1
|
Choi D, Na AY, Jeoung SW, Choi YH, Park N, Park HS, Kwon HK, Lee HS, Cho DH, Kim DH, Ryu HY. Histone H3 lysine 9 tri-methylation is associated with pterygium. BMC Ophthalmol 2025; 25:106. [PMID: 40033254 PMCID: PMC11874390 DOI: 10.1186/s12886-025-03939-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Pterygium, abnormal growths of conjunctival tissue onto the cornea, are common ocular surface conditions with a high risk of recurrence after surgery and potential ophthalmic complications. The exact cause of pterygium remains unclear, and the triggers are still unknown. This study aims to investigate the relationship between pterygium and epigenetics to uncover the cause of pterygium and identify biomarkers for its diagnosis. METHODS We performed a ChIP-seq assay to compare genome-wide histone modification levels between normal conjunctiva and stage 3 pterygium samples. RESULTS In this study, we investigate the epigenetic profiles of patients with pterygium, focusing on histone H3 lysine 4 (H3K4) and lysine 9 (H3K9) trimethylation (me3). While H3K4me3 levels showed no significant genome-wide change, they were significantly altered in genes related to development and ocular diseases. Conversely, H3K9me3 levels were markedly elevated genome-wide, particularly at the promoters of 82 genes involved in developmental pathways. Furthermore, we identify six genes, ANK2, AOAH, CBLN2, CDH8, CNTNAP4, and DPP6, with decreased gene expression correlated with substantially increased H3K9me3, suggesting their potential as biomarkers for pterygium. CONCLUSION This study represents the first report linking histone modification to pterygium progression, providing valuable insights into therapeutic strategies and potential drug targets.
Collapse
Affiliation(s)
- Dahee Choi
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Ann-Yae Na
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Seok-Won Jeoung
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Yun-Hee Choi
- Department of Ophthalmology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- School of Health and Environmental Science, Korea University, Seoul, South Korea
| | - Nayoon Park
- Department of Ophthalmology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyun-Sun Park
- Department of Pharmacology, Chonnam National University Medical School, Hwasun- gun, Jeonnam, 58128, Republic of Korea
| | - Hyuk-Kwon Kwon
- Division of Life Science, Division of Bio & Medical Bigdata Department (BK4 Program), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyun-Shik Lee
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Dong-Hyung Cho
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Dong Hyun Kim
- Department of Ophthalmology, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Hong-Yeoul Ryu
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
2
|
Jiang W, Chen Y, Yu CY, Zou B, Lu Y, Yang Q, Tang Z, Mao W, Li J, Han H, Shao L, Zeng J, Chu Y, Tang J, Lu M. Alveolar epithelial cells shape lipopolysaccharide-induced inflammatory responses and reprogramming of alveolar macrophages. Eur J Immunol 2025; 55:e2350378. [PMID: 39498697 DOI: 10.1002/eji.202350378] [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: 02/19/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 11/07/2024]
Abstract
Alveolar macrophages (AMs) are sentinels in the airways, where they sense and respond to invading microbes and other stimuli. Unlike macrophages in other locations, AMs can remain responsive to Gram-negative lipopolysaccharides (LPS) after they have responded to LPS in vivo (they do not develop "endotoxin tolerance"), suggesting that the alveolar microenvironment may influence their responses. Although alveolar epithelial cells (AECs) normally limit AMs' innate responses, preventing inflammation induced by harmless antigens in the lung, how AECs influence the innate responses of AMs to infectious agents has been uncertain. Here we report that (1) after exposure to aspirated (intranasal instillation) LPS, AMs increase their responses to TLR agonists and elevate their phagocytic and bactericidal activities in mice; (2) Aspirated LPS pre-exposure increases host resistance to pulmonary infection caused by Gram-negative bacteria and the protection effect lasts for at least 35 days; (3) LPS stimulation of AECs both increases AMs' innate immune responses and prevents AMs from developing tolerance in vitro; (4) Upon LPS stimulation, AMs secreted TNF-α induces AECs to release GM-CSF, which potentiates AMs' response. These experiments have revealed a previously unappreciated role that AECs may play in boosting the innate responses of AMs and promoting resistance to pulmonary infections.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yeying Chen
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Cheng-Yun Yu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Benkun Zou
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yimeng Lu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Yang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihui Tang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiying Mao
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Li
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Han Han
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiashun Zeng
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
3
|
Zeng Y, Li T, Chen X, Fang X, Fang C, Liang X, Liu J, Yang Y. Oral administration of Lactobacillus plantarum expressing aCD11c modulates cellular immunity alleviating inflammatory injury due to Klebsiella pneumoniae infection. BMC Vet Res 2024; 20:399. [PMID: 39244529 PMCID: PMC11380324 DOI: 10.1186/s12917-024-04248-9] [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: 04/06/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae (KP), responsible for acute lung injury (ALI) and inflammation of the gastrointestinal tract, is a zoonotic pathogen that poses a threat to livestock farming worldwide. Nevertheless, there is currently no validated vaccine to prevent KP infection. The development of mucosal vaccines against KP using Lactobacillus plantarum (L. plantarum) is an effective strategy. RESULTS Firstly, the L. plantarum strains NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c were constructed via homologous recombination to express the aCD11c protein either inducibly or constitutively. Both NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c strains could enhance the adhesion and invasion of L. plantarum on bone marrow-derived dendritic cells (BMDCs), and stimulate the activation of BMDCs compared to the control strain NC8-pSIP409 in vitro. Following oral immunization of mice with NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c, the cellular, humoral, and mucosal immunity were significantly improved, as evidenced by the increased expression of CD4+ IL-4+ T cells in the spleen, IgG in serum, and secretory IgA (sIgA) in the intestinal lavage fluid (ILF). Furthermore, the protective effects of L. plantarum against inflammatory damage caused by KP infection were confirmed by assessing the bacterial loads in various tissues, lung wet/dry ratio (W/D), levels of inflammatory cytokines, and histological evaluation, which influenced T helper 17 (Th17) and regulatory T (Treg) cells in peripheral blood and lung. CONCLUSIONS Both the inducible and constitutive L. plantarum strains NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c have been found to stimulate cellular and humoral immunity levels and alleviate the inflammatory response caused by KP infection. These findings have provided a basis for the development of a novel vaccine against KP.
Collapse
Affiliation(s)
- Yang Zeng
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Tiantian Li
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xueyang Chen
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xiaowei Fang
- College of Agriculture, Yangtze University, Jingzhou, 434025, China
| | - Chun Fang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xiongyan Liang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Jing Liu
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China.
| | - Yuying Yang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
4
|
Wang L, Shen J, Liu W, Li W, Tang W, Zha B, Wu H, Liu X, Shen Q. Abscisic acid for acute respiratory distress syndrome therapy by suppressing alveolar macrophage pyroptosis via upregulating acyloxyacyl hydrolase expression. Eur J Pharmacol 2024; 977:176672. [PMID: 38849041 DOI: 10.1016/j.ejphar.2024.176672] [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: 12/18/2023] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024]
Abstract
OBJECTIVE Abscisic acid (ABA) is a phytohormone that inhibits airway inflammation in acute respiratory distress syndrome (ARDS) mouse models. However, the molecular mechanism underlying this phenomenon remains unclear. METHODS Serum ABA level in patients and mice was measured via liquid chromatography-tandem mass spectrometry (LC-MS/MS). In-depth molecular mechanism was investigated through transmission electron microscopy, RNA-sequencing, and molecular docking in ARDS mice and cultured primary alveolar macrophages (AMs). RESULTS We found that the serum ABA level was remarkably decreased in ARDS mice and patients. ABA inhibited lipopolysaccharide (LPS)-induced airway inflammation in mice; moreover, it downregulated genes associated with pyroptosis, as shown by RNA-sequencing and lung protein immunoblots. ABA inhibited the formation of membrane pores in AMs and suppressed the cleavage of gasdermin D (GSDMD) and the activation of caspase-11 and caspase-1 in vivo and in vitro; however, the overexpression of caspase-11 reversed the protective effect of ABA on LPS-induced pyroptosis of primary AMs. ABA inhibited intra-AM LPS accumulation while increasing the level of acyloxyacyl hydrolase (AOAH) in AMs, whereas AOAH deficiency abrogated the suppressive action of ABA on inflammation, pyroptosis, and intra-AM LPS accumulation in vivo and in vitro. Importantly, ABA promoted its intracellular receptor lanthionine C-like receptor 2 interacting with transcription factor peroxisome proliferator-activated receptor γ, which ultimately leading to increase AOAH expression to inactivate LPS and inhibit pyroptosis in AMs. CONCLUSIONS ABA protected against LPS-induced lung injury by inhibiting pyroptosis in AMs via proliferator-activated receptor γ-mediated AOAH expression.
