1
|
Wei T, Lu C, Du H, Yang Q, Qi X, Liu Y, Zhang Y, Chen C, Li Y, Tang Y, Zhang WH, Tao X, Jiang N. DeepPBI-KG: a deep learning method for the prediction of phage-bacteria interactions based on key genes. Brief Bioinform 2024; 25:bbae484. [PMID: 39344712 PMCID: PMC11440089 DOI: 10.1093/bib/bbae484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Phages, the natural predators of bacteria, were discovered more than 100 years ago. However, increasing antimicrobial resistance rates have revitalized phage research. Methods that are more time-consuming and efficient than wet-laboratory experiments are needed to help screen phages quickly for therapeutic use. Traditional computational methods usually ignore the fact that phage-bacteria interactions are achieved by key genes and proteins. Methods for intraspecific prediction are rare since almost all existing methods consider only interactions at the species and genus levels. Moreover, most strains in existing databases contain only partial genome information because whole-genome information for species is difficult to obtain. Here, we propose a new approach for interaction prediction by constructing new features from key genes and proteins via the application of K-means sampling to select high-quality negative samples for prediction. Finally, we develop DeepPBI-KG, a corresponding prediction tool based on feature selection and a deep neural network. The results show that the average area under the curve for prediction reached 0.93 for each strain, and the overall AUC and area under the precision-recall curve reached 0.89 and 0.92, respectively, on the independent test set; these values are greater than those of other existing prediction tools. The forward and reverse validation results indicate that key genes and key proteins regulate and influence the interaction, which supports the reliability of the model. In addition, intraspecific prediction experiments based on Klebsiella pneumoniae data demonstrate the potential applicability of DeepPBI-KG for intraspecific prediction. In summary, the feature engineering and interaction prediction approaches proposed in this study can effectively improve the robustness and stability of interaction prediction, can achieve high generalizability, and may provide new directions and insights for rapid phage screening for therapy.
Collapse
Affiliation(s)
- Tongqing Wei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Chenqi Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Hanxiao Du
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Qianru Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Xin Qi
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
| | - Yankun Liu
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
| | - Yi Zhang
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Chen Chen
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Yutong Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Yuanhao Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Wen-Hong Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Xu Tao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| |
Collapse
|
2
|
Lee S, Yoon SJ, Oh JH, Ryu JS, Park Y, Hwang ES. MPoMA protects against lung epithelial cell injury via p65 degradation. Biomed Pharmacother 2024; 175:116674. [PMID: 38703509 DOI: 10.1016/j.biopha.2024.116674] [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: 02/13/2024] [Revised: 04/20/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Numerous cases of lung injury caused by viral infection were reported during the coronavirus disease-19 pandemic. While there have been significant efforts to develop drugs that block viral infection and spread, the development of drugs to reduce or reverse lung injury has been a lower priority. This study aimed to identify compounds from a library of compounds that prevent viral infection that could reduce and prevent lung epithelial cell damage. We investigated the cytotoxicity of the compounds, their activity in inhibiting viral spike protein binding to cells, and their activity in reducing IL-8 production in lung epithelial cells damaged by amodiaquine (AQ). We identified N-(4-(4-methoxyphenoxy)-3-methylphenyl)-N-methylacetamide (MPoMA) as a non-cytotoxic inhibitor against viral infection and AQ-induced cell damage. MPoMA inhibited the expression of IL-8, IL-6, IL-1β, and fibronectin induced by AQ and protected against AQ-induced morphological changes. However, MPoMA did not affect basal IL-8 expression in lung epithelial cells in the absence of AQ. Further mechanistic analysis confirmed that MPoMA selectively promoted the proteasomal degradation of inflammatory mediator p65, thereby reducing intracellular p65 expression and p65-mediated inflammatory responses. MPoMA exerted potent anti-inflammatory and protective functions in epithelial cells against LPS-induced acute lung injury in vivo. These findings suggest that MPoMA may have beneficial effects in suppressing viral infection and preventing lung epithelial cell damage through the degradation of p65 and inhibition of the production of inflammatory cytokines.
Collapse
Affiliation(s)
- Soheun Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Suh Jin Yoon
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ji Hyun Oh
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Sang Ryu
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yunjeong Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
3
|
Malainou C, Abdin SM, Lachmann N, Matt U, Herold S. Alveolar macrophages in tissue homeostasis, inflammation, and infection: evolving concepts of therapeutic targeting. J Clin Invest 2023; 133:e170501. [PMID: 37781922 PMCID: PMC10541196 DOI: 10.1172/jci170501] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Alveolar macrophages (AMs) are the sentinel cells of the alveolar space, maintaining homeostasis, fending off pathogens, and controlling lung inflammation. During acute lung injury, AMs orchestrate the initiation and resolution of inflammation in order to ultimately restore homeostasis. This central role in acute lung inflammation makes AMs attractive targets for therapeutic interventions. Single-cell RNA-Seq and spatial omics approaches, together with methodological advances such as the generation of human macrophages from pluripotent stem cells, have increased understanding of the ontogeny, function, and plasticity of AMs during infectious and sterile lung inflammation, which could move the field closer to clinical application. However, proresolution phenotypes might conflict with proinflammatory and antibacterial responses. Therefore, therapeutic targeting of AMs at vulnerable time points over the course of infectious lung injury might harbor the risk of serious side effects, such as loss of antibacterial host defense capacity. Thus, the identification of key signaling hubs that determine functional fate decisions in AMs is of the utmost importance to harness their therapeutic potential.
Collapse
Affiliation(s)
- Christina Malainou
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Shifaa M. Abdin
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- RESIST (Resolving Infection Susceptibility), Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
4
|
Noël F, Mauroy B. Propagation of an idealized infection in an airway tree, consequences of the inflammation on the oxygen transfer to blood. J Theor Biol 2023; 561:111405. [PMID: 36639022 DOI: 10.1016/j.jtbi.2023.111405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/02/2022] [Accepted: 12/31/2022] [Indexed: 01/12/2023]
Abstract
A mathematical model of infection, inflammation and immune response in an idealized bronchial tree is developed. This work is based on a model from the literature that is extended to account for the propagation dynamics of an infection between the airways. The inflammation affects the size of the airways, the air flows distribution in the airway tree, and, consequently, the oxygen transfers to blood. We test different infections outcomes and propagation speed. In the hypotheses of our model, the inflammation can reduce notably and sometimes drastically the oxygen flow to blood. Our model predicts how the air flows and oxygen exchanges reorganize in the tree during an infection. Our results highlight the links between the localization of the infection and the amplitude of the loss of oxygen flow to blood. We show that a compensation phenomena due to the reorganization of the flow exists, but that it remains marginal unless the power produced the ventilation muscles is increased. Our model forms a first step towards a better understanding of the dynamics of bronchial infections.
Collapse
Affiliation(s)
- Frédérique Noël
- Université Côte d'Azur, CNRS, LJAD, Vader center, Nice, France; INRIA Paris, France.
| | - Benjamin Mauroy
- Université Côte d'Azur, CNRS, LJAD, Vader center, Nice, France.
| |
Collapse
|
5
|
Yang H. Silencing of Long Non-coding RNA H19 Alleviates Lipopolysaccharide (LPS)-induced Apoptosis and Inflammation Injury by Regulating miR-140-5p/TLR4 Axis in Cell Models of Pneumonia. Curr Mol Med 2023; 23:275-284. [PMID: 35392782 DOI: 10.2174/1566524022666220407100949] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Mounting studies have clarified the link between long non-coding RNAs (lncRNAs) and pneumonia. This research aims to probe the function and regulatory mechanism of lncRNA H19 in lipopolysaccharide (LPS)-induced cell models of pneumonia. METHODS WI-38 cells were exposed to LPS for 12 h to mimic cell models of pneumonia. The relative expression of H19, miR-140-5p, and toll-like receptor 4 (TLR4) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The cell viability was detected by MTT assay. The protein expression of apoptosis-associated proteins (Bax and Bcl-2) and TLR4 were determined by western blot. Moreover, the content of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were measured by enzyme-linked immunosorbent assay (ELISA). The target relationship between miR- 140-5p and H19/ TLR4 was confirmed by Dual luciferase reporter (DLR) assay. RESULTS LncRNA H19 and TLR4 were up-regulated, while miR-140-5p was downregulated in peripheral blood of patients with pneumonia and LPS-treated WI-38 cells compared with their controls. Silencing of H19 or miR-140-5p mimics facilitated cell viability, whereas repressed apoptosis and reduced content of TNF-α, IL-6, and IL-1β in LPS-induced WI-38 cells. H19 targeted miR-140-5p and it inversely regulated miR-140- 5p expression. MiR-140-5p targeted TLR4 and it inversely regulated TLR4 expression. H19 positively regulated TLR4 expression. Moreover, inhibition of miR-140-5p or overexpression of TLR4 reversed the effects of H19 silencing on cell viability, inflammation, and apoptosis in LPS-induced WI-38 cells. CONCLUSION Silencing of H19 inhibited apoptosis and inflammation by miR-140- 5p/TLR4 pathway in LPS-induced WI-38 cells.
Collapse
Affiliation(s)
- Hong Yang
- Department of Pediatric, Affiliated Hospital of Beihua University, Jilin City, Jilin Province, 132011, China
| |
Collapse
|
6
|
Miao F, Shan C, Geng S, Ning D. Oleocanthal alleviated lipopolysaccharide-induced acute lung injury in chickens by inhibiting TLR4/NF-κB pathway activation. Poult Sci 2022; 102:102458. [PMID: 36640559 PMCID: PMC9842928 DOI: 10.1016/j.psj.2022.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/09/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022] Open
Abstract
This study aimed to investigate the ameliorative effect of oleocanthal (OC) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in chickens and its possible mechanisms. In total, 20 chickens were randomly divided into 4 groups: control (CON) group, LPS group, LPS + OC group, and OC group. LPS + OC and OC groups were intragastrically administered a 5 mg/kg·d OC dose for 7 d. On d 8, the LPS group and LPS + OC group were intratracheally administered 2 mg/kg LPS for 12 h. It was found that OC ameliorated the pathological morphology and significantly suppressed apoptosis after OC treatment in LPS-induced ALI chicken (P < 0.01). Antioxidant capacity was higher in the LPS + OC group compared with the LPS group (P < 0.01). OC downregulated the related genes and proteins expression of toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) pathway in LPS group (P < 0.01). In conclusion, OC supplementation can alleviate LPS-induced ALI in chickens by suppressing apoptosis, enhancing lung antioxidant capacities and inhibiting TLR4/NF-κB pathway activation.
