated rich-get-richer hypothesis of the interaction between atelectrauma and volutrauma. Together, these concepts explain why VILI appears in a normal lung only after an initial latent period of injurious mechanical ventilation. In addition, they provide a mechanistic explanation for the observed synergy between atelectrauma and volutrauma. The model recapitulates the key features of previously published in vitro measurements of barrier function in an epithelial monolayer and in vivo measurements of lung function in mice subjected to injurious mechanical ventilation. This provides a framework for understanding the dynamic balance between factors responsible for the generation of and recovery from VILI.
Collapse
Affiliation(s)
- Jason H T Bates
- Department of Medicine, University of Vermont, Burlington, VT, 05405, USA.
- Department of Medicine, Larner College of Medicine, 149 Beaumont Avenue, Burlington, 05405-0075, USA.
| | - Gary F Nieman
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Donald P Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| |
Collapse
|
9
|
Efficacy of Clinically Used PARP Inhibitors in a Murine Model of Acute Lung Injury. Cells 2022; 11:cells11233789. [PMID: 36497049 PMCID: PMC9738530 DOI: 10.3390/cells11233789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1), as a potential target for the experimental therapy of acute lung injury (ALI), was identified over 20 years ago. However, clinical translation of this concept was not possible due to the lack of clinically useful PARP inhibitors. With the clinical introduction of several novel, ultrapotent PARP inhibitors, the concept of PARP inhibitor repurposing has re-emerged. Here, we evaluated the effect of 5 clinical-stage PARP inhibitors in oxidatively stressed cultured human epithelial cells and monocytes in vitro and demonstrated that all inhibitors (1-30 µM) provide a comparable degree of cytoprotection. Subsequent in vivo studies using a murine model of ALI compared the efficacy of olaparib and rucaparib. Both inhibitors (1-10 mg/kg) provided beneficial effects against lung extravasation and pro-inflammatory mediator production-both in pre- and post-treatment paradigms. The underlying mechanisms include protection against cell dysfunction/necrosis, inhibition of NF-kB and caspase 3 activation, suppression of the NLRP3 inflammasome, and the modulation of pro-inflammatory mediators. Importantly, the efficacy of PARP inhibitors was demonstrated without any potentiation of DNA damage, at least as assessed by the TUNEL method. These results support the concept that clinically approved PARP inhibitors may be repurposable for the experimental therapy of ALI.
Collapse
|
10
|
Liu C, Zhen D, Du H, Gong G, Wu Y, Ma Q, Quan ZS. Synergistic anti-inflammatory effects of peimine, peiminine, and forsythoside a combination on LPS-induced acute lung injury by inhibition of the IL-17-NF-κB/MAPK pathway activation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115343. [PMID: 35533916 DOI: 10.1016/j.jep.2022.115343] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/16/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Forsythia suspensa (Thunb.) Vahl and Fritillaria thunbergii Miq are traditional Chinese medicines that exhibit the ability to clear heat and toxic material effects. In China, the combination of these two medicines is widely used to treat mucopurulent sputum and bloody phlegm, arising due to phlegm-heat obstruction in respiratory diseases. However, very limited information is available regarding the combined anti-inflammatory effect of important effective components of Forsythia suspensa (Thunb.) Vahl and Fritillaria thunbergii Miq, namely peimine, peiminine, and forsythoside A. AIM OF THIS STUDY To investigate synergistic anti-inflammatory effects of combined administration of peimine, peiminine, and forsythoside A on LPS-induced acute lung injury compared to combined administration of two compounds or individual administration, and unravel the underlying mechanism. MATERIAL AND METHODS In the present study, male BALB/c mice received an oral dosage of sodium carboxymethylcellulose (CMC-Na) (0.5%, 1 mL/100 g), peimine, peiminine, forsythoside A, peimine + forsythoside A, peiminine + forsythoside A, and peimine + peiminine + forsythoside A (suspended in CMC-Na; 0.5%), once daily for 7 days. Subsequently, intratracheal instillation of LPS was applied to establish acute lung injury model. After 6 h of administration, the mice were sacrificed, and bronchoalveolar lavage fluid (BALF) and lung tissues were collected. These samples were further used to determine lung W/D (wet/dry) weight ratio, total protein (TP) levels, inflammatory cytokines (IL-6, TNF-α, IL-1β, and IL-17), and expression of proteins involved in TLR4/MAPK/NF-κB pathway and IL-17 pathway. Further, tissue sections were subjected to H&E staining to assess the pathological alterations induced by LPS. The expression of IL-6 and TNF-α proteins in lung tissues was also analyzed using immunohistochemical staining. RESULTS A synergistic anti-inflammatory effect of peimine, peiminine, and forsythoside A was observed when administered in combination to LPS-induced acute lung injury. The combined administration of peimine, peiminine, and forsythoside A had a strongly inhibitory effects on the W/D weight ratio, total protein (TP) level and the inflammatory cytokines (TNF-α, IL-6, IL-1β, and IL-17) level in acute lung injury mice, compared to combined administration of two compounds or individual administration. The infiltration of inflammatory cells and thickened bronchoalveolar walls induced by LPS were also ameliorated through the combined administration of peimine, peiminine, and forsythoside A. More importantly, the upregulation of protein related to TLR4/MAPK/NF-κB signaling pathway and the activation of IL-17 were significantly suppressed by pretreatment with each of the three compounds alone, while the effects of individual compounds were synergistically augmented by the combined pretreatment of these three compounds. CONCLUSION The combined administration of peimine, peiminine, and forsythoside A ameliorated inflammatory response in acute lung injury mice induced by LPS in a synergistic manner, the mechanism may be related to the dampening of the TLR4/MAPK/NF-κB signaling pathway and IL-17 activation.
Collapse
Affiliation(s)
- Chunyan Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Inner Mongolia Autonomous Region, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia, PR China.
| | - Dong Zhen
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Inner Mongolia Autonomous Region, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia, PR China.
| | - Huanhuan Du
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Inner Mongolia Autonomous Region, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia, PR China.
| | - Guohua Gong
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Inner Mongolia Autonomous Region, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia, PR China; Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Yun Wu
- Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Qianqian Ma
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Inner Mongolia Autonomous Region, Tongliao, 028000, China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia, PR China.