Collapse
Affiliation(s)
- Lixia Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, China
| | - Jian Shen
- Department of Anesthesiology and Perioperative Medicine, Jiangsu Province Hospital, China
| | - Weiju Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, China
| | - Wei Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, China
| | - Weijie Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, China
| | - Binshan Zha
- Department of Vascular and Thyroid Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, China
| | - Huimei Wu
- Department of Geriatric Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, China.
| | - Qiying Shen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, China.
| |
Collapse
|
5
|
Zhu Y, Hu Y, Liu Z, Chang L, Geng X, Yin X, Zhao BQ, Fan W. The LPS-inactivating enzyme acyloxyacyl hydrolase protects the brain from experimental stroke. Transl Res 2024; 270:42-51. [PMID: 38522823 DOI: 10.1016/j.trsl.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/29/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Blood-brain-barrier (BBB) disruption is a pathological hallmark of ischemic stroke, and inflammation occurring at the BBB contributes to the pathogenesis of ischemic brain injury. Lipopolysaccharide (LPS), a cell wall component of Gram-negative bacteria, is elevated in patients with acute stroke. The activity of LPS is controlled by acyloxyacyl hydrolase (AOAH), a host enzyme that deacylates LPS to inactivated forms. However, whether AOAH influences the pathogenesis of ischemic stroke remain elusive. We performed in vivo experiments to explore the role and mechanism of AOAH on neutrophil extravasation, BBB disruption, and brain infarction. We found that AOAH was upregulated in neutrophils in peri-infarct areas from mice with transient focal cerebral ischemia. AOAH deficiency increased neutrophil extravasation into the brain parenchyma and proinflammatory cytokine production, broke down the BBB and worsened stroke outcomes in mice. These effects require Toll-like receptor 4 (TLR4) because absence of TLR4 or pharmacologic inhibition of TLR4 signaling prevented the exacerbated inflammation and BBB damage in Aoah-/- mice after ischemic stroke. Importantly, neutrophil depletion or inhibition of neutrophil trafficking by blocking LFA-1 integrin dramatically reduced stroke-induced BBB breakdown in Aoah-/- mice. Furthermore, virus-mediated overexpression of AOAH induced a substantial decrease in neutrophil recruitment that was accompanied by reducing BBB damage and stroke volumes. Our findings show the importance of AOAH in regulating neutrophil-dependent BBB breakdown and cerebral infarction. Consequently, strategies that modulate AOAH may be a new therapeutic approach for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yuanbo Zhu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yue Hu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhongwang Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Luping Chang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xue Geng
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xuhui Yin
- Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Bing-Qiao Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Wenying Fan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
6
|
Wu Z, Deng B, Shen Y, Li X, Li J, Li Y, Ma S, Pan Y, Ding F. Acyloxyacyl Hydrolase Protects against Kidney Injury via Inhibition of Tubular CD74-Macrophage Crosstalk. Int J Biol Sci 2024; 20:3061-3075. [PMID: 38904010 PMCID: PMC11186370 DOI: 10.7150/ijbs.91237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Renal fibrosis is the common pathway in the progression of chronic kidney disease (CKD). Acyloxyacyl hydrolase (AOAH) is expressed in various phagocytes and is highly expressed in proximal tubular epithelial cells (PTECs). Research shows that AOAH plays a critical role in infections and chronic inflammatory diseases, although its role in kidney injury is unknown. Here, we found that AOAH deletion led to exacerbated kidney injury and fibrosis after folic acid (FA) administration, which was reversed by overexpression of Aoah in kidneys. ScRNA-seq revealed that Aoah-/- mice exhibited increased subpopulation of CD74+ PTECs, though the percentage of total PTECs were decreased compared to WT mice after FA treatment. Additionally, exacerbated kidney injury and fibrosis seen in Aoah-/- mice was attenuated via administration of methyl ester of (S, R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1), an inhibitor of macrophage inhibition factor (MIF) and CD74 binding. Finally, AOAH expression was found positively correlated with estimated glomerular filtration rate while negatively correlated with the degree of renal fibrosis in kidneys of CKD patients. Thus, our work indicates that AOAH protects against kidney injury and fibrosis by inhibiting renal tubular epithelial cells CD74 signaling pathways. Targeting kidney AOAH represents a promising strategy to prevent renal fibrosis progression.
Collapse
Affiliation(s)
- Zhenkai Wu
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Deng
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqi Shen
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuezhu Li
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaolun Li
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Ma
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Pan
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research
| |
Collapse
|
7
|
Wang X, Wan W, Zhang J, Lu J, Liu P. Efficient pulmonary fibrosis therapy via regulating macrophage polarization using respirable cryptotanshinone-loaded liposomal microparticles. J Control Release 2024; 366:1-17. [PMID: 38154539 DOI: 10.1016/j.jconrel.2023.12.042] [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: 10/22/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Lung inflammation and fibrogenesis are the two main characteristics during the development of pulmonary fibrosis (PF), which are particularly associated with pulmonary macrophages. In this context, whether cryptotanshinone (CTS) could alleviate PF through regulating macrophage polarization were preliminarily demonstrated in vitro. Then the time course of PF and its relationship with macrophage polarization was determined in BLM-induced mice based on cytokine levels in bronchoalveolar lavage fluid (BALF), lung histopathology, flow cytometric analysis, mRNA and protein expression. CTS was loaded into macrophage-targeted and responsively released mannose-modified liposomes (Man-lipo), and the liposomes were then embedded into mannitol microparticles (M-MPs) using spray drying to achieve efficient pulmonary delivery. Afterwards, how CTS regulates macrophage polarization in vivo during different time courses of PF was probed. Furthermore, the molecular mechanisms of CTS against PF by regulating macrophage polarization were elucidated in vivo and in vitro. The full-course therapy group could achieve comparable therapeutic effects compared with the positive control drug PFD group. CTS can alleviate PF through regulating macrophage polarization, mainly by inhibiting NLRP3/TGF-β1 pathway during the inflammation course and modulating MMP-9/TIMP-1 balance during the fibrosis development course, providing new insights into chronic PF treatment.
Collapse
Affiliation(s)
- Xiuhua Wang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiguo Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Jing Lu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.
| |
Collapse
|
8
|
Wu J, Cai J, Tang Y, Lu B. The noncanonical inflammasome-induced pyroptosis and septic shock. Semin Immunol 2023; 70:101844. [PMID: 37778179 DOI: 10.1016/j.smim.2023.101844] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/10/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023]
Abstract
Sepsis remains one of the most common and lethal conditions globally. Currently, no proposed target specific to sepsis improves survival in clinical trials. Thus, an in-depth understanding of the pathogenesis of sepsis is needed to propel the discovery of effective treatment. Recently attention to sepsis has intensified because of a growing recognition of a non-canonical inflammasome-triggered lytic mode of cell death termed pyroptosis upon sensing cytosolic lipopolysaccharide (LPS). Although the consequences of activation of the canonical and non-canonical inflammasome are similar, the non-canonical inflammasome formation requires caspase-4/5/11, which enzymatically cleave the pore-forming protein gasdermin D (GSDMD) and thereby cause pyroptosis. The non-canonical inflammasome assembly triggers such inflammatory cell death by itself; or leverages a secondary activation of the canonical NLRP3 inflammasome pathway. Excessive cell death induced by oligomerization of GSDMD and NINJ1 leads to cytokine release and massive tissue damage, facilitating devastating consequences and death. This review summarized the updated mechanisms that initiate and regulate non-canonical inflammasome activation and pyroptosis and highlighted various endogenous or synthetic molecules as potential therapeutic targets for treating sepsis.
Collapse
Affiliation(s)
- Junru Wu
- Department of Cardiology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China
| | - Jingjing Cai
- Department of Cardiology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410000, PR China
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410000, PR China.
| |
Collapse
|
9
|
Singh AK, Wang R, Lombardo KA, Praharaj M, Bullen CK, Um P, Gupta M, Srikrishna G, Davis S, Komm O, Illei PB, Ordonez AA, Bahr M, Huang J, Gupta A, Psoter KJ, Creisher PS, Li M, Pekosz A, Klein SL, Jain SK, Bivalacqua TJ, Yegnasubramanian S, Bishai WR. Intravenous BCG vaccination reduces SARS-CoV-2 severity and promotes extensive reprogramming of lung immune cells. iScience 2023; 26:107733. [PMID: 37674985 PMCID: PMC10477068 DOI: 10.1016/j.isci.2023.107733] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/31/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Bacillus Calmette-Guérin (BCG) confers heterologous immune protection against viral infections and has been proposed as vaccine against SARS-CoV-2 (SCV2). Here, we tested intravenous BCG vaccination against COVID-19 using the golden Syrian hamster model. BCG vaccination conferred a modest reduction on lung SCV2 viral load, bronchopneumonia scores, and weight loss, accompanied by a reversal of SCV2-mediated T cell lymphopenia, and reduced lung granulocytes. BCG uniquely recruited immunoglobulin-producing plasma cells to the lung suggesting accelerated local antibody production. BCG vaccination also recruited elevated levels of Th1, Th17, Treg, CTLs, and Tmem cells, with a transcriptional shift away from exhaustion markers and toward antigen presentation and repair. Similarly, BCG enhanced recruitment of alveolar macrophages and reduced key interstitial macrophage subsets, that show reduced IFN-associated gene expression. Our observations indicate that BCG vaccination protects against SCV2 immunopathology by promoting early lung immunoglobulin production and immunotolerizing transcriptional patterns among key myeloid and lymphoid populations.