Collapse
Affiliation(s)
- Fujun Miao
- Yunnan Academy of Forestry and Grassland, Kunming, 650204, P. R. China.
| | - Chunlan Shan
- College of Animal Science, Guizhou University, Guiyang, 550000, P. R. China
| | - Shuxiang Geng
- Yunnan Academy of Forestry and Grassland, Kunming, 650204, P. R. China
| | - Delu Ning
- Yunnan Academy of Forestry and Grassland, Kunming, 650204, P. R. China
| |
Collapse
|
7
|
Xia F, Yang L, Zhu X. Knockdown of circ_0038467 alleviates lipopolysaccharides-induced 16HBE cell injury by regulating the miR-545-3p/TRAF1 axis in neonatal pneumonia. Microb Pathog 2022; 173:105819. [PMID: 36216207 DOI: 10.1016/j.micpath.2022.105819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/22/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Neonatal pneumonia is a common illness in the neonatal period with a high fatality rate. Accumulating proofs have attested to the crucial role of circular RNAs (circRNAs) in pneumonia. This study was intended to expound on the function of circ_0038467 and the underlying mechanism in lipopolysaccharide (LPS)-stimulated 16HBE cell injury in neonatal pneumonia. METHODS 16HBE cells were exposed to LPS to establish an in vitro neonatal pneumonia cell model. Quantitative real-time polymerase chain reaction (qRT-PCR) was implemented for detecting the levels of circ_0038467, microRNA-545-3p (miR-545-3p), and tumor necrosis factor receptor-associated factor 1 (TRAF1) in neonatal pneumonia serums and LPS-treated 16HBE cells. Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU) incorporation, and flow cytometry assays were used to examine cell viability, proliferation, and apoptosis, respectively. The protein abundances of proliferation/apoptosis/inflammation-correlated makers and TRAF1 were tested by Western blot. RNase R and Actinomycin D assays were implemented to determine the features of circ_0038467. The mutual effect between miR-545-3p and circ_0038467 or TRAF1 was affirmed by a dual-luciferase reporter and RNA pull-down assay assays. RESULTS Circ_0038467 was upregulated in neonatal pneumonia serum specimens and LPS-triggered 16HBE cells. LPS administration restrained 16HBE cell proliferation and promoted apoptosis and inflammation, whereas circ_0038467 silence recovered these influences. Meanwhile, miR-545-3p was targeted by circ_0038467, and circ_0038467 could modulate LPS-treated 16HBE cell injury through absorbing miR-545-3p. Furthermore, circ_0038467 controlled TRAF1 level via segregating miR-545-3p. Moreover, TRAF1 overexpression relieved the suppressive impact of circ_0038467 silence in LPS-triggered 16HBE cell detriment. CONCLUSION Circ_0038467 knockdown mitigated LPS-exposed 16HBE cell damage through regulating miR-545-3p/PPARA axis.
Collapse
Affiliation(s)
- Fang Xia
- Department of Neonatology, Jingzhou Central Hospital (Jingzhou Hospital Affiliated to Yangtze University), Jingzhou, Hubei, China
| | - Long Yang
- Department of Neonatology, Jingzhou Central Hospital (Jingzhou Hospital Affiliated to Yangtze University), Jingzhou, Hubei, China
| | - Xiaofang Zhu
- Department of Neonatology, Jingzhou Central Hospital (Jingzhou Hospital Affiliated to Yangtze University), Jingzhou, Hubei, China.
| |
Collapse
|
8
|
Rayees S, Joshi JC, Joshi B, Vellingiri V, Banerjee S, Mehta D. Protease-activated receptor 2 promotes clearance of Pseudomonas aeruginosa infection by inducing cAMP-Rac1 signaling in alveolar macrophages. Front Pharmacol 2022; 13:874197. [PMID: 36204227 PMCID: PMC9530345 DOI: 10.3389/fphar.2022.874197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Efficient phagocytosis of pathogens by the innate immune system during infectious injury is vital for restoring tissue integrity. Impaired phagocytosis, such as in the case of infection with Pseudomonas aeruginosa, a broad-spectrum antibiotic-resistant Gram-negative bacterium, can lead to a life threatening lung disorder, acute lung injury (ALI). Evidence indicates that loss of protease-activated receptor 2 (PAR2) impaired Pseudomonas aeruginosa clearance leading to non-resolvable ALI, but the mechanism remains unclear. Here, we focused on the alveolar macrophages (AMs), the predominant population of lung-resident macrophages involved in sensing bacteria, to understand their role in PAR2-mediated phagocytosis of Pseudomonas aeruginosa. We found that upon binding Pseudomonas aeruginosa, PAR2-expressing but not PAR2-null AMs had increased cAMP levels, which activated Rac1 through protein kinase A. Activated Rac1 increased actin-rich protrusions to augment the phagocytosis of Pseudomonas aeruginosa. Administration of liposomes containing constitutively active Rac1 into PAR2-null mice lungs rescued phagocytosis and enhanced the survival of PAR2-null mice from pneumonia. These studies showed that PAR2 drives the cAMP-Rac1 signaling cascade that activates Pseudomonas aeruginosa phagocytosis in AMs, thereby preventing death from bacterial pneumonia.
Collapse
|
9
|
Highly pathogenic coronavirus N protein aggravates inflammation by MASP-2-mediated lectin complement pathway overactivation. Signal Transduct Target Ther 2022; 7:318. [PMID: 36100602 PMCID: PMC9470675 DOI: 10.1038/s41392-022-01133-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 12/30/2022] Open
Abstract
Excessive inflammatory responses contribute to the pathogenesis and lethality of highly pathogenic human coronaviruses, but the underlying mechanism remains unclear. In this study, the N proteins of highly pathogenic human coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), middle east respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), were found to bind MASP-2, a key serine protease in the lectin pathway of complement activation, resulting in excessive complement activation by potentiating MBL-dependent MASP-2 activation, and the deposition of MASP-2, C4b, activated C3 and C5b-9. Aggravated inflammatory lung injury was observed in mice infected with adenovirus expressing the N protein. Complement hyperactivation was also observed in SARS-CoV-2-infected patients. Either blocking the N protein:MASP-2 interaction, MASP-2 depletion or suppressing complement activation can significantly alleviate N protein-induced complement hyperactivation and lung injury in vitro and in vivo. Altogether, these data suggested that complement suppression may represent a novel therapeutic approach for pneumonia induced by these highly pathogenic coronaviruses.
Collapse
|
10
|
Icariin Alleviates Escherichia coli Lipopolysaccharide-Mediated Endometritis in Mice by Inhibiting Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:ijms231810219. [PMID: 36142129 PMCID: PMC9499631 DOI: 10.3390/ijms231810219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Icariin (ICA) is a naturally occurring phytochemical agent primarily extracted from Epimedium Brevicornum Maxim (Family Berberidaceae) with a broad spectrum of bioactivities. Endometritis is a uterine disease that causes enormous losses in the dairy industry worldwide. In this study, anti-inflammatory and anti-oxidant properties of ICA were investigated against lipopolysaccharide (LPS)-induced endometritis in mice to investigate possible underlying molecular mechanisms. Sixty heathy female Kunming mice were randomly assigned to four groups (n = 15), namely control, LPS, LPS + ICA, and ICA groups. The endometritis was induced by intrauterine infusion of 50 µL of LPS (1 mg/mL). After 24 h of onset of LPS-induced endometritis, ICA groups were injected thrice by ICA intraperitoneally six hours apart. Histopathological examination, enzyme linked immunosorbent assay (ELISA), real time quantitative polymerase chain reaction (RT-qPCR), western blotting, and immunohistochemistry were used in this study. Histological alterations revealed that ICA markedly mitigated uterine tissue injury caused by LPS. The results showed that the ICA inhibited the production of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) and boosted the production of anti-inflammatory cytokines (IL-10). Additionally, ICA modulated the expression of malondialdehyde (MDA), reactive oxygen species (ROS), superoxide dismutase 1 (SOD1), catalase (CAT), and glutathione peroxidase 1 (Gpx1) induced by LPS. The administration of ICA significantly (p < 0.05) improved the mRNA and protein expression of Toll-like receptor (TLR) 4. The western blotting and ELISA finding revealed that the ICA repressed LPS-triggered NF-κB pathway activation. Moreover, ICA improved the antioxidant defense system via activation of the Nrf2 pathway. The results revealed that ICA up-regulated the mRNA and protein expression of Nuclear erythroid-2-related factor (Nrf2), NAD(P)H: quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1), and glutamate-cysteine ligase catalytic subunit (GCLC) under LPS exposure. Conclusively, our findings strongly suggested that ICA protects endometritis caused by LPS by suppressing TLR4-associated NF-κB and Nrf2 pathways. Altogether, these innovative findings may pave the way for future studies into the therapeutic application of ICA to protect humans and animals against endometritis.
Collapse
|
11
|
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: 12] [Impact Index Per Article: 6.0] [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
|
12
|
Liang Y, Zhu C, Tian C, Lin Q, Li Z, Li Z, Ni D, Ma X. Early prediction of ventilator-associated pneumonia in critical care patients: a machine learning model. BMC Pulm Med 2022; 22:250. [PMID: 35752818 PMCID: PMC9233772 DOI: 10.1186/s12890-022-02031-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022] Open
Abstract
Background This study was performed to develop and validate machine learning models for early detection of ventilator-associated pneumonia (VAP) 24 h before diagnosis, so that VAP patients can receive early intervention and reduce the occurrence of complications. Patients and methods This study was based on the MIMIC-III dataset, which was a retrospective cohort. The random forest algorithm was applied to construct a base classifier, and the area under the receiver operating characteristic curve (AUC), sensitivity and specificity of the prediction model were evaluated. Furthermore, We also compare the performance of Clinical Pulmonary Infection Score (CPIS)-based model (threshold value ≥ 3) using the same training and test data sets. Results In total, 38,515 ventilation sessions occurred in 61,532 ICU admissions. VAP occurred in 212 of these sessions. We incorporated 42 VAP risk factors at admission and routinely measured the vital characteristics and laboratory results. Five-fold cross-validation was performed to evaluate the model performance, and the model achieved an AUC of 84% in the validation, 74% sensitivity and 71% specificity 24 h after intubation. The AUC of our VAP machine learning model is nearly 25% higher than the CPIS model, and the sensitivity and specificity were also improved by almost 14% and 15%, respectively. Conclusions We developed and internally validated an automated model for VAP prediction using the MIMIC-III cohort. The VAP prediction model achieved high performance based on its AUC, sensitivity and specificity, and its performance was superior to that of the CPIS model. External validation and prospective interventional or outcome studies using this prediction model are envisioned as future work. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02031-w.
Collapse
Affiliation(s)
- Yingjian Liang
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China
| | - Chengrui Zhu
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China
| | - Cong Tian
- Philips Research China, 5F Building A2, 718 Ling Shi Road, Jing An District, Shanghai, 200072, China
| | - Qizhong Lin
- Philips Research China, 5F Building A2, 718 Ling Shi Road, Jing An District, Shanghai, 200072, China
| | - Zhiliang Li
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China
| | - Zhifei Li
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China
| | - Dongshu Ni
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China
| | - Xiaochun Ma
- Department of Critical Care Medicine, The First Hospital of China Medical University, North Nanjing Street 155, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
13
|
Lenárt S, Lenárt P, Knopfová L, Kotasová H, Pelková V, Sedláková V, Vacek O, Pokludová J, Čan V, Šmarda J, Souček K, Hampl A, Beneš P. TACSTD2 upregulation is an early reaction to lung infection. Sci Rep 2022; 12:9583. [PMID: 35688908 PMCID: PMC9185727 DOI: 10.1038/s41598-022-13637-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
TACSTD2 encodes a transmembrane glycoprotein Trop2 commonly overexpressed in carcinomas. While the Trop2 protein was discovered already in 1981 and first antibody–drug conjugate targeting Trop2 were recently approved for cancer therapy, the physiological role of Trop2 is still not fully understood. In this article, we show that TACSTD2/Trop2 expression is evolutionarily conserved in lungs of various vertebrates. By analysis of publicly available transcriptomic data we demonstrate that TACSTD2 level consistently increases in lungs infected with miscellaneous, but mainly viral pathogens. Single cell and subpopulation based transcriptomic data revealed that the major source of TACSTD2 transcript are lung epithelial cells and their progenitors and that TACSTD2 is induced directly in lung epithelial cells following infection. Increase in TACSTD2 expression may represent a mechanism to maintain/restore epithelial barrier function and contribute to regeneration process in infected/damaged lungs.