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
11
|
Chaudhary S, Rai P, Joshi A, Yadav P, Sesham K, Kumar S, Mridha AR, Baitha U, Nag TC, Soni KD, Trikha A, Yadav SC. Ultracellular Imaging of Bronchoalveolar Lavage from Young COVID-19 Patients with Comorbidities Showed Greater SARS-COV-2 Infection but Lesser Ultrastructural Damage Than the Older Patients. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-25. [PMID: 36065953 DOI: 10.1017/s1431927622012430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this study, we examined the cellular infectivity and ultrastructural changes due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in the various cells of bronchoalveolar fluid (BALF) from intubated patients of different age groups (≥60 years and <60 years) and with common comorbidities such as diabetes, liver and kidney diseases, and malignancies. BALF of 79 patients (38 cases >60 and 41 cases <60 years) were studied by light microscopy, immunofluorescence, scanning, and transmission electron microscopy to evaluate the ultrastructural changes in the ciliated epithelium, type II pneumocytes, macrophages, neutrophils, eosinophils, lymphocytes, and anucleated granulocytes. This study demonstrated relatively a greater infection and better preservation of subcellular structures in these cells from BALF of younger patients (<60 years compared with the older patients (≥60 years). The different cells of BALF from the patients without comorbidities showed higher viral load compared with the patients with comorbidities. Diabetic patients showed maximum ultrastructural damage in BALF cells in the comorbid group. This study highlights the comparative effect of SARS-CoV-2 infection on the different airway and inflammatory cells of BALF at the subcellular levels among older and younger patients and in patients with comorbid conditions.
Collapse
Affiliation(s)
- Shikha Chaudhary
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Preeti Rai
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Arti Joshi
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Pooja Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Kishore Sesham
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Shailendra Kumar
- Department of Anaesthesiology, Pain Medicine and Critical Care, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Asit Ranjan Mridha
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Upendra Baitha
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Tapas Chandra Nag
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Kapil Dev Soni
- Anaesthesia and Critical Care, JPN Apex Trauma Center, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Anjan Trikha
- Department of Anaesthesiology, Pain Medicine and Critical Care, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| | - Subhash Chandra Yadav
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi 110029, India
| |
Collapse
|
12
|
Li X, Wei Y, Li S, Liang J, Liu Z, Cui Y, Gao J, Yang Z, Li L, Zhou H, Chen S, Yang C. Zanubrutinib ameliorates lipopolysaccharide-induced acute lung injury via regulating macrophage polarization. Int Immunopharmacol 2022; 111:109138. [PMID: 35973369 DOI: 10.1016/j.intimp.2022.109138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a disease characterized by pulmonary diffusion dysfunction and its exacerbation stage is acute respiratory distress syndrome (ARDS), which may develop to multiple organ failure and seriously threatens human health. ALI has high mortality rates and few effective treatments, thus effective protection measures for ALI are becoming increasingly important. Macrophages play a key regulatory role in the pathogenesis of ALI, and the degree of macrophage polarization is closely related to the severity and prognosis of ALI. In this study, we evaluated the effects of Zanubrutinib (ZB), a BTK small molecule inhibitor approved by the FDA for the treatment of cell lymphoma, on macrophage polarization and acute lung injury. In the in vivo study, we constructed a mouse model of Lipopolysaccharide (LPS)-induced acute lung injury and found that ZB could improve the acute injury of mouse lungs by inhibiting the secretion of proinflammatory factors and promoting the secretion of anti-inflammatory factors, reduce the number of inflammatory cells in alveolar lavage fluid, and then alleviate the inflammatory response. In vivo and in vitro studies have shown that ZB could inhibit the M1 macrophage polarization and promote the M2 macrophage polarization. Subsequent mechanistic studies revealed that ZB could inhibit the macrophage M1 polarization via targeting BTK activation and inhibiting JAK2/STAT1 and TLR4/MyD88/NF-κB signaling pathways, and promote the macrophage M2 polarization by promoting the activation of STAT6 and PI3K / Akt signaling pathways. In summary, ZB has shown therapeutic effect in LPS-induced acute lung injury in mice, which provides a potential candidate drug to treat acute lung injury.
Collapse
Affiliation(s)
- Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Yuli Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Shimeng Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Jingjing Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Zhichao Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Yunyao Cui
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China
| | - Jingjing Gao
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China
| | - Zhongyi Yang
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, People's Republic of China
| | - Lei Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, People's Republic of China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China.
| | - Shanshan Chen
- The First Affiliated Hospital of Zhengzhou University, People's Republic of China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China.
| |
Collapse
|
13
|
Nguyen N, Xu S, Lam TYW, Liao W, Wong WSF, Ge R. ISM1 suppresses LPS-induced acute lung injury and post-injury lung fibrosis in mice. Mol Med 2022; 28:72. [PMID: 35752760 PMCID: PMC9233842 DOI: 10.1186/s10020-022-00500-w] [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: 01/18/2022] [Accepted: 06/14/2022] [Indexed: 11/15/2022] Open
Abstract
Background Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are clinical syndromes characterized by acute lung inflammation, pulmonary edema and hypoxemia, with up to 50% mortality rate without effective pharmacological therapy. Following the acute inflammation, repair and remodeling occurs which in some cases resulting in lung fibrosis. The pathophysiology of ALI/ARDS remains incompletely understood. Lipopolysaccharide (LPS)-induced ALI in mice have been widely used as a model to study human ALI/ARDS. Isthmin 1 (ISM1) is a secreted protein highly abundant in mouse lung. We have previously reported that upon intratracheal LPS instillation, ISM1 expression in the lung is further upregulated. Recently, we also reported that ISM1 is an anti-inflammatory protein in the lung with Ism1-/- mice presenting spontaneous chronic low-grade lung inflammation and obvious emphysema at young adult stage. However, what role ISM1 plays in ALI/ARDS and lung fibrosis remain unclear. Methods Using Ism1-/- mice and intratracheal LPS-induced ALI, and local delivery of recombinant ISM1 (rISM1), we investigated the role ISM1 plays in ALI and post-ALI lung fibrosis using flow cytometry, Western blot, antibody array, immunohistochemistry (IHC), immunofluorescent and other histological staining. Results We reveal that ISM1 deficiency in mice led to an intensified acute lung inflammation upon intratracheal LPS challenge, with a heightened leukocyte infiltration including neutrophils and monocyte-derived alveolar macrophages, as well as upregulation of multiple pro-inflammatory cytokines/chemokines including tumor necrosis factor α (TNF-α). Although innate immune cells largely subsided to the baseline by day 7 post-LPS challenge in both wild-type and Ism1−/− mice, Ism1−/− lung showed increased post-ALI fibrosis from day 9 post-LPS treatment with increased myofibroblasts, excessive collagen accumulation and TGF-β upregulation. The heightened lung fibrosis remained on day 28 post-LPS. Moreover, intranasal delivered recombinant ISM1 (rISM1) effectively suppressed LPS-induced acute lung inflammation and ALI, and rISM1 suppressed LPS-induced NF-κB activation in cultured mouse alveolar macrophages. Conclusion Together with our previous report, this work further established ISM1 as an endogenous anti-inflammation protein in the lung, restraining excessive host inflammatory response to LPS-triggered ALI and suppressing post-ALI lung fibrosis likely through suppressing NF-κB activation and pro-inflammatory cytokine/chemokine production. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00500-w.