Collapse
Affiliation(s)
- Alok K. Singh
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Rulin Wang
- Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Kara A. Lombardo
- Johns Hopkins University, School of Medicine, Department of Urology, Baltimore, MD, USA
| | - Monali Praharaj
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - C. Korin Bullen
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Peter Um
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Manish Gupta
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Geetha Srikrishna
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Stephanie Davis
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Oliver Komm
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Peter B. Illei
- Johns Hopkins University, School of Medicine, Department of Pathology, Baltimore, MD, USA
| | - Alvaro A. Ordonez
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore, MD, USA
| | - Melissa Bahr
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore, MD, USA
| | - Joy Huang
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Anuj Gupta
- Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Kevin J. Psoter
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of General Pediatrics, Baltimore, MD, USA
| | - Patrick S. Creisher
- Johns Hopkins University, Bloomberg School of Public Health, The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Maggie Li
- Johns Hopkins University, Bloomberg School of Public Health, The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Andrew Pekosz
- Johns Hopkins University, Bloomberg School of Public Health, The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Sabra L. Klein
- Johns Hopkins University, Bloomberg School of Public Health, The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Sanjay K. Jain
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore, MD, USA
| | - Trinity J. Bivalacqua
- Perelman School of Medicine at the University of Pennsylvania, Division of Urology, Department of Surgery, Philadelphia, PA, USA
| | | | - William R. Bishai
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| |
Collapse
|
10
|
Caputo MJ, Li W, Kendall SJ, Larsen A, Weigel KA, White HM. Liver and Muscle Transcriptomes Differ in Mid-Lactation Cows Divergent in Feed Efficiency in the Presence or Absence of Supplemental Rumen-Protected Choline. Metabolites 2023; 13:1023. [PMID: 37755303 PMCID: PMC10536747 DOI: 10.3390/metabo13091023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Improving dairy cow feed efficiency is critical to the sustainability and profitability of dairy production, yet the underlying mechanisms that contribute to individual cow variation in feed efficiency are not fully understood. The objectives of this study were to (1) identify genes and associated pathways that are altered in cows with high- or low-residual feed intake (RFI) using RNA sequencing, and (2) determine if rumen-protected choline supplementation during mid-lactation would influence performance or feed efficiency. Mid-lactation (134 ± 20 days in milk) multiparous Holstein cows were randomly assigned to either supplementation of 0 g/d supplementation (CTL; n = 32) or 30 g/d of a rumen-protected choline product (RPC; 13.2 g choline ion; n = 32; Balchem Corp., New Hampton, NY, USA). Residual feed intake was determined as dry matter intake regressed on milk energy output, days in milk, body weight change, metabolic body weight, and dietary treatment. The 12 cows with the highest RFI (low feed efficient; LE) and 12 cows with the lowest RFI (high feed efficient; HE), balanced by dietary treatment, were selected for blood, liver, and muscle analysis. No differences in production or feed efficiency were detected with RPC supplementation, although albumin was greater and arachidonic acid tended to be greater in RPC cows. Concentrations of β-hydroxybutyrate were greater in HE cows. Between HE and LE, 268 and 315 differentially expressed genes in liver and muscle tissue, respectively, were identified through RNA sequencing. Pathway analysis indicated differences in cell cycling, oxidative stress, and immunity in liver and differences in glucose and fatty acid pathways in muscle. The current work indicates that unique differences in liver and muscle post-absorptive nutrient metabolism contribute to sources of variation in feed efficiency and that differences in amino acid and fatty acid oxidation, cell cycling, and immune function should be further examined.
Collapse
Affiliation(s)
- Malia J. Caputo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Wenli Li
- United States Department of Agriculture-Agriculture Research Station, Madison, WI 53706, USA;
| | - Sophia J. Kendall
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Anna Larsen
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
- United States Department of Agriculture-Agriculture Research Station, Madison, WI 53706, USA;
| | - Kent A. Weigel
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Heather M. White
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| |
Collapse
|
11
|
Li X, Qu S, Song X, Wu C, Shen J, Zhu K. In Situ Neutralization and Detoxification of LPS to Attenuate Hyperinflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302950. [PMID: 37428467 PMCID: PMC10502683 DOI: 10.1002/advs.202302950] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 07/11/2023]
Abstract
Hyperinflammation elicited by lipopolysaccharide (LPS) that derives from multidrug-resistant Gram-negative pathogens, leads to a sharp increase in mortality globally. However, monotherapies aiming to neutralize LPS often fail to improve the prognosis. Here, an all-in-one drug delivery strategy equipped with bactericidal activity, LPS neutralization, and detoxification is shown to recognize, kill pathogens, and attenuate hyperinflammation by abolishing the activation of LPS-triggered acute inflammatory responses. First, bactericidal colistin results in rapid bacterial killing, and the released LPS is subsequently sequestered. The neutralized LPS is further cleared by acyloxyacyl hydrolase to remove secondary fatty chains and detoxify LPS in situ. Last, such a system shows high efficacy in two mouse infection models challenged with Pseudomonas aeruginosa. This approach integrates direct antibacterial activity with in situ LPS neutralizing and detoxifying properties, shedding light on the development of alternative interventions to treat sepsis-associated infections.
Collapse
Affiliation(s)
- Xiaoyu Li
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Shaoqi Qu
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Xiangbin Song
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhou510642China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health SecurityCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Guangdong Laboratory for Lingnan Modern AgricultureGuangzhou510642China
| |
Collapse
|
12
|
Cheng X, Jiang W, Chen Y, Zou B, Wang Z, Gan L, Xiao Z, Li C, Yu CY, Lu Y, Han Z, Zeng J, Gu J, Chu T, Fu M, Chu Y, Zhang W, Tang J, Lu M. Acyloxyacyl hydrolase promotes pulmonary defense by preventing alveolar macrophage tolerance. PLoS Pathog 2023; 19:e1011556. [PMID: 37498977 PMCID: PMC10409266 DOI: 10.1371/journal.ppat.1011556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/08/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
Although alveolar macrophages (AMs) play important roles in preventing and eliminating pulmonary infections, little is known about their regulation in healthy animals. Since exposure to LPS often renders cells hyporesponsive to subsequent LPS exposures ("tolerant"), we tested the hypothesis that LPS produced in the intestine reaches the lungs and stimulates AMs, rendering them tolerant. We found that resting AMs were more likely to be tolerant in mice lacking acyloxyacyl hydrolase (AOAH), the host lipase that degrades and inactivates LPS; isolated Aoah-/- AMs were less responsive to LPS stimulation and less phagocytic than were Aoah+/+ AMs. Upon innate stimulation in the airways, Aoah-/- mice had reduced epithelium- and macrophage-derived chemokine/cytokine production. Aoah-/- mice also developed greater and more prolonged loss of body weight and higher bacterial burdens after pulmonary challenge with Pseudomonas aeruginosa than did wildtype mice. We also found that bloodborne or intrarectally-administered LPS desensitized ("tolerized") AMs while antimicrobial drug treatment that reduced intestinal commensal Gram-negative bacterial abundance largely restored the innate responsiveness of Aoah-/- AMs. Confirming the role of LPS stimulation, the absence of TLR4 prevented Aoah-/- AM tolerance. We conclude that commensal LPSs may stimulate and desensitize (tolerize) alveolar macrophages in a TLR4-dependent manner and compromise pulmonary immunity. By inactivating LPS in the intestine, AOAH promotes antibacterial host defenses in the lung.