Collapse
Affiliation(s)
- Sára Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic
| | - Peter Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic.,Faculty of Science, Research Centre for Toxic Compounds in the Environment, Masaryk University, Brno, Czech Republic.,Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Hana Kotasová
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vendula Pelková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Sedláková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ondřej Vacek
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jana Pokludová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic
| | - Vladimír Čan
- Department of Surgery, University Hospital Brno, Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Aleš Hampl
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| |
Collapse
|
14
|
Chen L, Chen Y, Huang J, Zhang J. LncRNA LINC00707 serves as a sponge of miR-382-5p to alleviate lipopolysaccharide (LPS)-induced WI-38 cell injury through upregulating NKAP in infantile pneumonia. Autoimmunity 2022; 55:328-338. [PMID: 35593504 DOI: 10.1080/08916934.2022.2062594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Infantile pneumonia (IP) is an acute lower respiratory infection that imposes a heavy burden on children's health. Increasing evidence has demonstrated that long non-coding RNA (lncRNA) LINC00707 participates in the regulation of the pneumonia process. Cell proliferative ability and apoptosis were measured using Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. Bcl-2 related X protein (Bax), NF-kB activating protein (NKAP), p-P65, P65, p-IκBα, and IκBα protein levels were detected using western blot assay. The binding between miR-382-5p and LINC00707 or NKAP was predicted by starBase v2.0 and then verified by a dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. LINC00707 and NKAP levels were increased, and miR-382-5p was decreased in LPS-stimulated WI-38 cells. Furthermore, the silencing of LINC00707 could abrogate LPS-engendered WI-38 cell proliferation, apoptosis, and oxidative stress. LINC00707 deficiency could relieve LPS-triggered WI-38 cell damage by regulating the miR-382-5p/NKAP axis, providing a new therapeutic strategy for IP treatment.
Collapse
Affiliation(s)
- Lu Chen
- Department of Respiratory medicine, Hunnan Children's Hospital, Changsha, China
| | - Yanping Chen
- Department of Respiratory medicine, Hunnan Children's Hospital, Changsha, China
| | - Jianbao Huang
- Department of Respiratory medicine, Hunnan Children's Hospital, Changsha, China
| | - Jiyan Zhang
- Department of Respiratory medicine, Hunnan Children's Hospital, Changsha, China
| |
Collapse
|
15
|
Huangkui Capsule Attenuates Lipopolysaccharide-Induced Acute Lung Injury and Macrophage Activation by Suppressing Inflammation and Oxidative Stress in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2021:6626483. [PMID: 35528830 PMCID: PMC9068299 DOI: 10.1155/2021/6626483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 07/28/2021] [Accepted: 08/28/2021] [Indexed: 01/19/2023]
Abstract
Background Huangkui capsule (HKC) comprises the total flavonoid extract of flowers of Abelmoschus manihot (L.) Medicus. This study aimed to explore the effects of HKC on lipopolysaccharide- (LPS-) induced acute lung injury (ALI) in mice and LPS-stimulated RAW 264.7 cells. Methods Enzyme-linked immunosorbent assay, histopathology, spectrophotometry, and quantitative real-time polymerase chain reaction were used for the assessments. Statistical analysis was performed using a one-way analysis of variance. Results LPS significantly increased lung inflammation, neutrophil infiltration, and oxidative stress and downregulated lung miR-451 expression. Treatment with HKC dramatically, reduced the total cell count in the bronchoalveolar lavage fluid (BALF), and inhibited myeloperoxidase activity in the lung tissues 24 h after LPS challenge. Histopathological analysis demonstrated that HKC attenuated LPS-induced tissue oedema and neutrophil infiltration in the lung tissues. Additionally, the concentrations of tumour necrosis factor- (TNF-) α and interleukin- (IL-) 6 in BALF and IL-6 in the plasma reduced after HKC administration. Moreover, HKC could enhance glutathione peroxidase and catalase activities and upregulate the expression of miR-451 in the lung tissues. In vitro experiments revealed that HKC inhibited the production of nitric oxide, TNF-α, and IL-6 in LPS-induced RAW 264.7 cells and mouse primary peritoneal macrophages. Additionally, HKC downregulated LPS-induced transcription of TNF-α and IL-6 in RAW 264.7 cells. Conclusions These findings suggest that HKC has anti-inflammatory and antioxidative effects that may protect mice against LPS-induced ALI and macrophage activation.
Collapse
|
16
|
Cao J, Liu M, Feng S, Li Y, Zheng K. Glaucocalyxin A alleviates lipopolysaccharide‑induced inflammation and apoptosis in pulmonary microvascular endothelial cells and permeability injury by inhibiting STAT3 signaling. Exp Ther Med 2022; 23:313. [PMID: 35369532 PMCID: PMC8943557 DOI: 10.3892/etm.2022.11242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/29/2021] [Indexed: 11/05/2022] Open
Abstract
Glaucocalyxin A (GLA), an ent-kauranoid diterpene derived from Rabdosia japonica var. glaucocalyx, possesses antibacterial, anti-oxidative and anti-neuroinflammatory properties. The present study aimed to investigate the potential mechanisms underlying GLA in the pathogenesis of pneumonia. Human pulmonary microvascular endothelial cells (hPMVECs) treated with lipopolysaccharide (LPS) were treated with GLA, followed by the detection of cell viability, inflammation, apoptosis and cell permeability. Furthermore, the protein expression levels of apoptosis- and permeability-associated proteins were determined using western blot analysis. Following treatment with a signal transducer and activator of transcription 3 (STAT3) activator, the protein expression levels of STAT3 and endoplasmic reticulum stress-associated proteins were determined, to confirm whether STAT3 signaling was mediated by GLA. Lastly, the mRNA expression level of inflammatory cytokines, apoptosis and permeability injury were also determined following treatment with a STAT3 activator. The results revealed that GLA ameliorated inflammation, apoptosis and permeability injury in LPS-induced hPMVECs. Following treatment with a STAT3 activator, the therapeutic effects of GLA on LPS-induced hPMVECs were abrogated. In conclusion, GLA alleviated LPS-induced inflammation, apoptosis and permeability injury in hPMVECs by inhibiting STAT3 signaling, which highlighted the potential therapeutic value of GLA in the treatment of pneumonia.
Collapse
Affiliation(s)
- Jianwei Cao
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Meiling Liu
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Shufang Feng
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Yingying Li
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Kaijun Zheng
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| |
Collapse
|
17
|
Su R, Zhang Y, Zhang J, Wang H, Luo Y, Chan HF, Tao Y, Chen Z, Li M. Nanomedicine to advance the treatment of bacteria-induced acute lung injury. J Mater Chem B 2021; 9:9100-9115. [PMID: 34672317 DOI: 10.1039/d1tb01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacteria-induced acute lung injury (ALI) is associated with a high mortality rate due to the lack of an effective treatment. Patients often rely on supportive care such as low tidal volume ventilation to alleviate the symptoms. Nanomedicine has recently received much attention owing to its premium benefits of delivering drugs in a sustainable and controllable manner while minimizing the potential side effects. It can effectively improve the prognosis of bacteria-induced ALI through targeted delivery of drugs, regulation of multiple inflammatory pathways, and combating antibiotic resistance. Hence, in this review, we first discuss the pathogenesis of ALI and its potential therapeutics. In particular, the state-of-the-art nanomedicines for the treatment of bacteria-induced ALI are highlighted, including their administration routes, in vivo distribution, and clearance. Furthermore, the available bacteria-induced ALI animal models are also summarized. In the end, future perspectives of nanomedicine for ALI treatment are proposed.
Collapse
Affiliation(s)
- Ruonan Su
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca 14853, USA
| | - Jiabin Zhang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haixia Wang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhuanggui Chen
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
18
|
Wang J, Chen Z, Feng X, Yin L. Shikonin ameliorates injury and inflammatory response of LPS-stimulated WI-38 cells via modulating the miR-489-3p/MAP2K1 axis. ENVIRONMENTAL TOXICOLOGY 2021; 36:1775-1784. [PMID: 34089293 DOI: 10.1002/tox.23298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 05/16/2021] [Indexed: 06/12/2023]
Abstract
Pneumonia is an inflammatory disease induced by infection with different pathogens. Currently, multiple preclinical studies have revealed that shikonin, a natural naphthoquinone, can mitigate lipopolysaccharide (LPS)-induced inflammation, but its underlying mechanism in pneumonia remains unknown. This research was designed to explore the function and regulatory mechanism of shikonin in LPS-induced cell injury and inflammation in WI-38 cells. In-vitro model of pneumonia was constructed by treating WI-38 cells with LPS. Expression of miR-489-3p and MAP2K1 was tested by RT-qPCR and (or) Western blot analysis. Cell viability was examined by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide assay. The productions of pro-inflammatory cytokines were determined by enzyme-linked immunosorbent assays. Cell apoptosis was detected by Western blot and flow cytometry analysis. In the current study, LPS induced WI-38 cell damage by inhibiting cell viability and promoting cell apoptosis and inflammation. Shikonin ameliorated LPS-induced cell injury and elevated miR-489-3p expression. LPS-induced inflammatory injury was further mitigated by upregulation of miR-489-3p. In addition, MAP2K1, the target of miR-489-3p, was upregulated by LPS. Furthermore, upregulation of MAP2K1 reversed the influence of shikonin and miR-489-3p mimics on LPS-induced cell injury and inflammation. This study revealed that shikonin protected WI-38 cells against LPS-induced cell injury and inflammatory response by regulating the miR-489-3p/MAP2K1 axis, thus affecting the progression of pneumonia.
Collapse
Affiliation(s)
- Jinchun Wang
- Department of Pharmacy, Jiangsu Health vocational college, Nanjing 211800, Jiangsu, China
| | - Zhujing Chen
- Department of Outpatient, Jurong People's Hospital, Zhenjiang 212400, Jiangsu, China
| | - Xiaojing Feng
- Department of Comprehensive ICU, Luoyang Central Hospital, Luoyang 471009, Henan, China
| | - Lu Yin
- Department of Comprehensive ICU, Luoyang Central Hospital, Luoyang 471009, Henan, China
| |
Collapse
|
19
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
20
|
Kao TI, Chen PJ, Wang YH, Tseng HH, Chang SH, Wu TS, Yang SH, Lee YT, Hwang TL. Bletinib ameliorates neutrophilic inflammation and lung injury by inhibiting Src family kinase phosphorylation and activity. Br J Pharmacol 2021; 178:4069-4084. [PMID: 34131920 PMCID: PMC8518616 DOI: 10.1111/bph.15597] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/07/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overactivation is crucial in the pathogenesis of acute lung injury (ALI). Bletinib (3,3'-dihydroxy-2',6'-bis(p-hydroxybenzyl)-5-methoxybibenzyl), a natural bibenzyl, extracted from the Bletilla plant, exhibits anti-inflammatory, antibacterial, and antimitotic effects. In this study, we evaluated the therapeutic effects of bletinib in human neutrophilic inflammation and LPS-mediated ALI in mice. EXPERIMENTAL APPROACH In human neutrophils activated with the formyl peptide (fMLP), we assessed integrin expression, superoxide anion production, degranulation, neutrophil extracellular trap (NET) formation, and adhesion through flow cytometry, spectrophotometry, and immunofluorescence microscopy. Immunoblotting was used to measure phosphorylation of Src family kinases (SFKs) and downstream proteins. Finally, a LPS-induced ALI model in male BALB/c mice was used to investigate the potential therapeutic effects of bletinib treatment. KEY RESULTS In activated human neutrophils, bletinib reduced degranulation, respiratory burst, NET formation, adhesion, migration, and integrin expression; suppressed the enzymic activity of SFKs, including Src, Lyn, Fgr, and Hck; and inhibited the phosphorylation of SFKs as well as Vav and Bruton's tyrosine kinase (Btk). In mice with ALI, the pulmonary sections demonstrated considerable amelioration of prominent inflammatory changes, such as haemorrhage, pulmonary oedema, and neutrophil infiltration, after bletinib treatment. CONCLUSION AND IMPLICATIONS Bletinib regulates neutrophilic inflammation by inhibiting the SFK-Btk-Vav pathway. Bletinib ameliorates LPS-induced ALI in mice. Further biochemical optimisation of bletinib may be a promising strategy for the development of novel therapeutic agents for inflammatory diseases.