Collapse
Affiliation(s)
- Ngan Nguyen
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Simin Xu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Terence Yin Weng Lam
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Republic of Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore, 138602, Republic of Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Republic of Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore, 138602, Republic of Singapore.,Drug Discovery and Optimization Platform, National University Health System, Singapore, 117600, Republic of Singapore
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore.
| |
Collapse
|
14
|
Sen’kova AV, Savin IA, Brenner EV, Zenkova MA, Markov AV. Core genes involved in the regulation of acute lung injury and their association with COVID-19 and tumor progression: A bioinformatics and experimental study. PLoS One 2021; 16:e0260450. [PMID: 34807957 PMCID: PMC8608348 DOI: 10.1371/journal.pone.0260450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a specific form of lung damage caused by different infectious and non-infectious agents, including SARS-CoV-2, leading to severe respiratory and systemic inflammation. To gain deeper insight into the molecular mechanisms behind ALI and to identify core elements of the regulatory network associated with this pathology, key genes involved in the regulation of the acute lung inflammatory response (Il6, Ccl2, Cat, Serpine1, Eln, Timp1, Ptx3, Socs3) were revealed using comprehensive bioinformatics analysis of whole-genome microarray datasets, functional annotation of differentially expressed genes (DEGs), reconstruction of protein-protein interaction networks and text mining. The bioinformatics data were validated using a murine model of LPS-induced ALI; changes in the gene expression patterns were assessed during ALI progression and prevention by anti-inflammatory therapy with dexamethasone and the semisynthetic triterpenoid soloxolone methyl (SM), two agents with different mechanisms of action. Analysis showed that 7 of 8 revealed ALI-related genes were susceptible to LPS challenge (up-regulation: Il6, Ccl2, Cat, Serpine1, Eln, Timp1, Socs3; down-regulation: Cat) and their expression was reversed by the pre-treatment of mice with both anti-inflammatory agents. Furthermore, ALI-associated nodal genes were analysed with respect to SARS-CoV-2 infection and lung cancers. The overlap with DEGs identified in postmortem lung tissues from COVID-19 patients revealed genes (Saa1, Rsad2, Ifi44, Rtp4, Mmp8) that (a) showed a high degree centrality in the COVID-19-related regulatory network, (b) were up-regulated in murine lungs after LPS administration, and (c) were susceptible to anti-inflammatory therapy. Analysis of ALI-associated key genes using The Cancer Genome Atlas showed their correlation with poor survival in patients with lung neoplasias (Ptx3, Timp1, Serpine1, Plaur). Taken together, a number of key genes playing a core function in the regulation of lung inflammation were found, which can serve both as promising therapeutic targets and molecular markers to control lung ailments, including COVID-19-associated ALI.
Collapse
Affiliation(s)
- Aleksandra V. Sen’kova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Innokenty A. Savin
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenyi V. Brenner
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina A. Zenkova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Andrey V. Markov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
15
|
Li J, Zeng X, Wang W. miR-122-5p downregulation attenuates lipopolysaccharide-induced acute lung injury by targeting IL1RN. Exp Ther Med 2021; 22:1278. [PMID: 34594415 PMCID: PMC8456493 DOI: 10.3892/etm.2021.10713] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRs) and inflammatory cytokines can induce acute lung injury (ALI), which can develop into acute respiratory distress syndrome in severe cases. Previous research has revealed that miR-122-5p participates in the development of ALI, and that its expression is positively associated with ALI. However, the mechanism by which miR-122-5p contributes to ALI remains to be determined. In the current study, TargetScan and dual luciferase reporter gene assays were used to confirm that IL-1 receptor antagonist (IL1RN) was a target of miR-122-5p. Subsequently, by referring to previous literature, a lipopolysaccharide (LPS)-induced ALI cell model was established. A549 cells were transfected with mimic control or miR-122-5p mimics for 24 h, and 10 µg LPS was used to treat the transfected cells for 12 h. The results revealed that miR-122-5p mimics decreased cell viability and promoted apoptosis. Lactate dehydrogenase (LDH) release assays indicated that miR-122-5p mimics increased LDH release. ELISA demonstrated that miR-122-5p mimics promoted TNF-α, IL-1β and IL-6 expression levels. A549 cells were transfected with inhibitor control, miR-122-5p inhibitor, miR-122-5p inhibitor + control-small interfering (si)RNA or miR-122-5p inhibitor + IL1RN-siRNA for 24 h, after which the cells were treated with 10 µg LPS for 12 h. The results revealed that the effects of the miR-122-5p inhibitor were the opposite of those of the miR-122-5p mimic. All the effects of miR-122-5p inhibitor on LPS-treated A549 cells were significantly reversed by IL1RN-siRNA. Overall, the results highlighted miR-122-5p as a potential novel target for the treatment of ALI.