Collapse
Affiliation(s)
- Xiaofang Cheng
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yeying Chen
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Benkun Zou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyan Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Lu Gan
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zeling Xiao
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Changshun Li
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Cheng-Yun Yu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yimeng Lu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zeyao Han
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jiashun Zeng
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianqing Chu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mingsheng Fu
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China
| | - Wenhong Zhang
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Mingfang Lu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China
| |
Collapse
|
13
|
Chancharoenthana W, Kamolratanakul S, Schultz MJ, Leelahavanichkul A. The leaky gut and the gut microbiome in sepsis - targets in research and treatment. Clin Sci (Lond) 2023; 137:645-662. [PMID: 37083032 PMCID: PMC10133873 DOI: 10.1042/cs20220777] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/25/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023]
Abstract
Both a leaky gut (a barrier defect of the intestinal surface) and gut dysbiosis (a change in the intestinal microbial population) are intrinsic to sepsis. While sepsis itself can cause dysbiosis, dysbiosis can worsen sepsis. The leaky gut syndrome refers to a status with which there is an increased intestinal permeability allowing the translocation of microbial molecules from the gut into the blood circulation. It is not just a symptom of gastrointestinal involvement, but also an underlying cause that develops independently, and its presence could be recognized by the detection, in blood, of lipopolysaccharides and (1→3)-β-D-glucan (major components of gut microbiota). Gut-dysbiosis is the consequence of a reduction in some bacterial species in the gut microbiome, as a consequence of intestinal mucosal immunity defect, caused by intestinal hypoperfusion, immune cell apoptosis, and a variety of enteric neuro-humoral-immunity responses. A reduction in bacteria that produce short-chain fatty acids could change the intestinal barriers, leading to the translocation of pathogen molecules, into the circulation where it causes systemic inflammation. Even gut fungi might be increased in human patients with sepsis, even though this has not been consistently observed in murine models of sepsis, probably because of the longer duration of sepsis and also antibiotic use in patients. The gut virobiome that partly consists of bacteriophages is also detectable in gut contents that might be different between sepsis and normal hosts. These alterations of gut dysbiosis altogether could be an interesting target for sepsis adjuvant therapies, e.g., by faecal transplantation or probiotic therapy. Here, current information on leaky gut and gut dysbiosis along with the potential biomarkers, new treatment strategies, and future research topics are mentioned.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Marcus J Schultz
- Department of Intensive Care and Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
14
|
Roberts JL, Golloshi M, Harding DB, Conduah M, Liu G, Drissi H. Bifidobacterium longum supplementation improves age-related delays in fracture repair. Aging Cell 2023; 22:e13786. [PMID: 36704918 PMCID: PMC10086533 DOI: 10.1111/acel.13786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Abstract
Age-related delays in bone repair remains an important clinical issue that can prolong pain and suffering. It is now well established that inflammation increases with aging and that this exacerbated inflammatory response can influence skeletal regeneration. Recently, simple dietary supplementation with beneficial probiotic bacteria has been shown to influence fracture repair in young mice. However, the contribution of the gut microbiota to age-related impairments in fracture healing remains unknown. Here, we sought to determine whether supplementation with a single beneficial probiotic species, Bifidobacterium longum (B. longum), would promote fracture repair in aged (18-month-old) female mice. We found that B. longum supplementation accelerated bony callus formation which improved mechanical properties of the fractured limb. We attribute these pro-regenerative effects of B. longum to preservation of intestinal barrier, dampened systemic inflammation, and maintenance of the microbiota community structure. Moreover, B. longum attenuated many of the fracture-induced systemic pathologies. Our study provides evidence that targeting the gut microbiota using simple dietary approaches can improve fracture healing outcomes and minimize systemic pathologies in the context of aging.
Collapse
Affiliation(s)
- Joseph L Roberts
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Mateo Golloshi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Derek B Harding
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Madison Conduah
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Guanglu Liu
- Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| |
Collapse
|
15
|
Cheng X, Song X, Li Z, Yuan C, Lei X, Feng M, Hong Z, Zhang L, Hong D. Acyloxyacyl hydrolase deficiency induces chronic inflammation and bone loss in male mice. J Mol Med (Berl) 2022; 100:1599-1616. [PMID: 36112153 DOI: 10.1007/s00109-022-02252-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/10/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
Hormonal homeostasis is essential in bone remodeling. Recent studies have shown that the treatment of intestinal inflammation can result in the regulation of bone resorption in distant bones. Increased intestinal permeability may lead to systemic inflammation and bone loss, also known as gut-bone axis. However, the underlying mechanism remains to be elucidated. Lipopolysaccharide (LPS) is a component of gram-negative bacteria that can increase osteoclastic differentiation in vitro. Acyloxyacyl hydrolase (AOAH) is a specific degrading enzyme of LPS, but little is known about the role of AOAH in bone metabolism. In this study, adult Aoah-/- mice showed a chronic inflammatory state and osteopenic phenotype analyzed by micro-CT and HE staining. Tartrate-resistant acid phosphatase (TRAP) staining of femurs showed an increase in TRAP-positive cells from Aoah-/- mice. AOAH depletion enhanced the osteoclast differentiation and bone resorption capacity of bone marrow-derived macrophages (BMMs). The enhanced osteoclast differentiation and bone resorption capacity of Aoah-/- BMMs were reversed by rAOAH. In conclusion, the chronic inflammatory state of adult Aoah-/- mice promotes bone resorption. AOAH participates in bone metabolism, which is mainly mediated by inhibiting osteoclast differentiation. LPS may be a key mediator of the gut-bone axis, and targeting AOAH may represent a feasible strategy for the treatment of chronic inflammatory bone resorption. KEY MESSAGES : AOAH knockout mice exhibited chronic inflammation mediated by LPS, and LPS may also serve as an important mediator in the regulation of bone metabolism in the gut-bone axis. AOAH regulated bone resorption by blocking the osteoclast differentiation via classical ERK and JNK pathways. rAOAH could rescue the enhanced osteoclast differentiation caused by AOAH deficiency.
Collapse
Affiliation(s)
- Xu Cheng
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiaoting Song
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhiyan Li
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Chiting Yuan
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Xinhuan Lei
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Mingxuan Feng
- Department of Orthopedics, Taizhou Central Hospital Affiliated to Taizhou College, Taizhou, Zhejiang, China
| | - Zhenghua Hong
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.,Enze Medical Center, Taizhou, China.,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Liwei Zhang
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China. .,Enze Medical Center, Taizhou, China. .,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.
| | - Dun Hong
- Department of Orthopedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China. .,Enze Medical Center, Taizhou, China. .,Bone Metabolism and Development Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.
| |
Collapse
|
16
|
Szafran B, Borazjani A, Scheaffer HL, Crow JA, McBride AM, Adekanye O, Wonnacott CB, Lehner R, Kaplan BLF, Ross MK. Carboxylesterase 1d Inactivation Augments Lung Inflammation in Mice. ACS Pharmacol Transl Sci 2022; 5:919-931. [PMID: 36268116 PMCID: PMC9578131 DOI: 10.1021/acsptsci.2c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Indexed: 11/28/2022]
Abstract
Carboxylesterases are members of the serine hydrolase superfamily and metabolize drugs, pesticides, and lipids. Previous research showed that inhibition of carboxylesterase 1 (CES1) in human macrophages altered the immunomodulatory effects of lipid mediators called prostaglandin glyceryl esters, which are produced by cyclooxygenase-catalyzed oxygenation of the endocannabinoid 2-arachidonoylglycerol (2-AG). Ces1d - the mouse ortholog of human CES1 - is the most abundant Ces isoform in murine lung tissues and alveolar macrophages and a major target of organophosphate poisons. Monoacylglycerol lipase (Magl) is also expressed in murine lung and is the main enzyme responsible for 2-AG catabolism. Several metabolic benefits are observed in Ces1d-/- mice fed a high-fat diet; thus, we wondered whether pharmacological and genetic inactivation of Ces1d in vivo might also ameliorate the acute inflammatory response to lipopolysaccharide (LPS). C57BL/6 mice were treated with WWL229 (Ces1d inhibitor) or JZL184 (Magl inhibitor), followed 30 min later by either LPS or saline. Wild-type (WT) and Ces1d-/- mice were also administered LPS to determine the effect of Ces1d knockout. Mice were sacrificed at 6 and 24 h, and cytokines were assessed in serum, lung, liver, and adipose tissues. Lipid mediators were quantified in lung tissues, while activity-based protein profiling and enzyme assays determined the extent of lung serine hydrolase inactivation by the inhibitors. WWL229 was shown to augment LPS-induced lung inflammation in a female-specific manner, as measured by enhanced neutrophil infiltration and Il1b mRNA. The marked Ces inhibition in female lung by 4 h after drug treatment might explain this sex difference, although the degree of Ces inhibition in female and male lungs was similar at 6 h. In addition, induction of lung Il6 mRNA and prostaglandin E2 by LPS was more pronounced in Ces1d-/- mice than in WT mice. Thus, WWL229 inhibited lung Ces1d activity and augmented the female lung innate immune response, an effect observed in part in Ces1d-/- mice and Ces1d/CES1-deficient murine and human macrophages. In contrast, JZL184 attenuated LPS-induced Il1b and Il6 mRNA levels in female lung, suggesting that Ces1d and Magl have opposing effects. Mapping the immunomodulatory molecules/pathways that are regulated by Ces1d in the context of lung inflammation will require further research.