Collapse
Affiliation(s)
- Ting-I Kao
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jen Chen
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Hui Tseng
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsin Chang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tian-Shung Wu
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Sien-Hung Yang
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Yen-Tung Lee
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Cosmetic Science, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Chinese Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
21
|
Chen C, Zhang H, Ge M, Ye J, Li R, Wang D. LncRNA NEAT1 acts as a key regulator of cell apoptosis and inflammatory response by the miR-944/TRIM37 axis in acute lung injury. J Pharmacol Sci 2021; 145:202-212. [PMID: 33451755 DOI: 10.1016/j.jphs.2020.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Acute lung injury (ALI), a common complication of sepsis, is characterized by the impairment and injury of pulmonary function. The nuclear factor kappa-B (NF-κB) pathway is activated in ALI. Tripartite motif-containing 37 (TRIM37) can activate the NF-κB pathway and is closely associated with inflammation. The purpose of our study is to reveal the role of TRIM37 in ALI. The present study revealed that TRIM37 presented high levels in lung tissues of ALI mice, and knockdown of TRIM37 alleviated lipopolysaccharide (LPS)-induced lung injury, inflammatory response, and cell apoptosis in vivo. In addition, knockdown of TRIM37 inhibited the inflammatory response, and cell apoptosis of LPS-treated WI-38 cells. Mechanistically, miR-944 was identified to bind with and negatively regulate TRIM37. Furthermore, NEAT1 was indicated to act as a competitive endogenous RNA to promote TRIM37 expression by sequestering miR-944. Detailly, NEAT1 bound with miR-944, negatively modulated miR-944 expression, and positively modulated TRIM37 expression. The rescue assays suggested that overexpression of TRIM37 rescued the influence of NEAT1 knockdown on cell apoptosis and inflammatory response. Overall, NEAT1 facilitated cell apoptosis and inflammatory response of WI-38 cells by the miR-944/TRIM37 axis in sepsis-induced ALI, implying that NEAT1 may provide a novel insight for the treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Haitao Zhang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Graduate School of Peking Union Medical College, Nanjing, 210008, China
| | - Min Ge
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Jiaxin Ye
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Ruisha Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Dongjin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing 210008, Jiangsu, China.
| |
Collapse
|
22
|
Hajjar WM, Eldawlatly A, Alnassar SA, Ahmed I, Alghamedi A, Shakoor Z, Alrikabi AC, Hajjar AW, Ahmad AE. The effect of low versus high tidal volume ventilation on inflammatory markers in animal model undergoing lung ventilation: A prospective study. Saudi J Anaesth 2021; 15:1-6. [PMID: 33824635 PMCID: PMC8016054 DOI: 10.4103/sja.sja_650_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/30/2020] [Accepted: 07/05/2020] [Indexed: 11/23/2022] Open
Abstract
Background and Aims: Mechanical ventilation (MV) with high tidal volume (Vt.) may induce or aggravate lung injury in critically ill patients. It might also cause an overwhelming systemic inflammation leading to acute lung injury (ALI), diffuse alveolar damage (DAD) and multiple organ failure (MOF) with subsequent high mortality. The objective of this study was to compare the effects of different Vt. on the inflammatory markers of the broncho-alveolar lavage (BAL) fluid and lung biopsy in a group of animal model (Beagle dogs). Methods: A two-phased prospective study involving 30 Beagle dogs (15 dogs/phase), each phase divided into three groups (each 5 dogs/group). In the first phase each group received MV with Vt. of 8 (low), 10 (normal, control group), and 12 (high) ml/kg body weight (b.w.) respectively. BAL fluid was obtained at the time of induction of anesthesia immediately following tracheal intubation and one hour later following MV to count the macrophages, neutrophils and lymphocytes. In the second phase of the experiment, in addition to obtaining (BAL) fluid similar to the phase one, mini thoracotomy and lung biopsy obtained from the upper lobe of the right lung at same timings for histopathological examination study. Mann-Whitney-Wilcoxon test was used for statistical analysis of the data obtained. Results: BAL fluid analysis showed increase in the counts of macrophages and lymphocytes with Vt. of 12 ml/kg b.w. compared to the control group (10 ml/kg b.w.) (P < 0.05). in the second phase, similar findings obtained. The histopathological study of the lung tissue obtained in the second phase of the study from the group that received a high Vt. of 12 ml/kg b.w. showed significant inflammatory changes with presence of neutrophil infiltration and edema in the bronchial wall compared to the control group (10 ml/kg b.w.) (P < 0.05). Conclusions: The use of high Vt. in ventilated animal lung model may increase the risk of inflammation and subsequent damage in healthy lungs, these findings may help physicians to avoid using high Vt. in short-term mechanically ventilated patients in the operating room setting.
Collapse
Affiliation(s)
- Waseem M Hajjar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdelazeem Eldawlatly
- Department of Anesthesia, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sami A Alnassar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Iftikhar Ahmed
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Alaa Alghamedi
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Zahid Shakoor
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ammar C Alrikabi
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Adnan W Hajjar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulaziz Ejaz Ahmad
- Department of Anesthesia, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
23
|
Xu L, Song Q, Ouyang Z, Zhang X, Zhang C. let7f‑5p attenuates inflammatory injury in i n vitro pneumonia models by targeting MAPK6. Mol Med Rep 2020; 23:95. [PMID: 33300070 PMCID: PMC7723174 DOI: 10.3892/mmr.2020.11734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Pneumonia accounts for ~1.3 million mortalities in children per year worldwide. MicroRNAs are implicated in several diseases, including cancer and pneumonia; however, the role of let7f-5p in pneumonia is not completely understood. In the present study, lipopolysaccharide (LPS) was used to establish an in vitro pneumonia model in A549 and WI-38 cells. The reverse transcription-quantitative PCR (RT-qPCR) and western blotting results demonstrated that let7f-5p expression levels were significantly decreased, whereas MAPK6 expression levels were significantly increased in the peripheral venous blood of patients with pneumonia and in LPS-induced A549 and WI-38 cells compared with healthy volunteers and control cells, respectively. Furthermore, the dual-luciferase reporter assay demonstrated that let7f-5p targeted the 3′-untranslated region of MAPK6. The ELISA and RT-qPCR results demonstrated that let7f-5p mimic ameliorated LPS-induced inflammatory injury in A549 and WI-38 cells, as demonstrated by decreased expression levels of proinflammatory cytokines, including TNF-α and IL-6. In addition, the Cell Counting Kit-8 assay results indicated that let7f-5p mimic ameliorated LPS-induced reductions in cell viability, and the western blotting results demonstrated that let7f-5p mimic reversed LPS-induced activation of the STAT3 signaling pathway. Notably, the aforementioned let7f-5p-mediated effects were reversed by MAPK6 overexpression. Collectively, the results of the present study suggested that let7f-5p inhibited inflammation by targeting MAPK6 in the in vitro pneumonia model, thus let7f-5p may serve as a potential novel therapeutic target for pneumonia.
Collapse
Affiliation(s)
- Lin Xu
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| | - Qingying Song
- Department of Anesthesiology, The Second Affiliated Hospital of Guizhou College of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Zhanghong Ouyang
- Development Planning Division, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xiangyan Zhang
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| | - Cheng Zhang
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
24
|
Acetylharpagide Protects Mice from Staphylococcus Aureus-Induced Acute Lung Injury by Inhibiting NF-κB Signaling Pathway. Molecules 2020; 25:molecules25235523. [PMID: 33255656 PMCID: PMC7728067 DOI: 10.3390/molecules25235523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/15/2020] [Accepted: 11/24/2020] [Indexed: 12/30/2022] Open
Abstract
Staphylococcus aureus (S. aureus)-induced acute lung injury (ALI) is a serious disease that has a high risk of death among infants and teenagers. Acetylharpagide, a natural compound of Ajuga decumbens Thunb. (family Labiatae), has been found to have anti-tumor, anti-inflammatory and anti-viral effects. This study investigates the therapeutic effects of acetylharpagide on S. aureus-induced ALI in mice. Here, we found that acetylharpagide alleviated S. aureus-induced lung pathological morphology damage, protected the pulmonary blood-gas barrier and improved the survival of S. aureus-infected mice. Furthermore, S. aureus-induced myeloperoxidase (MPO) activity of lung homogenate and pro-inflammatory factors in bronchoalveolar lavage (BAL) fluid were suppressed by acetylharpagide. Mechanically, acetylharpagide inhibited the interaction between polyubiquitinated receptor interacting protein 1 (RIP1) and NF-κB essential modulator (NEMO), thereby suppressing NF-κB activity. In summary, these results show that acetylharpagide protects mice from S. aureus-induced ALI by suppressing the NF-κB signaling pathway. Acetylharpagide is expected to become a potential treatment for S. aureus-induced ALI.
Collapse
|
25
|
Wang H, Wen X, Yan M, Chang M, Zhang G, Peng W, Wu Y, Shen Y, Zhou J, Li H. The role of perilipin 2 in Pseudomonas aeruginosa pulmonary infection. Respir Physiol Neurobiol 2020; 281:103497. [DOI: 10.1016/j.resp.2020.103497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/23/2020] [Accepted: 07/14/2020] [Indexed: 01/24/2023]
|
26
|
Sikyungbanha-Tang Suppressing Acute Lung Injury in Mice Is Related to the Activation of Nrf2 and TNFAIP3. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8125758. [PMID: 32256655 PMCID: PMC7102461 DOI: 10.1155/2020/8125758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/13/2020] [Accepted: 02/06/2020] [Indexed: 12/30/2022]
Abstract
Sikyungbanha-Tang (SKBHT) is a Chinese traditional medicine popularly prescribed to patients with respiratory inflammatory symptoms in Korea. Although the Korea Food and Drug Administration approved SKBHT as a therapeutics for relieving the symptoms, experimental evidence for SKBHT suppressing inflammation is scarce. Here, we presented evidence that SKBHT can suppress inflammation in an acute lung injury (ALI) mouse model and explored the possible underlying mechanisms of SKBHT's anti-inflammatory activity. Single intratracheal (i.t.) injection of SKBHT (1 mg/kg or 10 mg/kg body weight) into mouse lungs decreased prototypic features of lung inflammation found in ALI, such as a high level of proinflammatory cytokines, neutrophil infiltration, and the formation of hyaline membrane, which were induced by a single i.t. LPS (2 mg/kg body weight). When added to a murine macrophage RAW 264.7 cells, SKBHT activated an anti-inflammatory factor Nrf2, increasing the expression of genes regulated by Nrf2. SKBHT suppressed the ubiquitination of Nrf2, suggesting that SKBHT increases the level of and thus activates Nrf2 by blunting the ubiquitin-dependent degradation of Nrf2. SKBHT induced the expression of tumor necrosis factor α-induced protein 3 (TNFAIP3), an ubiquitin-modulating protein that suppresses various cellular signals to NF-κB. Concordantly, SKBHT suppressed NF-κB activity and the expression of inflammatory cytokine genes regulated by NF-κB. Given that Nrf2 and TNFAIP3 are involved in regulating inflammation, our results suggest that SKBHT suppresses inflammation in the lung, the effect of which is related to SKBHT activating Nrf2 and TNFAIP3.