Collapse
Affiliation(s)
- Jie Li
- Department of Pulmonary Disease, Hubei Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430000, P.R. China
| | - Xiaoxia Zeng
- Department of Emergency, Hubei Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430000, P.R. China
| | - Weiqing Wang
- Department of Emergency, Hubei Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
16
|
Li Y, Zou L, Chu L, Ye L, Ni J, Chu X, Guo T, Yang X, Zhu Z. Identification and Integrated Analysis of circRNA and miRNA of Radiation-Induced Lung Injury in a Mouse Model. J Inflamm Res 2021; 14:4421-4431. [PMID: 34511976 PMCID: PMC8422032 DOI: 10.2147/jir.s322736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Radiation-induced lung injury (RILI) is a main threat to patients who received thoracic radiotherapy. Thus, understanding the molecular mechanism of RILI is of great importance. Circular RNAs (circRNAs) have been found to act as a regulator of multiple biological processes, and the circRNA-microRNA (miRNA)-mRNA axis could play an important role in the signaling pathway of many human diseases including radiation injury. Methods First, the circRNA and miRNA of RILI in a mouse model were investigated. The mice received 12 Gy of thoracic irradiation, and the irradiated lung tissues at 48 hours after irradiation were analyzed by RNA sequencing (RNA-seq) compared with normal lung tissues. Then, Gene Ontology analysis of the target mRNAs of the significantly differently expressed circRNAs was performed. Results In the irradiated group, inflammatory changes in lungs were observed; 21 significantly up-regulated and 33 down-regulated significantly miRNAs were identified (p < 0.05). Among 27 differentially expressed circRNAs, 10 were down-regulated and 17 were up-regulated in the irradiated group [log2 (fold change) > 1 or < −1, p<0.05]. These differentially expressed miRNAs took part in a series of cellular processes, such as positive regulation of alpha-beta T-cell proliferation, interstitial matrix, collagen fibril organization, chemokine receptor activity, cellular defense response, and B-cell receptor signaling pathway. The differentially expressed circRNAs were related to Th1 and Th2 differentiation pathways, and the predicted mRNAs were verified. Conclusion This study revealed immune-related molecular pathways play an important role in the early response after radiotherapy. In the future, research on the target mechanism and early intervention of circRNAs with associated miRNAs such as circRNA5229, circRNA544, and circRNA3340, could benefit the treatment of RILI.
Collapse
Affiliation(s)
- Yida Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Luxi Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiao Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
17
|
Kosyreva A, Dzhalilova D, Lokhonina A, Vishnyakova P, Fatkhudinov T. The Role of Macrophages in the Pathogenesis of SARS-CoV-2-Associated Acute Respiratory Distress Syndrome. Front Immunol 2021; 12:682871. [PMID: 34040616 PMCID: PMC8141811 DOI: 10.3389/fimmu.2021.682871] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages are cells that mediate both innate and adaptive immunity reactions, playing a major role in both physiological and pathological processes. Systemic SARS-CoV-2-associated complications include acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation syndrome, edema, and pneumonia. These are predominantly effects of massive macrophage activation that collectively can be defined as macrophage activation syndrome. In this review we focus on the role of macrophages in COVID-19, as pathogenesis of the new coronavirus infection, especially in cases complicated by ARDS, largely depends on macrophage phenotypes and functionalities. We describe participation of monocytes, monocyte-derived and resident lung macrophages in SARS-CoV-2-associated ARDS and discuss possible utility of cell therapies for its treatment, notably the use of reprogrammed macrophages with stable pro- or anti-inflammatory phenotypes.
Collapse
Affiliation(s)
- Anna Kosyreva
- Department of Neuromorphology, Science Research Institute of Human Morphology, Moscow, Russia
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| | - Dzhuliia Dzhalilova
- Department of Immunomorphology of Inflammation, Science Research Institute of Human Morphology, Moscow, Russia
| | - Anastasia Lokhonina
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Polina Vishnyakova
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Growth and Development, Science Research Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
18
|
Paget TL, Parkinson-Lawrence EJ, Trim PJ, Autilio C, Panchal MH, Koster G, Echaide M, Snel MF, Postle AD, Morrison JL, Pérez-Gil J, Orgeig S. Increased Alveolar Heparan Sulphate and Reduced Pulmonary Surfactant Amount and Function in the Mucopolysaccharidosis IIIA Mouse. Cells 2021; 10:849. [PMID: 33918094 PMCID: PMC8070179 DOI: 10.3390/cells10040849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disease with significant neurological and skeletal pathologies. Respiratory dysfunction is a secondary pathology contributing to mortality in MPS IIIA patients. Pulmonary surfactant is crucial to optimal lung function and has not been investigated in MPS IIIA. We measured heparan sulphate (HS), lipids and surfactant proteins (SP) in pulmonary tissue and bronchoalveolar lavage fluid (BALF), and surfactant activity in healthy and diseased mice (20 weeks of age). Heparan sulphate, ganglioside GM3 and bis(monoacylglycero)phosphate (BMP) were increased in MPS IIIA lung tissue. There was an increase in HS and a decrease in BMP and cholesteryl esters (CE) in MPS IIIA BALF. Phospholipid composition remained unchanged, but BALF total phospholipids were reduced (49.70%) in MPS IIIA. There was a reduction in SP-A, -C and -D mRNA, SP-D protein in tissue and SP-A, -C and -D protein in BALF of MPS IIIA mice. Captive bubble surfactometry showed an increase in minimum and maximum surface tension and percent surface area compression, as well as a higher compressibility and hysteresis in MPS IIIA surfactant upon dynamic cycling. Collectively these biochemical and biophysical changes in alveolar surfactant are likely to be detrimental to lung function in MPS IIIA.
Collapse
Affiliation(s)
- Tamara L. Paget
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Emma J. Parkinson-Lawrence
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| | - Paul J. Trim
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Chiara Autilio
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Madhuriben H. Panchal
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Grielof Koster
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Mercedes Echaide
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Marten F. Snel
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (P.J.T.); (M.F.S.)
| | - Anthony D. Postle
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.H.P.); (G.K.); (A.D.P.)
| | - Janna L. Morrison
- Early Origins Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Jésus Pérez-Gil
- Department of Biochemistry, Faculty of Biology and Research Institute Hospital 12 de Octubre (Imas12), Complutense University, 28003 Madrid, Spain; (C.A.); (M.E.); (J.P.-G.)
| | - Sandra Orgeig
- Mechanisms in Cell Biology and Disease Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (T.L.P.); (E.J.P.-L.)