Collapse
Affiliation(s)
- Brittany
N. Szafran
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Abdolsamad Borazjani
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Hannah L. Scheaffer
- Department
of Biochemistry, Molecular Biology, Entomology, and Plant Pathology,
College of Agriculture and Life Sciences, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - J. Allen Crow
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Ann Marie McBride
- Department
of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Oluwabori Adekanye
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Caitlin B. Wonnacott
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Richard Lehner
- Departments
of Cell Biology and Pediatrics, Group on Molecular & Cell Biology
of Lipids, University of Alberta, Edmonton, ABT6G 2R3, Canada
| | - Barbara L. F. Kaplan
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| | - Matthew K. Ross
- Department
of Comparative Biomedical Sciences, Center for Environmental Health
Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi39762, United States
| |
Collapse
|
17
|
Fang H, Sun Z, Chen Z, Chen A, Sun D, Kong Y, Fang H, Qian G. Bioinformatics and systems-biology analysis to determine the effects of Coronavirus disease 2019 on patients with allergic asthma. Front Immunol 2022; 13:988479. [PMID: 36211429 PMCID: PMC9537444 DOI: 10.3389/fimmu.2022.988479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background The coronavirus disease (COVID-19) pandemic has posed a significant challenge for global health systems. Increasing evidence shows that asthma phenotypes and comorbidities are major risk factors for COVID-19 symptom severity. However, the molecular mechanisms underlying the association between COVID-19 and asthma are poorly understood. Therefore, we conducted bioinformatics and systems biology analysis to identify common pathways and molecular biomarkers in patients with COVID-19 and asthma, as well as potential molecular mechanisms and candidate drugs for treating patients with both COVID-19 and asthma. Methods Two sets of differentially expressed genes (DEGs) from the GSE171110 and GSE143192 datasets were intersected to identify common hub genes, shared pathways, and candidate drugs. In addition, murine models were utilized to explore the expression levels and associations of the hub genes in asthma and lung inflammation/injury. Results We discovered 157 common DEGs between the asthma and COVID-19 datasets. A protein–protein-interaction network was built using various combinatorial statistical approaches and bioinformatics tools, which revealed several hub genes and critical modules. Six of the hub genes were markedly elevated in murine asthmatic lungs and were positively associated with IL-5, IL-13 and MUC5AC, which are the key mediators of allergic asthma. Gene Ontology and pathway analysis revealed common associations between asthma and COVID-19 progression. Finally, we identified transcription factor–gene interactions, DEG–microRNA coregulatory networks, and potential drug and chemical-compound interactions using the hub genes. Conclusion We identified the top 15 hub genes that can be used as novel biomarkers of COVID-19 and asthma and discovered several promising candidate drugs that might be helpful for treating patients with COVID-19 and asthma.
Collapse
Affiliation(s)
- Hongwei Fang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhun Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhouyi Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Donglin Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yan Kong
- Department of Anesthesiology (High-Tech Branch), The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Guojun Qian, ; Hao Fang,
| | - Guojun Qian
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Guojun Qian, ; Hao Fang,
| |
Collapse
|
18
|
Zheng X, Guo J, Cao C, Qin T, Zhao Y, Song X, Lv M, Hu L, Zhang L, Zhou D, Fang T, Yang W. Time-Course Transcriptome Analysis of Lungs From Mice Infected With Hypervirulent Klebsiella pneumoniae via Aerosolized Intratracheal Inoculation. Front Cell Infect Microbiol 2022; 12:833080. [PMID: 35573776 PMCID: PMC9097095 DOI: 10.3389/fcimb.2022.833080] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/11/2022] [Indexed: 11/21/2022] Open
Abstract
Hypervirulent Klebsiella pneumoniae (hvKp) can cause life-threatening community-acquired infections among healthy young individuals and is thus of concern for global dissemination. In this study, a mouse model of acute primary hvKp pneumonia was established via aerosolized intratracheal (i.t.) inoculation, laying the foundation for conducting extensive studies related to hvKp. Subsequently, a time-course transcriptional profile was created of the lungs from the mouse model at 0, 12, 24, 48 and 60 hours post-infection (hpi) using RNA Sequencing (RNA-Seq). RNA-Seq data were analyzed with the use of Mfuzz time clustering, weighted gene co-expression network analysis (WGCNA) and Immune Cell Abundance Identifier for mouse (ImmuCellAI-mouse). A gradual change in the transcriptional profile of the lungs was observed that reflected expected disease progression. At 12 hpi, genes related to acute phase inflammatory response increased in expression and lipid metabolism appeared to have a pro-inflammatory effect. At 24 hpi, exacerbation of inflammation was observed and active IFN-γ suggested that signaling promoted activation and recruitment of macrophages occurred. Genes related to maintaining the structural integrity of lung tissues showed a sustained decrease in expression after infection and the decrease was especially marked at 48 hpi. TNF, IL-17, MAPK and NF-kB signaling pathways may play key roles in the immunopathogenesis mechanism at all stages of infection. Natural killer (NK) cells consistently decreased in abundance after infection, which has rarely been reported in hvKp infection and could provide a new target for treatment. Genes Saa1 and Slpi were significantly upregulated during infection. Both Saa1, which is associated with lipopolysaccharide (LPS) that elicits host inflammatory response, and Slpi, which encodes an antimicrobial protein, have not previously been reported in hvKp infections and could be important targets for subsequent studies. To t our knowledge, this paper represents the first study to investigate the pulmonary transcriptional response to hvKp infection. The results provide new insights into the molecular mechanisms underlying the pathogenesis of hvKp pulmonary infection that can contribute to the development of therapies to reduce hvKp pneumonia.
Collapse
Affiliation(s)
- Xinying Zheng
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jianshu Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Chaoyue Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tongtong Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing, China
| | - Yue Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolin Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Meng Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lili Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tongyu Fang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- *Correspondence: Tongyu Fang, ; Wenhui Yang,
| | - Wenhui Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- *Correspondence: Tongyu Fang, ; Wenhui Yang,
| |
Collapse
|
19
|
Li J, Pan C, Tang C, Tan W, Liu H, Guan J. The Reaction Pathway of miR-30c-5p Activates Lipopolysaccharide Promoting the Course of Traumatic and Hemorrhagic Shock Acute Lung Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3330552. [PMID: 35463979 PMCID: PMC9021990 DOI: 10.1155/2022/3330552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022]
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory failure caused by diffuse inflammatory injury in alveolar epithelial cells during severe infection, trauma, and shock. Among them, trauma/hemorrhagic shock (T/HS) is the main type of indirect lung injury. Despite a great number of clinical studies, indirect factor trauma/hemorrhagic shock to the function and the mechanism in acute lung injury is not clear yet. Therefore, it is still necessary to carry on relevant analysis in order to thoroughly explore its molecular and cellular mechanisms and the pathway of disease function. In our research, we aimed to identify potential pathogenic genes and do modular analysis by downloading disease-related gene expression profile data. And our dataset is from the NCBI-GEO database. Then, we used the Clusterprofiler R package, GO function, and KEGG pathway enrichment analysis to analyze the core module genes. In addition, we also identified key transcription factors and noncoding RNAs. Based on the high degree of interaction of potential pathogenic genes and their involved functions and pathways, we identified 17 dysfunction modules. Among them, up to 9 modules significantly regulate the response to bacterial-derived molecules, and the response to lipopolysaccharide and other related functional pathways that mediate disease development. In addition, miR-290, miR-30c-5p, miR-195-5p, and miR-1-3p-based ncRNA and Jun, Atf1, and Atf3-based transcription factors have a total of 80 transcription drivers for functional modules. In summary, this study confirmed that miR-30c-5p activates lipopolysaccharide response pathway to promote the pathogenesis of ALI induced by hemorrhagic shock. This result can be an important direction for further research on related deepening diseases such as acute respiratory distress syndrome (ARDS). It further provides a piece of scientific medical evidence for revealing the pathogenic principle and cure difficulty of acute lung injury and also provides important guidance for the design of therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Jianmin Li
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan, China
| | - Chanyuan Pan
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan, China
| | - Chao Tang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan, China
| | - Wenwen Tan
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan, China
| | - Hui Liu
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005 Hunan, China
| | - Jing Guan
- Department of Science and Education, The First Hospital of Changsha, Changsha, 410008 Hunan, China
| |
Collapse
|
20
|
Papoutsopoulou S, Morris L, Bayliff A, Mair T, England H, Stagi M, Bergey F, Alam MT, Sheibani-Tezerji R, Rosenstiel P, Müller W, Martins Dos Santos VAP, Campbell BJ. Effects of Human RelA Transgene on Murine Macrophage Inflammatory Responses. Biomedicines 2022; 10:biomedicines10040757. [PMID: 35453507 PMCID: PMC9027775 DOI: 10.3390/biomedicines10040757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
The NFκB transcription factors are major regulators of innate immune responses, and NFκB signal pathway dysregulation is linked to inflammatory disease. Here, we utilised bone marrow-derived macrophages from the p65-DsRedxp/IκBα-eGFP transgenic strain to study the functional implication of xenogeneic (human) RelA(p65) protein introduced into the mouse genome. Confocal imaging showed that human RelA is expressed in the cells and can translocate to the nucleus following activation of Toll-like receptor 4. RNA sequencing of lipid A-stimulated macrophages, revealed that human RelA impacts on murine gene transcription, affecting both non-NFκB and NFκB target genes, including immediate-early and late response genes, e.g., Fos and Cxcl10. Validation experiments on NFκB targets revealed markedly reduced mRNA levels, but similar kinetic profiles in transgenic cells compared to wild-type. Enrichment pathway analysis of differentially expressed genes revealed interferon and cytokine signaling were affected. These immune response pathways were also affected in macrophages treated with tumor necrosis factor. Data suggests that the presence of xenogeneic RelA protein likely has inhibitory activity, altering specific transcriptional profiles of key molecules involved in immune responses. It is therefore essential that this information be taken into consideration when designing and interpreting future experiments using this transgenic strain.