Collapse
|
27
|
Zhang J, Xiang J, Liu T, Wang X, Tang Y, Liang Y. miR-495 targets ROCK1 to inhibit lipopolysaccharides-induced WI-38 cells apoptosis and inflammation. Kaohsiung J Med Sci 2020; 36:607-614. [PMID: 32237054 DOI: 10.1002/kjm2.12210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/12/2020] [Indexed: 12/20/2022] Open
Abstract
Pneumonia is an inflammatory disease with leading mortality rate in children. It has been well established that microRNAs (miRNAs) have been regarded as critical regulator in acute lung injury. We intended to explore the effect and underlying mechanism of miR-495 on lipopolysaccharides (LPS)-induced WI-38 cells. Here, we first found that miR-495 was downregulated in serum of patients with acute stage pneumonia. To establish cell model of acute pneumonia, WI-38 cells were treated with 20 μg/mL LPS, and qRT-PCR analysis also confirmed the downregulation of miR-495 in LPS-induced WI-38 cells. Data from MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) and flow cytometry assays showed that the decreased cell viability and induced cell apoptosis by LPS treatment were also reversed by miR-495 over-expression. Moreover, miR-495 inhibited expression of associated inflammatory factors, which were induced by LPS treatment. Second, ROCK1 (rho-associated, coiled-coil-containing protein kinase 1) was identified as functional target gene of miR-495, whose expression was decreased by miR-495. Mechanically, combination of miR-495 and ROCK1 over-expression reversed the influence of miR-495 on LPS-induced inflammation, viability, and apoptosis. In conclusion, our findings indicated that miR-495 inhibited LPS-induced inflammation injury and apoptosis in WI-38 cells via targeting ROCK1, which would shed light on therapeutic schedule in acute pneumonia.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Clinical Laboratory, Wuhan Medical Treatment Center, Wuhan City, Hubei Province, China
| | - Jie Xiang
- Department of Clinical Laboratory, Wuhan Medical Treatment Center, Wuhan City, Hubei Province, China
| | - Ting Liu
- Department of Clinical Laboratory, Wuhan Medical Treatment Center, Wuhan City, Hubei Province, China
| | - Xinwei Wang
- Department of Respiratory and Critical Medicine, Hubei No.3 People's Hospital of Jianghan University, Wuhan City, Hubei Province, China
| | - Ying Tang
- Department of Clinical Laboratory, Wuhan Medical Treatment Center, Wuhan City, Hubei Province, China
| | - Yin Liang
- Department of Respiratory and Critical Medicine, Hubei No.3 People's Hospital of Jianghan University, Wuhan City, Hubei Province, China
| |
Collapse
|
28
|
Kim KH, Lee JY, Ahn S, Won R, Kim SJ, Jeong SI, Lee JJ, Kim JI, Choi JY, Joo M. The methanol extract of Guettarda speciosa Linn. Ameliorates acute lung injury in mice. BMC Complement Med Ther 2020; 20:40. [PMID: 32033557 PMCID: PMC7076890 DOI: 10.1186/s12906-020-2828-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Background Guettarda speciosa is mainly found in tropical areas in Asia. Although G. speciosa is traditionally used to treat some of the inflammatory disorders, the experimental evidence supporting the anti-inflammatory effect of G. speciosa is limited. Here, we sought to obtain evidence that G. speciosa has anti-inflammatory activity using an acute lung injury (ALI) mouse model and to explore possible underlying mechanisms for the activity. Methods The methanol extract of G. speciosa Linn. (MGS) was fingerprinted by HPLC. Cytotoxicity was determined by MTT and flow cytometer. As for an ALI mouse model, C57BL/6 mice received an intratracheal (i.t.) injection of lipopolysaccharide (LPS). The effects of MGS on lung inflammation in the ALI mice were assessed by differential cell counting and FACS of inflammatory cells and hematoxylin and eosin staining of lung tissue. Proteins were analyzed by immunoprecipitation and immunoblotting, and gene expression was by real-time qPCR. Neutrophil elastase activity was measured by ELISA. Results MGS did not cause metabolic disarray or produce reactive oxygen species that could induce cytotoxicity. Similar to ALI patients, C57BL/6 mice that received an i.t. LPS developed a high level of neutrophils, increased pro-inflammatory cytokines, and inflicted tissue damage in the lung, which was suppressed by i.t. MGS administered at 2 h after LPS. Mechanistically, MGS activated Nrf2, which was related to MGS interrupting the ubiquitin-dependent degradation of Nrf2. MGS suppressed the nuclear localization of NF-κB induced by LPS, suggesting the inhibition of NF-κB activity. Furthermore, MGS inhibited the enzymatic activity of neutrophil elastase. Conclusion MGS could suppress lung inflammation in an ALI mouse model, the effect of which could be attributed to multiple mechanisms, including the activation of Nrf2 and the suppression of NF-κB and neutrophil elastase enzymatic activity by MGS.
Collapse
Affiliation(s)
- Kyun Ha Kim
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ji Yeon Lee
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seonju Ahn
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ran Won
- Department ofs Biomedical Laboratory Science, Division of Health Sciences, Dongseo University, Busan, 47011, Republic of Korea
| | - Sang-Jun Kim
- Jeonju AgroBio-Materials Institute, Jeonju, 57810, Republic of Korea
| | - Seung-Il Jeong
- Jeonju AgroBio-Materials Institute, Jeonju, 57810, Republic of Korea
| | - Jung Ju Lee
- Department of Acupuncture and Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jong-In Kim
- Department of Acupuncture and Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jun-Yong Choi
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea. .,Department of Internal Medicine, Korean Medicine Hospital of Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Myungsoo Joo
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
29
|
Yang J, Chen Y, Jiang K, Zhao G, Guo S, Liu J, Yang Y, Deng G. MicroRNA-182 supplies negative feedback regulation to ameliorate lipopolysaccharide-induced ALI in mice by targeting TLR4. J Cell Physiol 2020; 235:5925-5937. [PMID: 32003008 DOI: 10.1002/jcp.29504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022]
Abstract
Acute lung injury (ALI), characterized by increased excessive pulmonary inflammation, is a pervasive inflammatory disease with clinically high incidence. MicroRNA (miRNAs) have been associated with the progression of multiple diseases and are regarded as novel regulators of inflammation. However, it remains largely unknown whether the miRNAs-mediated regulatory mechanism has an effect on lipopolysaccharide (LPS)-induced inflammation in ALI. We discovered that miR-182 distinctly lessened expression in the lung tissue of mice with ALI and macrophages stimulated by LPS. We also found that overexpression of miR-182 significantly cut down the secretion of inflammatory cytokines, while this change was reversed by inhibition of miR-182. In addition, miR-182 suppressed the activation of NF-κB by targeting TLR4 expression. And it was confirmed that miR-182 directly regulated TLR4 expression at the posttranscriptional level by binding to the 3'-UTR of TLR4. Together, these data suggested that inhibition of TLR4 expression assuaged LPS-stimulated inflammation through negative feedback regulation of miR-182.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Junfeng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
30
|
Zhang J, Mao F, Zhao G, Wang H, Yan X, Zhang Q. Long non-coding RNA SNHG16 promotes lipopolysaccharides-induced acute pneumonia in A549 cells via targeting miR-370-3p/IGF2 axis. Int Immunopharmacol 2019; 78:106065. [PMID: 31841752 DOI: 10.1016/j.intimp.2019.106065] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/23/2019] [Accepted: 11/17/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pneumonia is an infectious lung inflammation in children with high mortality and morbidity rates. Small nucleolar RNA host gene 16 (SNHG16) has been verified to accelerate the progression of acute pneumonia. However, the role of SNHG16 in acute pneumonia has not yet been fully elucidated. The study was aimed to explore the regulatory mechanism of SNHG16 in LPS-induced acute pneumonia in A549 cells. METHODS The levels of SNHG16, miR-370-3p and IGF2 in serum samples and LPS-induced A549 cells were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The cell viability and apoptosis of A549 cells were examined by Cell Counting Kit-8 (CCK-8) assay and flow cytometer, respectively. The levels of interleukin 1β (IL-1β), interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). The binding relationships among SNHG16, miR-370-3p and IGF2 were predicted by online database and verified by Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The protein levels of IGF2 were tested by Western blot. RESULTS SNHG16 and IGF2 were upregulated while miR-370-3p was downregulated in serum of acute pneumonia patients and LPS-induced A549 cells. SNHG16 regulated proliferation, apoptosis and inflammatory cytokines by inhibiting miR-370-3p in LPS-induced A549 cells. MiR-370-3p targeted IGF2 and inhibited LPS-induced inflammatory injury via IGF2 in A549 cells. Furthermore, SNHG16 was verified to promote IGF2 expression by sponging miR-370-3p in A549 cells. CONCLUSION SNHG16 impeded cell viability and promoted apoptosis, inflammatory injury by targeting IGF2 mediated by miR-370-3p in LPS-induced A549 cells.