| |
Collapse
|
19
|
Duan XZ, Zhang X, Tong DK, Ji F, Xu KH, He RZ. Risk factors for and predictive nomogram of postoperative hypoxaemia in elderly patients with femoral neck fractures. J Int Med Res 2020; 48:300060520945132. [PMID: 33028126 PMCID: PMC7550957 DOI: 10.1177/0300060520945132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the related risk factors and predictive nomogram of postoperative hypoxaemia in elderly patients with femoral neck fractures. Methods This study included patients aged ≥65 years who underwent surgical treatment of acute femoral neck fractures. Univariate and multivariate logistic analyses were performed to determine the incidence of and risk factors for postoperative hypoxaemia. A predictive nomogram was constructed based on the multivariable model. Using the bootstrap method, discrimination was determined by the C-index and calibration plot. Results The logistic regression analysis showed that the anaesthesia type, surgical procedure, American Society of Anesthesiologists (ASA) classification, preoperative hypoxaemia occurrence, and age were independent predictors of development of postoperative hypoxaemia. The predictive formula for hypoxaemia was established as follows: hypoxaemia=−0.8668×spinal anaesthesia (whether)+0.1162×nerve anaesthesia (whether)+1.9555×plate/screw fixation (whether)+1.4950×hip replacement (whether)+0.4883×ASA classification+1.7153×preoperative oxygenation index+0.1608×age. With the bootstrap method, the prediction curve fit well with the ideal curve, suggesting that the prediction curve constructed in this study has good predictive ability. Conclusions Anaesthesia type, surgical procedure, ASA classification, preoperative hypoxaemia occurrence, and age were risk factors for postoperative hypoxaemia in elderly patients with femoral neck fractures. The predictive nomogram was designed for preoperative assessment of the risk of postoperative hypoxaemia by calculating the risk score.
Collapse
Affiliation(s)
- Xu-Zhou Duan
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| | - Xin Zhang
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| | - Da-Ke Tong
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| | - Fang Ji
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| | - Kai-Hang Xu
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| | - Rong-Zhi He
- Department of Orthopedics, Changhai Hospital Affiliated to the Navy Military Medical University, Shanghai, China
| |
Collapse
|
20
|
Mesenchymal stem cells alleviate LPS-induced acute lung injury by inhibiting the proinflammatory function of Ly6C + CD8 + T cells. Cell Death Dis 2020; 11:829. [PMID: 33024074 PMCID: PMC7538431 DOI: 10.1038/s41419-020-03036-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022]
Abstract
Systemic inflammatory processes, including alveolar injury, cytokine induction, and neutrophil accumulation, play key roles in the pathophysiology of acute lung injury (ALI). The immunomodulatory effects of mesenchymal stem cells (MSCs) can contribute to the treatment of inflammatory disorders. In previous studies, the focus was on innate immune cells and the effects of MSCs on ALI through CD8+ T cells remain unclear. In the present study, lipopolysaccharide (LPS) was used to induce ALI in mice. ALI mice were treated with MSCs via intratracheal instillation. Survival rate, histopathological changes, protein levels, total cell count, cytokine levels, and chemokine levels in alveolar lavage fluid were used to determine the efficacy of MSCs. Mass cytometry and single-cell RNA sequencing (scRNA-seq) were used to characterize the CD8+ T cells in the lungs. Ly6C- CD8+ T cells are prevalent in normal mice, whereas a specialized effector phenotype expressing a high level of Ly6C is predominant in advanced disease. MSCs significantly mitigated ALI and improved survival. MSCs decreased the infiltration of CD8+ T cells, especially Ly6C+ CD8+ T cells into the lungs. Mass cytometry revealed that CD8+ T cells expressing high Ly6C and CXCR3 levels caused tissue damage in the lungs of ALI mice, which was alleviated by MSCs. The scRNA-seq showed that Ly6C+ CD8+ T cells exhibited a more activated phenotype and decreased expression of proinflammatory factors that were enriched the most in immune chemotaxis after treatment with MSCs. We showed that CD8+ T cells play an important role in MSC-mediated ALI remission, and both infiltration quantity and proinflammatory function were inhibited by MSCs, indicating a potential mechanism for therapeutic intervention.
Collapse
|
21
|
Wyman AE, Nguyen TTT, Karki P, Tulapurkar ME, Zhang CO, Kim J, Feng TG, Dabo AJ, Todd NW, Luzina IG, Geraghty P, Foronjy RF, Hasday JD, Birukova AA, Atamas SP, Birukov KG. SIRT7 deficiency suppresses inflammation, induces EndoMT, and increases vascular permeability in primary pulmonary endothelial cells. Sci Rep 2020; 10:12497. [PMID: 32719338 PMCID: PMC7385158 DOI: 10.1038/s41598-020-69236-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
Acute lung injury (ALI), a common condition in critically ill patients, has limited treatments and high mortality. Aging is a risk factor for ALI. Sirtuins (SIRTs), central regulators of the aging process, decrease during normal aging and in aging-related diseases. We recently showed decreased SIRT7 expression in lung tissues and fibroblasts from patients with pulmonary fibrosis compared to controls. To gain insight into aging-related mechanisms in ALI, we investigated the effects of SIRT7 depletion on lipopolysaccharide (LPS)-induced inflammatory responses and endothelial barrier permeability in human primary pulmonary endothelial cells. Silencing SIRT7 in pulmonary artery or microvascular endothelial cells attenuated LPS-induced increases in ICAM1, VCAM1, IL8, and IL6 and induced endomesenchymal transition (EndoMT) with decreases in VE-Cadherin and PECAM1 and increases in collagen, alpha-smooth muscle actin, TGFβ receptor 1, and the transcription factor Snail. Loss of endothelial adhesion molecules was accompanied by increased F-actin stress fibers and increased endothelial barrier permeability. Together, these results show that an aging phenotype induced by SIRT7 deficiency promotes EndoMT with impaired inflammatory responses and dysfunction of the lung vascular barrier.