Collapse
Affiliation(s)
- Stamatia Papoutsopoulou
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (H.E.); (W.M.)
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 413 34 Larissa, Greece
- Correspondence: (S.P.); (B.J.C.)
| | - Lorna Morris
- LifeGlimmer GmbH, Markelstr. 39A, 12163 Berlin, Germany; (L.M.); (F.B.); (V.A.P.M.D.S.)
| | - Andrew Bayliff
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3GE, UK; (A.B.); (T.M.)
| | - Thomas Mair
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3GE, UK; (A.B.); (T.M.)
| | - Hazel England
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (H.E.); (W.M.)
| | - Massimiliano Stagi
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK;
| | - François Bergey
- LifeGlimmer GmbH, Markelstr. 39A, 12163 Berlin, Germany; (L.M.); (F.B.); (V.A.P.M.D.S.)
| | - Mohammad Tauqeer Alam
- Warwick Medical School, Bioinformatics RTP, University of Warwick, Coventry CV4 7AL, UK;
- Department of Biology, College of Science, United Arab Emirates University, Abu Dhabi P.O. Box 15551, United Arab Emirates
| | - Raheleh Sheibani-Tezerji
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, 6708 WE Kiel, Germany; (R.S.-T.); (P.R.)
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, 6708 WE Kiel, Germany; (R.S.-T.); (P.R.)
| | - Werner Müller
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (H.E.); (W.M.)
| | - Vitor A. P. Martins Dos Santos
- LifeGlimmer GmbH, Markelstr. 39A, 12163 Berlin, Germany; (L.M.); (F.B.); (V.A.P.M.D.S.)
- Laboratory of Systems & Synthetic Biology, Wageningen University & Research, P.O. Box 8033, 6700 EJ Wageningen, The Netherlands
| | - Barry J. Campbell
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3GE, UK; (A.B.); (T.M.)
- Correspondence: (S.P.); (B.J.C.)
| |
Collapse
|
21
|
Singh AK, Wang R, Lombardo KA, Praharaj M, Bullen CK, Um P, Davis S, Komm O, Illei PB, Ordonez AA, Bahr M, Huang J, Gupta A, Psoter KJ, Jain SK, Bivalacqua TJ, Yegnasubramanian S, Bishai WR. Dynamic single-cell RNA sequencing reveals BCG vaccination curtails SARS-CoV-2 induced disease severity and lung inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.15.484018. [PMID: 35313583 PMCID: PMC8936112 DOI: 10.1101/2022.03.15.484018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
COVID-19 continues to exact a toll on human health despite the availability of several vaccines. Bacillus Calmette Guérin (BCG) has been shown to confer heterologous immune protection against viral infections including COVID-19 and has been proposed as vaccine against SARS-CoV-2 (SCV2). Here we tested intravenous BCG vaccination against COVID-19 using the golden Syrian hamster model together with immune profiling and single cell RNA sequencing (scRNAseq). We observed that BCG reduced both lung SCV2 viral load and bronchopneumonia. This was accompanied by an increase in lung alveolar macrophages, a reversal of SCV2-mediated T cell lymphopenia, and reduced lung granulocytes. Single cell transcriptome profiling showed that BCG uniquely recruits immunoglobulin-producing plasma cells to the lung suggesting accelerated antibody production. BCG vaccination also recruited elevated levels of Th1, Th17, Treg, CTLs, and Tmem cells, and differentially expressed gene (DEG) analysis showed a transcriptional shift away from exhaustion markers and towards antigen presentation and repair. Similarly, BCG enhanced lung recruitment of alveolar macrophages and reduced key interstitial macrophage subsets, with both cell-types also showing reduced IFN-associated gene expression. Our observations indicate that BCG vaccination protects against SCV2 immunopathology by promoting early lung immunoglobulin production and immunotolerizing transcriptional patterns among key myeloid and lymphoid populations.
Collapse
Affiliation(s)
- Alok K. Singh
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Rulin Wang
- Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Kara A. Lombardo
- Johns Hopkins University, School of Medicine, Department of Urology, Baltimore, MD, USA
| | - Monali Praharaj
- Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - C. Korin Bullen
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Peter Um
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Stephanie Davis
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Oliver Komm
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Peter B. Illei
- Johns Hopkins University, School of Medicine, Department of Pathology, Baltimore, MD, USA
| | - Alvaro A. Ordonez
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore MD, USA
| | - Melissa Bahr
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore MD, USA
| | - Joy Huang
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| | - Anuj Gupta
- Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Kevin J. Psoter
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of General Pediatrics, Baltimore, MD, USA
| | - Sanjay K. Jain
- Johns Hopkins University, School of Medicine, Department of Pediatrics, Division of Infectious Diseases, Baltimore MD, USA
| | - Trinity J. Bivalacqua
- Perelman School of Medicine at the University of Pennsylvania, Division of Urology, Department of Surgery, Philadelphia, PA, USA
| | | | - William R. Bishai
- Johns Hopkins University, School of Medicine, Department of Medicine, Center for Tuberculosis Research, Baltimore, MD, USA
| |
Collapse
|
22
|
Pussinen PJ, Kopra E, Pietiäinen M, Lehto M, Zaric S, Paju S, Salminen A. Periodontitis and cardiometabolic disorders: The role of lipopolysaccharide and endotoxemia. Periodontol 2000 2022; 89:19-40. [PMID: 35244966 PMCID: PMC9314839 DOI: 10.1111/prd.12433] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lipopolysaccharide is a virulence factor of gram-negative bacteria with a crucial importance to the bacterial surface integrity. From the host's perspective, lipopolysaccharide plays a role in both local and systemic inflammation, activates both innate and adaptive immunity, and can trigger inflammation either directly (as a microbe-associated molecular pattern) or indirectly (by inducing the generation of nonmicrobial, danger-associated molecular patterns). Translocation of lipopolysaccharide into the circulation causes endotoxemia, which is typically measured as the biological activity of lipopolysaccharide to induce coagulation of an aqueous extract of blood cells of the assay. Apparently healthy subjects have a low circulating lipopolysaccharide activity, since it is neutralized and cleared rapidly. However, chronic endotoxemia is involved in the pathogenesis of many inflammation-driven conditions, especially cardiometabolic disorders. These include atherosclerotic cardiovascular diseases, obesity, liver diseases, diabetes, and metabolic syndrome, where endotoxemia has been recognized as a risk factor. The main source of endotoxemia is thought to be the gut microbiota. However, the oral dysbiosis in periodontitis, which is typically enriched with gram-negative bacterial species, may also contribute to endotoxemia. As endotoxemia is associated with an increased risk of cardiometabolic disorders, lipopolysaccharide could be considered as a molecular link between periodontal microbiota and cardiometabolic diseases.