Collapse
Affiliation(s)
- Ju Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Fengxia Mao
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Gai Zhao
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Haixia Wang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiaomin Yan
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Qian Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
31
|
Yu X, Qian X, Sun R, Yang B, Zheng H, Jiang P, Li X. MiR-370-3p targets TLR4 to regulate LPS-induced acute pneumonia in WI-38 cells. Panminerva Med 2019; 64:121-122. [PMID: 31663304 DOI: 10.23736/s0031-0808.19.03712-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Xi Yu
- Department of Respiratory, Tianjin First Central Hospital, Tianjin, China
| | - Xuejiao Qian
- Department of Respiratory, Tianjin First Central Hospital, Tianjin, China
| | - Rongfei Sun
- Department of Respiratory, Tianjin First Central Hospital, Tianjin, China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Hong Zheng
- Department of Respiratory, Tianjin First Central Hospital, Tianjin, China
| | - Ping Jiang
- Department of Respiratory, Tianjin First Central Hospital, Tianjin, China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, China -
| |
Collapse
|
32
|
Yang J, Chen Y, Jiang K, Yang Y, Zhao G, Guo S, Deng G. MicroRNA-106a Provides Negative Feedback Regulation in Lipopolysaccharide-Induced Inflammation by targeting TLR4. Int J Biol Sci 2019; 15:2308-2319. [PMID: 31595149 PMCID: PMC6775322 DOI: 10.7150/ijbs.33432] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/03/2019] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical disease with high incidence and mortality rate, which is characterized by severe inflammatory response and tissues damage. MicroRNAs (miRNAs) have been regarded as novel regulators of inflammation, and play an important role in various inflammatory diseases. However, it remains unknown whether the regulatory mechanisms mediated by miR-106a is involved in LPS-induced ALI. In this study, we found that expression of miR-106a was significantly decreased in lung tissues of ALI mice and LPS-stimulated macrophages. We also revealed that over-expression of miR-106a significantly decreased the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α, whereas this effect was reversed by the inhibition of miR-106a. Moreover, miR-106a inhibits NF-κB activation by targeting TLR4 expression. We further demonstrated that miR-106a inhibited TLR4 expression via binding directly to the 3'-UTR of TLR4. Taken together, the results of the present study illuminated that miR-106a is a negative feedback regulator in LPS-stimulated inflammation through TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| |
Collapse
|
33
|
Zhang Y, Zhu Y, Gao G, Zhou Z. Knockdown XIST alleviates LPS-induced WI-38 cell apoptosis and inflammation injury via targeting miR-370-3p/TLR4 in acute pneumonia. Cell Biochem Funct 2019; 37:348-358. [PMID: 31066476 PMCID: PMC6618287 DOI: 10.1002/cbf.3392] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
Pneumonia is an inflammatory disease that occurs in the lungs associated with pathogens or other factors. It has been well established that long noncoding RNA X inactivate-specific transcript (XIST) is involved in several cancers. The present study focused on the effect and detailed mechanism of XIST in lipopolysaccharide (LPS)-induced injury in pneumonia. Here, XIST was silenced by transfection with XIST-targeted siRNA, and then, mRNA expression, cell viability, apoptosis, and protein expression were, respectively, assessed by qRT-PCR, CCK-8, flow cytometry, and Western blotting. Luciferase reporter, RIP, and RNA pull-down assays were used to detect the combination of miR-370-3p and XIST. Besides, the tested proinflammatory factors were analysed by qRT-PCR and Western blot, and their productions were quantified by ELISA. The results showed that XIST expression was robustly increased in serum of patients with acute-stage pneumonia and LPS-induced WI-38 human lung fibroblasts cells. Functional analyses demonstrated that knockdown of XIST remarkably alleviated LPS-induced cell injury through increasing cell viability and inhibiting apoptosis and inflammatory cytokine levels. Mechanistically, XIST functioned as a competitive endogenous RNA (ceRNA) by effectively binding to miR-370-3p and then restoring TLR4 expression. More importantly, miR-370-3p inhibitor abolished the function of XIST knockdown on cell injury and JAK/STAT and NF-κB pathways. Taken together, XIST may be involved in progression of cell inflammatory response, and XIST/miR-370-3p/TLR4 axis thus may shed light on the development of novel therapeutics to the treatment of acute stage of pneumonia. SIGNIFICANCE OF THE STUDY: Our study demonstrated that XIST was highly expressed in patients with acute stage of pneumonia. Knockdown of XIST remarkably alleviated LPS-induced cell injury through increasing cell viability and inhibiting apoptosis and inflammatory cytokine levels through regulating JAK/STAT and NF-κB pathways.
Collapse
Affiliation(s)
- Yena Zhang
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Yuyin Zhu
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Guosheng Gao
- Department of Laboratory, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| | - Zhiming Zhou
- Department of Pulmonary Medicine, HwaMei HospitalUniversity Of Chinese Academy Of SciencesNingboChina
| |
Collapse
|
34
|
Long noncoding RNA SNHG16 targets miR-146a-5p/CCL5 to regulate LPS-induced WI-38 cell apoptosis and inflammation in acute pneumonia. Life Sci 2019; 228:189-197. [PMID: 31071307 DOI: 10.1016/j.lfs.2019.05.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/29/2019] [Accepted: 05/05/2019] [Indexed: 11/21/2022]
Abstract
AIMS Aberrant expression of the lncRNA small nucleolar RNA host gene 16 (SNHG16) has been researched in multiple cancers and inflammatory diseases. This study was intended to investigate the effect of SNHG16 in vitro model of pneumonia and explore the potential mechanism. MAIN METHODS The LPS-induced pulmonary injury model was established in WI-38 human lung fibroblasts cells. SNHG16 and miR-146a-5p expression levels were altered by transfection assay and were evaluated by qRT-PCR. Cell viability and apoptosis were respectively assessed by CCK-8 assay and flow cytometry analysis. The combination of miR-146a-5p and SNHG16 were demonstrated by luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay. Associated inflammatory factors expression levels and productions were determined by qRT-PCR, western blotting and Enzyme-linked immunosorbent (ELISA) assay, respectively. Main proteins related apoptosis, c-Jun N-terminal kinase (JNK) pathway and nuclear factor (NF)-κB pathway were also analyzed by western blotting. KEY FINDINGS SNHG16 was highly expressed in serum of acute stage pneumonia patients. SNHG16 was up-regulated in LPS-treated WI-38 cell model and SNHG16 knockdown obviously mitigated LPS-induced cell injury by promoting viability, restraining apoptosis and production of inflammatory cytokines. SNHG16 functioned as a competitive endogenous RNA (ceRNA) by efficaciously binding to miR-146a-5p and then restoring CC motif chemokine ligand 5 (CCL5) expression. Besides, miR-146a-5p inhibitor abolished the function of SNHG16 knockdown on cell injury, JNK and NF-κB pathways. SIGNIFICANCE SNHG16 regulated LPS-induced inflammation injury in WI-38 cells through competitively binding miR-146a-5p with CCL5 further mediating JNK and NF-κB pathways, which sheds novel light on diagnostics and therapeutics in pneumonia.
Collapse
|
35
|
Design and synthesis novel di-carbonyl analogs of curcumin (DACs) act as potent anti-inflammatory agents against LPS-induced acute lung injury (ALI). Eur J Med Chem 2019; 167:414-425. [DOI: 10.1016/j.ejmech.2019.02.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 01/30/2023]
|
36
|
Jiang X, Chen L, Zhang Z, Sun Y, Wang X, Wei J. Protective and Therapeutic Effects of Engeletin on LPS-Induced Acute Lung Injury. Inflammation 2018; 41:1259-1265. [PMID: 29704150 DOI: 10.1007/s10753-018-0773-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) is a serious disease with morbidity and mortality in patients. Engeletin (dihydrokaempferol 3-rhamnoside) is a flavanonol glycoside. It can be found in the skin of white grapes and white wine and is widely distributed in southeast Asia. In our study, we evaluated the protective and therapeutic effects of engeletin on lipopolysaccharide (LPS)-induced ALI in animal model. We determined the level of peroxisome proliferator-activated receptor-γ (PPAR-γ), nuclear factor kappaB (NF-κB), and IκBα by western blotting. The myeloperoxidase (MPO) activity and lung wet/dry (W/D) ratio in lung tissues were also detected. Histopathological changes and the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β were determined by H&E staining and ELISA. The MPO activity and lung W/D ratio induced by LPS were attenuated by engeletin. The numbers of inflammatory cells and the levels of inflammatory cytokines in bronchoalveolar lavage fluid (BALF) were ameliorated by engeletin. Furthermore, the results also showed that engeletin significantly suppressed LPS-induced NF-κB activation. The expression of PPAR-γ was upregulated by treatment of engeletin. In conclusion, we found that engeletin had protective and therapeutic effects against LPS-induced ALI by activating PPAR-γ. Engeletin is a potentially effective agent for the treatment of lung injury.
Collapse
Affiliation(s)
- Xian Jiang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lijuan Chen
- Department of Pharmacology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacology of Pharmacy College of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuhong Sun
- Department of Pharmacology of Pharmacy College of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaobin Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jicheng Wei
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
37
|
Wang S, Gao J, Li M, Wang L, Wang Z. A facile approach for development of a vaccine made of bacterial double-layered membrane vesicles (DMVs). Biomaterials 2018; 187:28-38. [PMID: 30292939 DOI: 10.1016/j.biomaterials.2018.09.042] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/15/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022]
Abstract
Bacterial infections cause acute and chronic diseases. Antimicrobial resistance and aging-related immune weakness remain challenging in therapy of infectious diseases. Vaccines are however an alternative to prevent bacterial infections. Here we report a facile method to rapidly generate bacterium-membrane-formed nanovesicles as a vaccine using nitrogen cavitation. The vaccine is comprised of double-layered membrane vesicles (DMVs) characterized by cryo-TEM, biochemistry and proteomics, showing DMVs possess the integrity of bacterial membrane and contain a wide range of membrane proteins required for vaccination. In the mouse sepsis model induced by Pseudomonas aeruginosa, we found that DMVs can improve mouse survival after mice were immunized with DMVs. The increased adaptive immunity and unique biodistribution of DMVs were responsible for enhanced protection of bacterial infection. Our studies demonstrate that this simple and innovative approach using nitrogen cavitation would be a promising technology for vaccine developments.
Collapse
Affiliation(s)
- Sihan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jin Gao
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Mo Li
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Liguo Wang
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
38
|
Li X, Yu Y, Gorshkov B, Haigh S, Bordan Z, Weintraub D, Rudic RD, Chakraborty T, Barman SA, Verin AD, Su Y, Lucas R, Stepp DW, Chen F, Fulton DJR. Hsp70 Suppresses Mitochondrial Reactive Oxygen Species and Preserves Pulmonary Microvascular Barrier Integrity Following Exposure to Bacterial Toxins. Front Immunol 2018; 9:1309. [PMID: 29951058 PMCID: PMC6008539 DOI: 10.3389/fimmu.2018.01309] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/25/2018] [Indexed: 01/22/2023] Open
Abstract
Pneumonia is a leading cause of death in children and the elderly worldwide, accounting for 15% of all deaths of children under 5 years old. Streptococcus pneumoniae is a common and aggressive cause of pneumonia and can also contribute to meningitis and sepsis. Despite the widespread use of antibiotics, mortality rates for pneumonia remain unacceptably high in part due to the release of bacterial toxins. Pneumolysin (PLY) is a cholesterol-dependent toxin that is produced by Streptococcus, and it is both necessary and sufficient for the development of the extensive pulmonary permeability edema that underlies acute lung injury. The mechanisms by which PLY disrupts the pulmonary endothelial barrier are not fully understood. Previously, we found that reactive oxygen species (ROS) contribute to the barrier destructive effects of PLY and identified an unexpected but potent role of Hsp70 in suppressing ROS production. The ability of Hsp70 to influence PLY-induced barrier dysfunction is not yet described, and the goal of the current study was to identify whether Hsp70 upregulation is an effective strategy to protect the lung microvascular endothelial barrier from G+ bacterial toxins. Overexpression of Hsp70 via adenovirus-mediated gene transfer attenuated PLY-induced increases in permeability in human lung microvascular endothelial cells (HLMVEC) with no evidence of cytotoxicity. To adopt a more translational approach, we employed a pharmacological approach using geranylgeranylacetone (GGA) to acutely upregulate endogenous Hsp70 expression. Following acute treatment (6 h) with GGA, HLMVECs exposed to PLY displayed improved cell viability and enhanced endothelial barrier function as measured by both Electric Cell-substrate Impedance Sensing (ECIS) and transwell permeability assays compared to control treated cells. PLY promoted increased mitochondrial ROS, decreased mitochondrial oxygen consumption, and increased caspase 3 cleavage and cell death, which were collectively improved in cells pretreated with GGA. In mice, IP pretreatment with GGA 24 h prior to IT administration of PLY resulted in significantly less Evans Blue Dye extravasation compared to vehicle, indicating preserved endothelial barrier integrity and suggesting that the acute upregulation of Hsp70 may be an effective therapeutic approach in the treatment of lung injury associated with pneumonia.