Collapse
Affiliation(s)
- Anne E Wyman
- Geriatric Research Education and Clinical Center (GRECC), VA Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD, USA. .,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA. .,Research Service, Baltimore VA Medical Center, Baltimore, MD, USA. .,Departments of Medicine and Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA.
| | - Trang T T Nguyen
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mohan E Tulapurkar
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chen-Ou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Junghyun Kim
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Theresa G Feng
- Department of Anesthesiology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Abdoulaye J Dabo
- Departments of Medicine and Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Research Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Research Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Patrick Geraghty
- Departments of Medicine and Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Robert F Foronjy
- Departments of Medicine and Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jeffrey D Hasday
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Research Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Research Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Konstantin G Birukov
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
22
|
Lu Z, Feng H, Shen X, He R, Meng H, Lin W, Geng Q. MiR-122-5p protects against acute lung injury via regulation of DUSP4/ERK signaling in pulmonary microvascular endothelial cells. Life Sci 2020; 256:117851. [PMID: 32470454 DOI: 10.1016/j.lfs.2020.117851] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Abstract
AIMS The aim of this study was to explore the role of miR-122-5p in acute lung injury. MATERIALS AND METHODS Mice were subjected to intratracheal injection of lipopolysaccharide to establish an acute lung injury model. The mice also received miR-122-5p antagonist and mimic via injection to inhibit or overexpress miR-122-5p in the lung tissue, respectively. In an in vitro experiment, we isolated primary mouse lung microvascular endothelial cells and established a cell injury model via lipopolysaccharide treatment. KEY FINDINGS Mice injected with an miR-122-5p antagonist exhibited reduced lung injury, inflammation and oxidative stress, while mice injected with a miR-122-5p mimic exhibited exaggerated lung injury, inflammation and oxidative stress. In an in vitro experiment, we found that the miR-122-5p antagonist suppressed lipopolysaccharide-induced inflammation, apoptosis and oxidative stress. Moreover, miR-122-5p regulated the promoter activity of DUSP4, which negatively regulated ERK1/2 signaling. The use of DUSP4 siRNA counteracted the effects of the miR-122-5p antagonist. SIGNIFICANCE Taken together, these results show that miR-122-5p protected against acute lung injury via regulation of DUSP4/ERK signaling in pulmonary microvascular endothelial cells. MiR-122-5p antagonism may be a promising treatment method for acute lung injury.
Collapse
Affiliation(s)
- Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Haojie Feng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaokang Shen
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Heng Meng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weichen Lin
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
23
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Calkovska A, Mokra D. Effects of PDE3 Inhibitor Olprinone on the Respiratory Parameters, Inflammation, and Apoptosis in an Experimental Model of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E3382. [PMID: 32403267 PMCID: PMC7247002 DOI: 10.3390/ijms21093382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1β, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Pavol Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Sona Balentova
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Andrea Calkovska
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Daniela Mokra
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| |
Collapse
|
24
|
Ye C, Li H, Bao M, Zhuo R, Jiang G, Wang W. Alveolar macrophage - derived exosomes modulate severity and outcome of acute lung injury. Aging (Albany NY) 2020; 12:6120-6128. [PMID: 32259794 PMCID: PMC7185135 DOI: 10.18632/aging.103010] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/20/2020] [Indexed: 12/31/2022]
Abstract
Severe acute lung injury (ALI) can cause death, and the survivals may develop acute respiratory distress syndrome (ARDS) due to fibrotic repair of the lung. Alveolar macrophages play a demonstrative role during the pathogenesis of ALI, and the timing and degree of differentially polarization of macrophages determine the severity of disease and outcome. Exosomes are important mediators of cellular communication and play critical roles during macrophage differentiation, proliferation and function. Nevertheless, the exact effects of alveolar macrophage - derived exosomes on ALI remain unknow. Here, we used lipopolysaccharide (LPS) to induce ALI in mice and analyzed the exosome population in bronchoalveolar lavage fluid (BALF) from macrophages, neutrophils and epithelial cells at different time points after treatment. Our data showed that macrophages were the major secretors for early secreted pro-inflammatory cytokines in the BALF-exosomes, which likely activated neutrophils to produce a variety of pro-inflammatory cytokines and IL-10. IL-10 by neutrophils in BALF-exosomes likely in turn polarized macrophages to M2c, which may be responsible for post-ALI fibrosis. Our study thus reveals a previous non-acknowledged role of BALF-exosomes as a mediator of inflammatory response and cell crosstalk during ALI.
Collapse
Affiliation(s)
- Cong Ye
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Huiting Li
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Minwei Bao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Ran Zhuo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Weixi Wang
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
25
|
Wu XN, Yang Y, Zhang HH, Zhong YS, Wu F, Yu B, Yu CH. Robustaflavone-4'-dimethyl ether from Selaginella uncinata attenuated lipopolysaccharide-induced acute lung injury via inhibiting FLT3-mediated neutrophil activation. Int Immunopharmacol 2020; 82:106338. [PMID: 32109679 DOI: 10.1016/j.intimp.2020.106338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 12/20/2022]
Abstract
Neutrophils act as both messenger and effector which contributed to the pathogenesis of acute lung injury (ALI). Targeting neutrophils could be a novel strategy for prevention and treatment of ALI. Selaginella uncinata is widely used as an antitussive, antipyretic and anti-inflammatory herb to treat various pulmonary diseases, including lung cancer, asthma, pulmonary fibrosis and pneumonia. However, its effective constituents remain unknown. In the present study, the protective effects of flavonoids from S. uncinata (SUF) and its major compound robustaflavone-4'-dimethyl ether (RDE) against lipopolysaccharide (LPS)-induced ALI were investigated in mice and in neutrophils.The results showed that both SUF and RDE had the same inhibition on LPS-induced lung edema and neutrophil infiltration as well as the increased levels of IL-6, TNF-α, P-selectin and ICAM-1 in serum of LPS-challenged mice. Furthermore, RDE significantly inhibited inducible neutrophil activation in a concentration-dependent manner, and also reduced the levels of intracellular calcium as well as the expressions of CCR2. Rescue experiment showed that RDE suppressed FLT3 and its downstream p-p38 and p-AKT, which could be abolished by FLT3 agonist FLT3L but partly by MAPK agonist PDBu or AKT agonist SC79. Therefore, these results indicated that RDE as the main bioactive compound in SUF alleviated LPS-induced acute lung injury and inhibited neutrophil activation via inhibition of FLT3-mediatied AKT and MAPK pathways.