Collapse
Affiliation(s)
- Pirkko J Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Elisa Kopra
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Milla Pietiäinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Svetislav Zaric
- Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, London, UK
| | - Susanna Paju
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aino Salminen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
23
|
Zhang S, Tang C, Wang X. Octreotide activates autophagy to alleviate lipopolysaccharide-induced human pulmonary epithelial cell injury by inhibiting the protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway. Bioengineered 2021; 13:217-226. [PMID: 34898367 PMCID: PMC8805934 DOI: 10.1080/21655979.2021.2012908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Octreotide is a synthetic octapeptide of natural somatostatin. We aimed to investigate the influence of Octreotide on lipopolysaccharide (LPS)-stimulated human pulmonary epithelial cell damage. After stimulated by LPS, BEAS-2B cells were treated with various concentrations of Octreotide. CCK-8 assay and LDH kits were to evaluate cell cytotoxicity. ELISA kits were to analyze the levels of inflammatory factors. TUNEL staining was to measure cell apoptosis. Western blot assay was used to assess the expression of apoptosis-related proteins, autophagy-related proteins and AKT/mTOR signaling-related proteins. Then, 3-methyladenine (3-MA) was adopted for treating BEAS-2B cells to determine its effects on inflammation and apoptosis. Afterward, adding AKT agonist (SC79) or mTOR antagonist (rapamycin) to explore the impact of Octreotide on autophagy. Results revealed that Octreotide notably enhanced cell viability and reduced LDH activity. The levels of inflammatory factors were significantly decreased following Octreotide treatment. Additionally, Octreotide attenuated the apoptotic capacity of LPS-induced BEAS-2B cells, led to the up-regulation of Bcl-2 protein level while cut down the protein levels of Bax and cleaved caspase3. Remarkably, the expression of autophagy-related protein LC3II/I and Beclin1 was elevated after Octreotide administration. Importantly, the suppressive effects of Octreotide on the inflammation and apoptosis of LPS-induced BEAS-2B cells was abrogated by 3-MA. Further experiments suggested that Octreotide downregulated p-AKT and mTOR expression in LPS-stimulated BEAS-2B cells. SC79 addition inhibited autophagy, evidenced by downregulated LC3II/I and Beclin1 expression while rapamycin presented the opposite effects. To conclude, Octreotide activates autophagy to alleviate LPS-induced pulmonary epithelial cell injury by inhibiting the AKT/mTOR signaling.
Collapse
Affiliation(s)
- Sumian Zhang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cijun Tang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuebin Wang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Zou B, Goodwin M, Saleem D, Jiang W, Tang J, Chu Y, Munford RS, Lu M. A highly conserved host lipase deacylates oxidized phospholipids and ameliorates acute lung injury in mice. eLife 2021; 10:70938. [PMID: 34783310 PMCID: PMC8594946 DOI: 10.7554/elife.70938] [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: 06/02/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
Abstract
Oxidized phospholipids have diverse biological activities, many of which can be pathological, yet how they are inactivated in vivo is not fully understood. Here, we present evidence that a highly conserved host lipase, acyloxyacyl hydrolase (AOAH), can play a significant role in reducing the pro-inflammatory activities of two prominent products of phospholipid oxidation, 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine and 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine. AOAH removed the sn-2 and sn-1 acyl chains from both lipids and reduced their ability to induce macrophage inflammasome activation and cell death in vitro and acute lung injury in mice. In addition to transforming Gram-negative bacterial lipopolysaccharide from stimulus to inhibitor, its most studied activity, AOAH can inactivate these important danger-associated molecular pattern molecules and reduce tissue inflammation and injury.
Collapse
Affiliation(s)
- Benkun Zou
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Michael Goodwin
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, United States
| | - Danial Saleem
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, United States
| | - Wei Jiang
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Robert S Munford
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, United States
| | - Mingfang Lu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.,Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Wen Y, Xiao H, Liu Y, Yang Y, Wang Y, Xu S, Huang S, Hou S, Liang J. Polysaccharides from Dendrobium officinale ameliorate colitis-induced lung injury via inhibiting inflammation and oxidative stress. Chem Biol Interact 2021; 347:109615. [PMID: 34363819 DOI: 10.1016/j.cbi.2021.109615] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 01/13/2023]
Abstract
It has been reported that Dendrobium officinale polysaccharides (DOPS) could alleviate colitis in animal model and suppress the activation of NLRP3 inflammasome and β-arrestin1 in vitro. However, it remains unclear whether DOPS has effect on protecting against colitis-induced pulmonary injury. The purpose of this study was to explore the protective effect and mechanism of DOPS on colitis-induced lung injury. A dextran sodium sulfate (DSS)-induced mice colitis model and lipopolysaccharide (LPS)-stimulated BEAS-2B cells model were applied in this study. The results showed that DOPS treatment restored histopathological changes, reduced inflammatory cells infiltration, pro-inflammatory cytokines levels, reactive oxygen species (ROS) formation and MDA generation, and increased anti-oxidative enzymes activities including SOD and GSH-Px in colitis mice. Further investigation showed that DOPS significantly inhibited the protein expression of TLR4, and apparently up-regulated proteins expressions of nuclear-Nrf2, HO-1 and NQO-1 in lung tissues of colitis mice and in BEAS-2B cells. These results indicated that DOPS significantly inhibited inflammation and oxidative stress to alleviate colitis-induced secondary lung injury, and its mechanisms are closely related to the inhibition of TLR4 signaling pathway and the activation of Nrf2 signaling pathway. DOPS may be a promising drug for alleviating colitis-induced lung injury.
Collapse
Affiliation(s)
- Yifan Wen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Hongyu Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Ying Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Yiqi Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, PR China
| | - Yumin Wang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, School of Chemistry, South China Normal University, Guangzhou, Guangdong, 510006, PR China
| | - Shijie Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Song Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| |
Collapse
|
26
|
Feng J, Jiang W, Cheng X, Zou B, Varley AW, Liu T, Qian G, Zeng W, Tang J, Zhao Q, Chu Y, Wei Y, Li X, Munford RS, Lu M. A host lipase prevents lipopolysaccharide-induced foam cell formation. iScience 2021; 24:103004. [PMID: 34522852 PMCID: PMC8426562 DOI: 10.1016/j.isci.2021.103004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/14/2021] [Accepted: 08/15/2021] [Indexed: 01/04/2023] Open
Abstract
Although microbe-associated molecular pattern (MAMP) molecules can promote cholesterol accumulation in macrophages, the existence of a host-derived MAMP inactivation mechanism that prevents foam cell formation has not been described. Here, we tested the ability of acyloxyacyl hydrolase (AOAH), the host lipase that inactivates gram-negative bacterial lipopolysaccharides (LPSs), to prevent foam cell formation in mice. Following exposure to small intraperitoneal dose(s) of LPSs, Aoah -/- macrophages produced more low-density lipoprotein receptor and less apolipoprotein E and accumulated more cholesterol than did Aoah +/+ macrophages. The Aoah -/- macrophages also maintained several pro-inflammatory features. Using a perivascular collar placement model, we found that Aoah -/- mice developed more carotid artery foam cells than did Aoah +/+ mice after they had been fed a high fat, high cholesterol diet, and received small doses of LPSs. This is the first demonstration that an enzyme that inactivates a stimulatory MAMP in vivo can reduce cholesterol accumulation and inflammation in arterial macrophages.
Collapse
Affiliation(s)
- Jintao Feng
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei Jiang
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200040, China
| | - Xiaofang Cheng
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Benkun Zou
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Alan W. Varley
- Department of Internal Medicine, UT-Southwestern Medical Center at Dallas, Texas 75390, USA
| | - Ting Liu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Guojun Qian
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wenjiao Zeng
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200040, China
| | - Qiang Zhao
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Yiwei Chu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanyuan Wei
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Robert S. Munford
- Antibacterial Host Defense Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mingfang Lu
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE, NHC, CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
27
|
Fei X, Ziqian Y, Bingwu Y, Min L, Xinmiao X, Zhen M, Lirong G, Song W. Aldosterone alleviates lipopolysaccharide-induced acute lung injury by regulating epithelial sodium channel through PI3K/Akt/SGK1 signaling pathway. Mol Cell Probes 2021; 57:101709. [PMID: 33713776 DOI: 10.1016/j.mcp.2021.101709] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
Reduced alveolar fluid clearance (AFC) is a major pathological feature of acute lung injury (ALI). Epithelial sodium channel (ENaC) plays a key role in regulating the transport of Na+ and clearing alveolar edema fluid effectively. ENaC has been reported to be regulated by aldosterone in the distal collecting tube of the kidney. We hypothesized whether aldosterone regulated ENaC in alveolar epithelium and correspondingly played a role in ALI. In this study we found that the expression of aldosterone synthesis encoding gene, CYP11B2, and ENaC were decreased in the lung tissue of LPS-induced ALI mice. Furthermore, aldosterone alleviated ALI by increasing the expression of ENaC-α and relieving pulmonary edema. Besides, we found that aldosterone upregulated ENaC-α through PI3K/Akt/SGK1 pathway. In conclusion, our study demonstrated that aldosterone attenuated pulmonary edema by upregulating ENaC-α through the PI3K/Akt/SGK1 pathway in LPS-induced ALI, indicating that aldosterone might be a promising adjuvant drug for ALI treatment.