Collapse
Affiliation(s)
- Xueyi Li
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yanfang Yu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephen Haigh
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Daniel Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Radu Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Scott A Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - David W Stepp
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Feng Chen
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
39
|
Jiang K, Guo S, Zhang T, Yang Y, Zhao G, Shaukat A, Wu H, Deng G. Downregulation of TLR4 by miR-181a Provides Negative Feedback Regulation to Lipopolysaccharide-Induced Inflammation. Front Pharmacol 2018. [PMID: 29535629 PMCID: PMC5834510 DOI: 10.3389/fphar.2018.00142] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Acute lung injury (ALI) is a progressive clinical disease with a high mortality rate, and characterized by an excessive uncontrolled inflammatory response. MicroRNAs (miRNAs) play a critical role in various human inflammatory diseases, and have been recognized as important regulators of inflammation. However, the regulatory mechanisms mediated by miRNAs involved in Lipopolysaccharide (LPS)-induced inflammation in ALI remain hazy. In this study, we found that miR-181a expression in the lung tissues of ALI mice and LPS-stimulated RAW 264.7 macrophages is dramatically reduced. We also show that over-expression of miR-181a significantly decreased the production of inflammatory cytokines, such as IL-1β, IL-6, and TNF-α, whereas inhibition of miR-181a reversed this decrease. Moreover, miR-181a inhibits NF-κB activation and accumulation of reactive oxygen species (ROS) by targeting TLR4 expression. We further verify that miR-181a suppresses TLR4 expression by binding directly to the 3′-UTR of TLR4. Therefore, we provide the first evidence for the negative regulation of miR-181a in LPS-induced inflammation via the suppression of ROS generation and TLR4-NF-κB pathway.
Collapse
Affiliation(s)
- Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
40
|
Pulido L, Burgos D, García Morato J, Luna CM. Does animal model on ventilator-associated pneumonia reflect physiopathology of sepsis mechanisms in humans? ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:452. [PMID: 29264369 PMCID: PMC5721223 DOI: 10.21037/atm.2017.11.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 11/21/2017] [Indexed: 11/06/2022]
Abstract
Ventilator-associated pneumonia (VAP) is the leading cause of death in critically ill patients in intensive care units. In the last 20 years, different animal models have been a valuable tool for the study of pathophysiology and phenotypic characteristics of different lung infections observed in humans, becoming an essential link between ''in vitro'' testing and clinical studies. Different animal models have been used to study the mechanism of a deregulated inflammatory response and host tissue damage of sepsis in VAP, as well as different infection parameters such as clinical, physiological, microbiological and pathological facts in several large and small mammals. In addition, the dosage of inflammatory modulators and their consequences in local and systemic inflammation, or even the administration of antibiotics, have been evaluated with very interesting results. Although some bronchial inoculation ways do not resemble the common pathophysiologic mechanisms, the experimental model of VAP induced by the inoculation of high concentrations of pathogens in mechanically ventilated animals is useful for studying the local and systemic responses of sepsis in VAP and it reproduces biological mechanisms such as acute lung injury, distress response, cardiac events and immune modulation comparable with clinical studies.
Collapse
Affiliation(s)
- Laura Pulido
- Department of Pulmonary Medicine, Experimental Surgery University Center, Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Diego Burgos
- Department of Pulmonary Medicine, Experimental Surgery University Center, Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Joaquín García Morato
- Thoracic Surgery Division, Department of Surgery, Experimental Surgery University Center, Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos M. Luna
- Department of Pulmonary Medicine, Experimental Surgery University Center, Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
41
|
Fu PK, Yang CY, Huang SC, Hung YW, Jeng KC, Huang YP, Chuang H, Huang NC, Li JP, Hsu MH, Chen JK. Evaluation of LPS-Induced Acute Lung Injury Attenuation in Rats by Aminothiazole-Paeonol Derivatives. Molecules 2017; 22:molecules22101605. [PMID: 28946699 PMCID: PMC6151495 DOI: 10.3390/molecules22101605] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/19/2022] Open
Abstract
Paeonol is a key phenolic compound in the root bark of Moutan Cortex Radicis that has been used in traditional Chinese Medicine to ameliorate inflammation. A series of aminothiazole-paeonol derivatives (APDs) were synthesized in this work and subjected to preliminary evaluation in cells followed by verification in animals. Quantification of monocyte chemotactic protein-1 (MCP-1) and interleukin-6 (IL-6) in culture media of LPS-activated A549 cells, a lung epithelial adenocarcinoma cell line, were used to investigate the anti-inflammatory capability of APDs. ALI-bearing rats were employed to verify therapeutic efficacy of APDs according to observations of total cells, protein amounts, MCP-1 and IL-6 in bronchoalveolar lavage fluid (BALF). Histopathological examinations of lung tissues were consequently applied for validation of APDs. Among these compounds, 2-(2-aminothiazol-4-yl)-5-methoxyphenol (4) had the most potent activity, showing comparable inhibition of MCP-1/IL-6 and superior elimination of neutrophil infiltration and protein exudation in lungs compared to others as well as dexamethasone. This study demonstrated a comprehensive strategy to evaluate APDs through integration of cell-based screening and animal-based verification. In order to fulfill unmet needs of treating acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), APDs introduced in this work could be promising lead compounds to develop high potent anti-inflammation agents.
Collapse
Affiliation(s)
- Pin-Kuei Fu
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
- Department of Biotechnology, Hungkuang University, Taichung 43302, Taiwan.
- School of Chinese Medicine, China Medical University, Taichung 40447, Taiwan.
| | - Chi-Yu Yang
- Animal Technology Laboratory, Agriculture Technology Research Institute, Miaoli 35053, Taiwan.
| | - Su-Chin Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
| | - Yu-Wen Hung
- Animal Technology Laboratory, Agriculture Technology Research Institute, Miaoli 35053, Taiwan.
| | - Kee-Ching Jeng
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung 43503, Taiwan.
| | - Ying-Pei Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hong Chuang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Nai-Chun Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
| | - Jui-Ping Li
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua County 50007, Taiwan.
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
- School of Dentistry, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
42
|
Ordija CM, Chiou TTY, Yang Z, Deloid GM, de Oliveira Valdo M, Wang Z, Bedugnis A, Noah TL, Jones S, Koziel H, Kobzik L. Free actin impairs macrophage bacterial defenses via scavenger receptor MARCO interaction with reversal by plasma gelsolin. Am J Physiol Lung Cell Mol Physiol 2017; 312:L1018-L1028. [PMID: 28385809 PMCID: PMC5495953 DOI: 10.1152/ajplung.00067.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 12/20/2022] Open
Abstract
Lung injury can release intracellular actin into the alveolar milieu and is also associated with increased susceptibility to secondary infections. We investigated the effect of free (extracellular) actin on lung macrophage host defense functions. Western blot analysis demonstrated free actin release into the lung lavage fluids of mouse models of ozone injury, influenza infection, and secondary pneumococcal pneumonia and in samples from patients following burn and inhalation injury. Using levels comparable with those observed in lung injury, we found that free actin markedly inhibited murine lung macrophage binding and uptake in vitro of S. pneumoniae, S. aureus, and E. coli, (e.g., S. pneumoniae, mean %inhibition, actin vs. vehicle: 85 ± 0.3 (SD); n = 22, P < .001). Similar effects were observed on the ability of primary human macrophages to bind and ingest fluorescent Saureus (~75% inhibition). Plasma gelsolin (pGSN), a protein that functions to bind and cleave actin, restored bacterial binding and uptake by both murine and human macrophages. Scavenger receptor inhibitors reduced binding of fluorescent actin by murine macrophages [fluorescence index (×10-3) after incubation with vehicle, actin, or actin + polyinosinic acid, respectively: 0.8 ± 0.7, 101.7 ± 50.7, or 52.7 ± 16.9; n = 5-6, P < 0.05]. In addition, actin binding was reduced in a MARCO/SR-AI/II-deficient cell line and by normal AMs obtained from MARCO-/- mice. After release from injured cells during lung injury, free actin likely contributes to impaired host defense by blocking scavenger receptor binding of bacteria. This mechanism for increased risk of secondary infections after lung injury or inflammation may represent another target for therapeutic intervention with pGSN.
Collapse
Affiliation(s)
- Christine M Ordija
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Terry Ting-Yu Chiou
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts.,Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang-Gung University College of Medicine, Kaohsiung, Taiwan
| | - Zhiping Yang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Glen M Deloid
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Melina de Oliveira Valdo
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Zhi Wang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Alice Bedugnis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Terry L Noah
- Department of Pediatrics, Pulmonology Division, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Samuel Jones
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Henry Koziel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Lester Kobzik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts;
| |
Collapse
|
43
|
Walsh DM, McCullough SD, Yourstone S, Jones SW, Cairns BA, Jones CD, Jaspers I, Diaz-Sanchez D. Alterations in airway microbiota in patients with PaO2/FiO2 ratio ≤ 300 after burn and inhalation injury. PLoS One 2017; 12:e0173848. [PMID: 28358811 PMCID: PMC5373524 DOI: 10.1371/journal.pone.0173848] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Injury to the airways after smoke inhalation is a major mortality risk factor in victims of burn injuries, resulting in a 15-45% increase in patient deaths. Damage to the airways by smoke may induce acute respiratory distress syndrome (ARDS), which is partly characterized by hypoxemia in the airways. While ARDS has been associated with bacterial infection, the impact of hypoxemia on airway microbiota is unknown. Our objective was to identify differences in microbiota within the airways of burn patients who develop hypoxemia early after inhalation injury and those that do not using next-generation sequencing of bacterial 16S rRNA genes. RESULTS DNA was extracted from therapeutic bronchial washings of 48 patients performed within 72 hours of hospitalization for burn and inhalation injury at the North Carolina Jaycee Burn Center. DNA was prepared for sequencing using a novel molecule tagging method and sequenced on the Illumina MiSeq platform. Bacterial species were identified using the MTToolbox pipeline. Patients with hypoxemia, as indicated by a PaO2/FiO2 ratio ≤ 300, had a 30% increase in abundance of Streptococcaceae and Enterobacteriaceae and 84% increase in Staphylococcaceae as compared to patients with a PaO2/FiO2 ratio > 300. Wilcoxon rank-sum test identified significant enrichment in abundance of OTUs identified as Prevotella melaninogenica (p = 0.042), Corynebacterium (p = 0.037) and Mogibacterium (p = 0.048). Linear discriminant effect size analysis (LefSe) confirmed significant enrichment of Prevotella melaninognica among patients with a PaO2/FiO2 ratio ≤ 300 (p<0.05). These results could not be explained by differences in antibiotic treatment. CONCLUSIONS The airway microbiota following burn and inhalation injury is altered in patients with a PaO2/FiO2 ratio ≤ 300 early after injury. Enrichment of specific taxa in patients with a PaO2/FiO2 ratio ≤ 300 may indicate airway environment and patient changes that favor these microbes. Longitudinal studies are necessary to identify stably colonizing taxa that play roles in hypoxemia and ARDS pathogenesis.