Collapse
Affiliation(s)
- Xiao-Ning Wu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Yang Yang
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu-Sen Zhong
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Fang Wu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Bing Yu
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
26
|
Cui SN, Chen L, Yang YY, Wang YX, Li SN, Zhou T, Xiao HR, Qin L, Yang W, Yuan SY, Yao SL, Shang Y. Activation of death-associated protein kinase 1 promotes neutrophil apoptosis to accelerate inflammatory resolution in acute respiratory distress syndrome. J Transl Med 2019; 99:1143-1156. [PMID: 30911150 DOI: 10.1038/s41374-019-0242-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a uniform progression of overwhelming inflammation in lung tissue with extensive infiltration of inflammatory cells. Neutrophil apoptosis is thought to be a significant process in the control of the resolution phase of inflammation. It has been proved that 5-Aza-2'-deoxycytidine (Aza) can inhibit cancer by activating death-associated protein kinase 1 (DAPK1) to promote apoptosis. However, the effect of DAPK1 on neutrophil apoptosis is unclear, and research on the role of Aza in inflammation is lacking. Here, we investigated whether Aza can regulate DAPK1 expression to influence the fate of neutrophils in ARDS. In vitro, we stimulated neutrophil-like HL-60 (dHL-60) cells with different concentrations of Aza for different durations and used RNA interference to up- or downregulate DAPK1 expression. We observed that culturing dHL-60 cells with Aza increased apoptosis by inhibiting NF-κB activation to modulate the expression of Bcl-2 family proteins, which was closely related to the levels of DAPK1. In vivo, ARDS was evoked by intratracheal instillation of lipopolysaccharide (LPS; 3 mg/kg). One hour after LPS administration, mice were treated with Aza (1 mg/kg, i.p.). To inhibit DAPK1 expression, mice were intraperitoneally injected with a DAPK1 inhibitor. Aza treatment accelerated inflammatory resolution in LPS-induced ARDS by suppressing pulmonary edema, alleviating lung injury and decreasing the infiltration of inflammatory cells in bronchoalveolar lavage fluid (BALF). Moreover, Aza reduced the production of proinflammatory cytokines. However, administration of the DAPK1 inhibitor attenuated the protective effects of Aza. Similarly, the proapoptotic function of Aza was prevented when DAPK1 was inhibited either in vivo or in vitro. In summary, Aza promotes neutrophil apoptosis by activating DAPK1 to accelerate inflammatory resolution in LPS-induced ARDS. This study provides the first evidence that Aza prevents LPS-induced neutrophil survival by modulating DAPK1 expression.
Collapse
Affiliation(s)
- Shu-Nan Cui
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Chen
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Yi Yang
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng-Nan Li
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Rong Xiao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Qin
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Yang
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Ying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shang-Long Yao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
27
|
Liang GP, Xu J, Cao LL, Zeng YH, Chen BX, Yang J, Zhang ZW, Kang Y. Piezo1 induced apoptosis of type II pneumocytes during ARDS. Respir Res 2019; 20:118. [PMID: 31186017 PMCID: PMC6558715 DOI: 10.1186/s12931-019-1083-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/24/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The mechanisms of lung injury in acute respiratory distress syndrome (ARDS) are not well understood.Piezo1 was recently identified as a mechanotransduction protein. The present study found the expression of Piezo1 in type II pneumocytes and investigated its role in mediating ARDS-related lung injury. METHODS Sprague-Dawley rats were used to establish an ARDS model, the expression of Piezo1,lung injuries, apoptosis as well as calcium influx were assessed. RESULTS Piezo1 was expressed in type II pneumocytes as shown by immunofluorescence staining and expression was increased in the ARDS model. Knockdown of Piezo1 reduced apoptosis which was related to the elevation of Bcl-2.Calcium influx played a vital role in Piezo1-induced apoptosis. CONCLUSION Piezo1 was expressed in type II pneumocytes. Mechanical stretch of alveoli during ARDS induced activation of the Piezo1 channel,which resulted in calcium influx. The increased intracellular Ca2+ induced the apoptosis of type II pneumocytes, which may be related to the Bcl-2 pathway.
Collapse
Affiliation(s)
- Guo-Peng Liang
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li-Li Cao
- Chengdu Medical College, Chengdu, 610041, Sichuan, China
| | - Yi-Hua Zeng
- Department of Respiratory Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bai-Xu Chen
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Yang
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhong-Wei Zhang
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yan Kang
- Department of Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
West J, Chen X, Yan L, Gladson S, Loyd J, Rizwan H, Talati M. Adverse effects of BMPR2 suppression in macrophages in animal models of pulmonary hypertension. Pulm Circ 2019; 10:2045894019856483. [PMID: 31124398 PMCID: PMC7074495 DOI: 10.1177/2045894019856483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 01/11/2023] Open
Abstract
Inflammatory cells contribute to irreversible damage in pulmonary arterial hypertension (PAH). We hypothesized that in PAH, dysfunctional BMPR2 signaling in macrophages contributes to pulmonary vascular injury and phenotypic changes via proinflammatory cytokine production. Studies were conducted in: (1) Rosa26-rtTA2 3 X TetO7-Bmpr2delx4 FVB/N mice (mutant Bmpr2 is universally expressed, BMPR2delx4 mice) given a weekly intra-tracheal liposomal clodronate injections for four weeks; and (2) LysM-Cre X floxed BMPR2 X floxed eGFP monocyte lineage-specific BMPR2 knockout (KO) mouse model (Bmpr2 gene expression knockdown in monocytic lineage cells) (BMPR2KO) following three weeks of sugen/hypoxia treatment. In the BMPR2delx4 mice, increased right ventricular systolic pressure (RVSP; P < 0.05) was normalized by clodronate, and in monocyte lineage-specific BMPR2KO mice sugen hypoxia treatment increased (P < 0.05) RVSP compared to control littermates, suggesting that suppressed BMPR2 in macrophages modulate RVSP in animal models of PH. In addition, in these mouse models, muscularized pulmonary vessels were increased (P < 0.05) and surrounded by an increased number of macrophages. Elimination of macrophages in BMPR2delx4 mice reduced the number of muscularized pulmonary vessels and macrophages surrounding these vessels. Further, in monocyte lineage-specific BMPR2KO mice, there was significant increase in proinflammatory cytokines, including C-X-C Motif Chemokine Ligand 12 (CXCL12), complement component 5 a (C5a), Interleukin-16 (IL-16), and secretory ICAM. C5a positive inflammatory cells present in and around the pulmonary vessels in the PAH lung could potentially be involved in pulmonary vessel remodeling. In summary, our data indicate that, in BMPR2-related PAH, macrophages with dysfunctional BMPR2 influence pulmonary vascular remodeling and phenotypic outcomes via proinflammatory cytokine production.