Collapse
Affiliation(s)
- Dana M. Walsh
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Shaun D. McCullough
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Chapel Hill, North Carolina, United States of America
| | - Scott Yourstone
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Samuel W. Jones
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, United States of America
- North Carolina Jaycee Burn Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Bruce A. Cairns
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, United States of America
- North Carolina Jaycee Burn Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Corbin D. Jones
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Program in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Ilona Jaspers
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - David Diaz-Sanchez
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
44
|
Cong X, Hubmayr RD, Li C, Zhao X. Plasma membrane wounding and repair in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2017; 312:L371-L391. [PMID: 28062486 PMCID: PMC5374305 DOI: 10.1152/ajplung.00486.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Various pathophysiological conditions such as surfactant dysfunction, mechanical ventilation, inflammation, pathogen products, environmental exposures, and gastric acid aspiration stress lung cells, and the compromise of plasma membranes occurs as a result. The mechanisms necessary for cells to repair plasma membrane defects have been extensively investigated in the last two decades, and some of these key repair mechanisms are also shown to occur following lung cell injury. Because it was theorized that lung wounding and repair are involved in the pathogenesis of acute respiratory distress syndrome (ARDS) and idiopathic pulmonary fibrosis (IPF), in this review, we summarized the experimental evidence of lung cell injury in these two devastating syndromes and discuss relevant genetic, physical, and biological injury mechanisms, as well as mechanisms used by lung cells for cell survival and membrane repair. Finally, we discuss relevant signaling pathways that may be activated by chronic or repeated lung cell injury as an extension of our cell injury and repair focus in this review. We hope that a holistic view of injurious stimuli relevant for ARDS and IPF could lead to updated experimental models. In addition, parallel discussion of membrane repair mechanisms in lung cells and injury-activated signaling pathways would encourage research to bridge gaps in current knowledge. Indeed, deep understanding of lung cell wounding and repair, and discovery of relevant repair moieties for lung cells, should inspire the development of new therapies that are likely preventive and broadly effective for targeting injurious pulmonary diseases.
Collapse
Affiliation(s)
- Xiaofei Cong
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Rolf D Hubmayr
- Emerius, Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota; and
| | - Changgong Li
- Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia;
| |
Collapse
|
45
|
Peng S, Hang N, Liu W, Guo W, Jiang C, Yang X, Xu Q, Sun Y. Andrographolide sulfonate ameliorates lipopolysaccharide-induced acute lung injury in mice by down-regulating MAPK and NF-κB pathways. Acta Pharm Sin B 2016; 6:205-11. [PMID: 27175331 PMCID: PMC4856950 DOI: 10.1016/j.apsb.2016.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/06/2016] [Accepted: 01/26/2016] [Indexed: 01/24/2023] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a severe, life-threatening medical condition characterized by widespread inflammation in the lungs, and is a significant source of morbidity and mortality in the patient population. New therapies for the treatment of ALI are desperately needed. In the present study, we examined the effect of andrographolide sulfonate, a water-soluble form of andrographolide (trade name: Xi-Yan-Ping Injection), on lipopolysaccharide (LPS)-induced ALI and inflammation. Andrographolide sulfonate was administered by intraperitoneal injection to mice with LPS-induced ALI. LPS-induced airway inflammatory cell recruitment and lung histological alterations were significantly ameliorated by andrographolide sulfonate. Protein levels of pro-inflammatory cytokines in bronchoalveolar lavage fluid (BALF) and serum were reduced by andrographolide sulfonate administration. mRNA levels of pro-inflammatory cytokines in lung tissue were also suppressed. Moreover, andrographolide sulfonate markedly suppressed the activation of mitogen-activated protein kinase (MAPK) as well as p65 subunit of nuclear factor-κB (NF-κB). In summary, these results suggest that andrographolide sulfonate ameliorated LPS-induced ALI in mice by inhibiting NF-κB and MAPK-mediated inflammatory responses. Our study shows that water-soluble andrographolide sulfonate may represent a new therapeutic approach for treating inflammatory lung disorders.
Collapse
Key Words
- ALI, acute lung injury
- ARDS, acute respiratory distress syndrome
- Andrographolide sulfonate
- BALF, bronchoalveolar lavage fluid
- DSS, dextran sulfate sodium
- H&E, hematoxylin & eosin
- HRP, horseradish peroxidase
- IL-6, interleukin-6
- JNK, c-Jun N-terminal kinase
- LPS, lipopolysaccharide
- Lipopolysaccharide
- Lung injury
- MAPK
- MAPK, mitogen-activated protein kinase
- Mouse
- NF-κB
- NF-κB, nuclear factor-κB
- TNBS, trinitrobenzenesulfonic acid
- TNF, tumor necrosis factor
Collapse
|
46
|
miR-429 regulates alveolar macrophage inflammatory cytokine production and is involved in LPS-induced acute lung injury. Biochem J 2015; 471:281-91. [PMID: 26431850 DOI: 10.1042/bj20131510] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 08/20/2015] [Indexed: 12/13/2022]
Abstract
p38 MAPK (mitogen-activated protein kinase) is a critical regulator in lung inflammation. It can be inactivated by DUSP1 (dual-specificity phosphatase 1) which was identified as a putative target of miR-429. miR-429 mimics directly targeted to the 3'-UTR of the gene encoding DUSP1 may result in the translational attenuation of DUSP1. Moreover, the phosphorylation of p38 MAPK was prolonged after miR-429 mimic treatment. Additionally, miR-429 expression was sensitive to LPS (lipopolysaccharide) stimulation and the miR-429 mimics increased the production of pro-inflammatory cytokines. However, anti-miR-429 reduced the LPS-induced production of pro-inflammatory cytokines. These results provide direct evidence that miR-429 is involved in the LPS-induced inflammatory response. In parallel with miR-429, miR-200b and miR-200c, but not miR-200a or miR-141, shared similar effects. In vivo, LPS induced the expression of miR-429, miR-200b and miR-200c in lung. At the same time, inhibiting these miRNAs by anti-miRNAs attenuated the LPS-induced pulmonary inflammatory response and injury. These findings reveal that miR-429 possesses pro-inflammatory activities and may be a potential therapy target for LPS-induced lung injury.
Collapse
|
47
|
Protective Effect of Isorhamnetin on Lipopolysaccharide-Induced Acute Lung Injury in Mice. Inflammation 2015; 39:129-137. [DOI: 10.1007/s10753-015-0231-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
48
|
Poroyko V, Meng F, Meliton A, Afonyushkin T, Ulanov A, Semenyuk E, Latif O, Tesic V, Birukova AA, Birukov KG. Alterations of lung microbiota in a mouse model of LPS-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2015; 309:L76-83. [PMID: 25957290 DOI: 10.1152/ajplung.00061.2014] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/12/2015] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) and the more severe acute respiratory distress syndrome are common responses to a variety of infectious and noninfectious insults. We used a mouse model of ALI induced by intratracheal administration of sterile bacterial wall lipopolysaccharide (LPS) to investigate the changes in innate lung microbiota and study microbial community reaction to lung inflammation and barrier dysfunction induced by endotoxin insult. One group of C57BL/6J mice received LPS via intratracheal injection (n = 6), and another received sterile water (n = 7). Bronchoalveolar lavage (BAL) was performed at 72 h after treatment. Bacterial DNA was extracted and used for qPCR and 16S rRNA gene-tag (V3-V4) sequencing (Illumina). The bacterial load in BAL from ALI mice was increased fivefold (P = 0.03). The community complexity remained unchanged (Simpson index, P = 0.7); the Shannon diversity index indicated the increase of community evenness in response to ALI (P = 0.07). Principal coordinate analysis and analysis of similarity (ANOSIM) test (P = 0.005) revealed a significant difference between microbiota of control and ALI groups. Bacteria from families Xanthomonadaceae and Brucellaceae increased their abundance in the ALI group as determined by Metastats test (P < 0.02). In concordance with the 16s-tag data, Stenotrohomonas maltophilia (Xanthomonadaceae) and Ochrobactrum anthropi (Brucellaceae) were isolated from lungs of mice from both groups. Metabolic profiling of BAL detected the presence of bacterial substrates suitable for both isolates. Additionally, microbiota from LPS-treated mice intensified IL-6-induced lung inflammation in naive mice. We conclude that the morbid transformation of ALI microbiota was attributed to the set of inborn opportunistic pathogens thriving in the environment of inflamed lung, rather than the external infectious agents.
Collapse
Affiliation(s)
- Valeriy Poroyko
- Department of Pediatrics, The University of Chicago, Chicago, Illinois;
| | - Fanyong Meng
- Section of Pulmonary and Critical Medicine, Lung Injury Center, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Angelo Meliton
- Section of Pulmonary and Critical Medicine, Lung Injury Center, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Taras Afonyushkin
- Section of Pulmonary and Critical Medicine, Lung Injury Center, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Alexander Ulanov
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign
| | - Ekaterina Semenyuk
- Department of Microbiology and Immunology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Omar Latif
- Department of Medicine, The University of Chicago, Chicago, Illinois; and
| | - Vera Tesic
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Anna A Birukova
- Section of Pulmonary and Critical Medicine, Lung Injury Center, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Konstantin G Birukov
- Section of Pulmonary and Critical Medicine, Lung Injury Center, Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
49
|
Carlier L, Kempf M, Aaziz R, Jolivet‐Gougeon A, Laroucau K. A severe case of pneumopathy in a duck breeder due to Chlamydia psittaci diagnosed by 16S rDNA sequencing. JMM Case Rep 2014. [DOI: 10.1099/jmmcr.0.001537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- L. Carlier
- Département d’Anesthésie Réanimation, Plateau Technique Ouest, CHU, F‐49933 Angers, France
| | - M. Kempf
- Laboratoire de Bactériologie, Institut de Biologie en Santé – PBH, CHU, F‐49933 Angers, France
| | - R. Aaziz
- Anses, Laboratoire de santé animale, Unité Zoonoses bactériennes, F‐94706 Maisons‐Alfort
| | - A. Jolivet‐Gougeon
- EA 1254 Microbiologie, Université Rennes 1, Service de Bactériologie et Hygiène, CHU Rennes, F‐35043 Rennes, France
| | - K. Laroucau
- Anses, Laboratoire de santé animale, Unité Zoonoses bactériennes, F‐94706 Maisons‐Alfort
| |
Collapse
|
50
|
Immunomodulatory effect of chinese herbal medicine formula sheng-fei-yu-chuan-tang in lipopolysaccharide-induced acute lung injury mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:976342. [PMID: 23997804 PMCID: PMC3755419 DOI: 10.1155/2013/976342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/21/2013] [Accepted: 06/26/2013] [Indexed: 02/06/2023]
Abstract
Traditional Chinese medicine formula Sheng-Fei-Yu-Chuan-Tang (SFYCT), consisting of 13 medicinal plants, was used to treat patients with lung diseases. This study investigated the immunoregulatory effect of SFYCT on intratracheal lipopolysaccharides- (LPS-) challenged acute lung injury (ALI) mice. SFYCT attenuated pulmonary edema, macrophages, and neutrophils infiltration in the airways. SFYCT decreased inflammatory cytokines, including tumor necrosis factor-α (TNFα), interleukin-1β, and interleukin-6 and inhibited nitric oxide (NO) production but increased anti-inflammatory cytokines, interleukin-4, and interleukin-10, in the bronchoalveolar lavage fluid of LPS-challenged mice. TNFα and monocyte chemotactic protein-1 mRNA expression in the lung of LPS-challenged mice as well as LPS-stimulated lung epithelial cell and macrophage were decreased by SFYCT treatment. SFYCT treatment also decreased the inducible nitric oxide synthase expression and phosphorylation of nuclear factor-κB (NF-κB) in the lung of mice and macrophage with LPS stimulation. SFYCT treatment dose dependently decreased the LPS-induced NO and reactive oxygen species generation in LPS-stimulated macrophage. In conclusion, SFYCT attenuated lung inflammation during LPS-induced ALI through decreasing inflammatory cytokines production while increasing anti-inflammatory cytokines production. The immunoregulatory effect of SFYCT is related to inhibiting NF-κB phosphorylation.
Collapse
|