Collapse
Affiliation(s)
- James West
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Xinping Chen
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ling Yan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Santhi Gladson
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James Loyd
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hamid Rizwan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Megha Talati
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
29
|
Zhang YF, Zhang J, Sun CC, Tang CY, Sun GY, Luo WJ, Zhou Y, Guan CX. Vasoactive intestinal peptide inhibits the activation of murine fibroblasts and expression of interleukin 17 receptor C. Cell Biol Int 2019; 43:770-780. [PMID: 31026365 DOI: 10.1002/cbin.11151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/01/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is an acute, severe, and refractory pulmonary inflammation with high morbidity and mortality. Excessive activation of fibroblast during the fibroproliferative phase plays a pivotal role in the prognosis of ARDS. Our previous study demonstrated that the vasoactive intestinal peptide (VIP) is mediated by lentivirus attenuates lipopolysaccharide (LPS)-induced ARDS in a murine model, and VIP inhibits the release of interleukin-17A (IL-17A) from activation macrophages. However, the effects of VIP on the activation of murine fibroblast and expression of IL-17 receptor (IL-17R) in ARDS remain unclear. Here, a mouse model of ARDS was established by an intratracheal injection of LPS. We found that the gene expression of col3a1 and hydroxyproline contents in the lungs were significantly increased 24 h after LPS injection. IL-17RC rather than IL-17RA was increased in the lungs of mice with ARDS. In vitro, LPS activated NIH3T3 cells, which was suppressed by VIP in a dose-dependent manner. In detail, VIP reduced the hydroxyproline content and col3a1 messenger RNA induced by LPS in NIH3T3 cells, as well as the expression of α-smooth muscle actin. Furthermore, we found that VIP inhibited the expression of IL-17R in the lungs of mice with ARDS and NIH3T3 cells stimulated with LPS, which was partly inhibited by antagonists of protein kinase A and protein kinase C. Taken together, our results demonstrated that VIP inhibited the activation of fibroblast via downregulation of IL-17RC, which may contribute to the protective effects of VIP against ARDS in mice.
Collapse
Affiliation(s)
- Yan-Feng Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Zhang
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chun-Yan Tang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guo-Ying Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wan-Jun Luo
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
30
|
Lanyu Z, Feilong H. Emerging role of extracellular vesicles in lung injury and inflammation. Biomed Pharmacother 2019; 113:108748. [DOI: 10.1016/j.biopha.2019.108748] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/19/2019] [Accepted: 02/27/2019] [Indexed: 12/26/2022] Open
|
31
|
Effects of calcium gluconate on lipopolysaccharide-induced acute lung injury in mice. Biochem Biophys Res Commun 2018; 503:2931-2935. [DOI: 10.1016/j.bbrc.2018.08.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022]
|
32
|
Bi G, Wu L, Huang P, Islam S, Heruth DP, Zhang LQ, Li DY, Sampath V, Huang W, Simon BA, Easley RB, Ye SQ. Up-regulation of SFTPB expression and attenuation of acute lung injury by pulmonary epithelial cell-specific NAMPT knockdown. FASEB J 2018; 32:3583-3596. [PMID: 29452569 PMCID: PMC5998971 DOI: 10.1096/fj.201701059r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022]
Abstract
Although a deficiency of surfactant protein B (SFTPB) has been associated with lung injury, SFTPB expression has not yet been linked with nicotinamide phosphoribosyltransferase (NAMPT), a potential biomarker of acute lung injury (ALI). The effects of Nampt in the pulmonary epithelial cell on both SFTPB expression and lung inflammation were investigated in a LPS-induced ALI mouse model. Pulmonary epithelial cell-specific knockdown of Nampt gene expression, achieved by the crossing of Nampt gene exon 2 floxed mice with mice expressing epithelial-specific transgene Cre or by the use of epithelial-specific expression of anti-Nampt antibody cDNA, significantly attenuated LPS-induced ALI. Knockdown of Nampt expression was accompanied by lower levels of bronchoalveolar lavage (BAL) neutrophil infiltrates, total protein and TNF-α levels, as well as lower lung injury scores. Notably, Nampt knockdown was also associated with significantly increased BAL SFTPB levels relative to the wild-type control mice. Down-regulation of NAMPT increased the expression of SFTPB and rescued TNF-α-induced inhibition of SFTPB, whereas overexpression of NAMPT inhibited SFTPB expression in both H441 and A549 cells. Inhibition of NAMPT up-regulated SFTPB expression by enhancing histone acetylation to increase its transcription. Additional data indicated that these effects were mainly mediated by NAMPT nonenzymatic function via the JNK pathway. This study shows that pulmonary epithelial cell-specific knockdown of NAMPT expression attenuated ALI, in part, via up-regulation of SFTPB expression. Thus, epithelial cell-specific knockdown of Nampt may be a potential new and viable therapeutic modality to ALI.-Bi, G., Wu, L., Huang, P., Islam, S., Heruth, D. P., Zhang, L. Q., Li, D.-Y., Sampath, V., Huang, W., Simon, B. A., Easley, R. B., Ye, S. Q. Up-regulation of SFTPB expression and attenuation of acute lung injury by pulmonary epithelial cell-specific NAMPT knockdown.
Collapse
Affiliation(s)
- Guangliang Bi
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Wu
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
- Department of Pediatrics, Changsha Central Hospital, Changsha, China
| | - Peixin Huang
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Shamima Islam
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Daniel P. Heruth
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Li Qin Zhang
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Ding-You Li
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Venkatesh Sampath
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Weimin Huang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Brett A. Simon
- Department of Anesthesiology, Josie Robertson Surgery Center, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Ronald Blaine Easley
- Department of Pediatrics-Anesthesiology, Baylor College of Medicine, Houston, Texas, USA
| | - Shui Qing Ye
- Department of Pediatrics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
- Department of Biomedical and Health Informatics, Children’s Mercy, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| |
Collapse
|
33
|
Development, remodeling and regeneration of the lung: coping with the structural and functional challenges of breathing. Cell Tissue Res 2017; 367:407-411. [DOI: 10.1007/s00441-016-2568-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|