Collapse
Affiliation(s)
- Xiu Fei
- Department of Blood Transfusion, Liaocheng People's Hospital, #67 Dongchangxi Road, Liaocheng, 252000, China
| | - Yu Ziqian
- Department of Clinical Laboratory, Liaocheng Veterans Hospital, #2 Gaodong Street, Liaocheng, 252000, China
| | - Yang Bingwu
- Procesion Biomedical Lab, Liaocheng People's Hospital, Medical College of Liaocheng University, #67 Dongchangxi Road, Liaocheng, 252000, China
| | - Li Min
- Procesion Biomedical Lab, Liaocheng People's Hospital, Medical College of Liaocheng University, #67 Dongchangxi Road, Liaocheng, 252000, China
| | - Xian Xinmiao
- Procesion Biomedical Lab, Liaocheng People's Hospital, Medical College of Liaocheng University, #67 Dongchangxi Road, Liaocheng, 252000, China
| | - Meng Zhen
- Procesion Biomedical Lab, Liaocheng People's Hospital, Medical College of Liaocheng University, #67 Dongchangxi Road, Liaocheng, 252000, China
| | - Guo Lirong
- Nursing School of Jilin University, Changchun, Jilin, 130021, China.
| | - Wang Song
- Procesion Biomedical Lab, Liaocheng People's Hospital, Medical College of Liaocheng University, #67 Dongchangxi Road, Liaocheng, 252000, China.
| |
Collapse
|
28
|
Wang M, Zhong H, Zhang X, Huang X, Wang J, Li Z, Chen M, Xiao Z. EGCG promotes PRKCA expression to alleviate LPS-induced acute lung injury and inflammatory response. Sci Rep 2021; 11:11014. [PMID: 34040072 PMCID: PMC8154949 DOI: 10.1038/s41598-021-90398-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI), which could be induced by multiple factors such as lipopolysaccharide (LPS), refer to clinical symptoms of acute respiratory failure, commonly with high morbidity and mortality. Reportedly, active ingredients from green tea have anti-inflammatory and anticancer properties, including epigallocatechin-3-gallate (EGCG). In the present study, protein kinase C alpha (PRKCA) is involved in EGCG protection against LPS-induced inflammation and ALI. EGCG treatment attenuated LPS-stimulated ALI in mice as manifested as improved lung injury scores, decreased total cell amounts, neutrophil amounts and macrophage amounts, inhibited the activity of MPO, decreased wet-to-dry weight ratio of lung tissues, and inhibited release of inflammatory cytokines TNF-α, IL-1β, and IL-6. PRKCA mRNA and protein expression showed to be dramatically decreased by LPS treatment while reversed by EGCG treatment. Within LPS-stimulated ALI mice, PRKCA silencing further aggravated, while PRKCA overexpression attenuated LPS-stimulated inflammation and ALI through MAPK signaling pathway. PRKCA silencing attenuated EGCG protection. Within LPS-induced RAW 264.7 macrophages, EGCG could induce PRKCA expression. Single EGCG treatment or Lv-PRKCA infection attenuated LPS-induced increases in inflammatory factors; PRKCA silencing could reverse the suppressive effects of EGCG upon LPS-stimulated inflammatory factor release. In conclusion, EGCG pretreatment inhibits LPS-induced ALI in mice. The protective mechanism might be associated with the inhibitory effects of PRKCA on proinflammatory cytokine release via macrophages and MAPK signaling pathway.
Collapse
Affiliation(s)
- Mian Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
- Department of Epidemiology and Health Statistics, School of Public Health, University of South China, Hengyang, 421001, China
| | - Hua Zhong
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xian Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Xin Huang
- Department of Epidemiology and Health Statistics, Hunan Normal University, Changsha, 410006, China
| | - Jing Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Zihao Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China
| | - Mengshi Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Xiangya RD 110, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, 410078, China.
| | - Zhenghui Xiao
- Hunan Provincial Key Laboratory of Pediatric Emergency, Hunan Children's Hospital, Changsha, 410006, China
| |
Collapse
|
29
|
Munford RS, Weiss JP, Lu M. Biochemical transformation of bacterial lipopolysaccharides by acyloxyacyl hydrolase reduces host injury and promotes recovery. J Biol Chem 2020; 295:17842-17851. [PMID: 33454018 DOI: 10.1074/jbc.rev120.015254] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/22/2020] [Indexed: 12/26/2022] Open
Abstract
Animals can sense the presence of microbes in their tissues and mobilize their own defenses by recognizing and responding to conserved microbial structures (often called microbe-associated molecular patterns (MAMPs)). Successful host defenses may kill the invaders, yet the host animal may fail to restore homeostasis if the stimulatory microbial structures are not silenced. Although mice have many mechanisms for limiting their responses to lipopolysaccharide (LPS), a major Gram-negative bacterial MAMP, a highly conserved host lipase is required to extinguish LPS sensing in tissues and restore homeostasis. We review recent progress in understanding how this enzyme, acyloxyacyl hydrolase (AOAH), transforms LPS from stimulus to inhibitor, reduces tissue injury and death from infection, prevents prolonged post-infection immunosuppression, and keeps stimulatory LPS from entering the bloodstream. We also discuss how AOAH may increase sensitivity to pulmonary allergens. Better appreciation of how host enzymes modify LPS and other MAMPs may help prevent tissue injury and hasten recovery from infection.
Collapse
Affiliation(s)
- Robert S Munford
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, Maryland, USA.
| | - Jerrold P Weiss
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA
| | - Mingfang Lu
- Department of Immunology and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Pierce CF, Brown VR, Olsen SC, Boggiatto P, Pedersen K, Miller RS, Speidel SE, Smyser TJ. Loci Associated With Antibody Response in Feral Swine ( Sus scrofa) Infected With Brucella suis. Front Vet Sci 2020; 7:554674. [PMID: 33324693 PMCID: PMC7724110 DOI: 10.3389/fvets.2020.554674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Feral swine (Sus scrofa) are a destructive invasive species widespread throughout the United States that disrupt ecosystems, damage crops, and carry pathogens of concern for the health of domestic stock and humans including Brucella suis-the causative organism for swine brucellosis. In domestic swine, brucellosis results in reproductive failure due to abortions and infertility. Contact with infected feral swine poses spillover risks to domestic pigs as well as humans, companion animals, wildlife, and other livestock. Genetic factors influence the outcome of infectious diseases; therefore, genome wide association studies (GWAS) of differential immune responses among feral swine can provide an understanding of disease dynamics and inform management to prevent the spillover of brucellosis from feral swine to domestic pigs. We sought to identify loci associated with differential antibody responses among feral swine naturally infected with B. suis using a case-control GWAS. Tissue, serum, and genotype data (68,516 bi-allelic single nucleotide polymorphisms) collected from 47 feral swine were analyzed in this study. The 47 feral swine were culture positive for Brucella spp. Of these 47, 16 were antibody positive (cases) whereas 31 were antibody negative (controls). Single-locus GWAS were performed using efficient mixed-model association eXpedited (EMMAX) methodology with three genetic models: additive, dominant, and recessive. Eight loci associated with seroconversion were identified on chromosome 4, 8, 9, 10, 12, and 18. Subsequent bioinformatic analyses revealed nine putative candidate genes related to immune function, most notably phagocytosis and induction of an inflammatory response. Identified loci and putative candidate genes may play an important role in host immune responses to B. suis infection, characterized by a detectable bacterial presence yet a differential antibody response. Given that antibody tests are used to evaluate brucellosis infection in domestic pigs and for disease surveillance in invasive feral swine, additional studies are needed to fully understand the genetic component of the response to B. suis infection and to more effectively translate estimates of Brucella spp. antibody prevalence among feral swine to disease control management action.
Collapse
Affiliation(s)
- Courtney F. Pierce
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Vienna R. Brown
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Feral Swine Damage Management Program, Fort Collins, CO, United States
| | - Steven C. Olsen
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Paola Boggiatto
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Kerri Pedersen
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Raleigh, NC, United States
| | - Ryan S. Miller
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, Fort Collins, CO, United States
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Timothy J. Smyser
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
| |
Collapse
|
31
|
Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2020; 78:1233-1261. [PMID: 33057840 PMCID: PMC7904555 DOI: 10.1007/s00018-020-03656-y] [Citation(s) in RCA: 777] [Impact Index Per Article: 155.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Toll-like receptor (TLR) 4 belongs to the TLR family of receptors inducing pro-inflammatory responses to invading pathogens. TLR4 is activated by lipopolysaccharide (LPS, endotoxin) of Gram-negative bacteria and sequentially triggers two signaling cascades: the first one involving TIRAP and MyD88 adaptor proteins is induced in the plasma membrane, whereas the second engaging adaptor proteins TRAM and TRIF begins in early endosomes after endocytosis of the receptor. The LPS-induced internalization of TLR4 and hence also the activation of the TRIF-dependent pathway is governed by a GPI-anchored protein, CD14. The endocytosis of TLR4 terminates the MyD88-dependent signaling, while the following endosome maturation and lysosomal degradation of TLR4 determine the duration and magnitude of the TRIF-dependent one. Alternatively, TLR4 may return to the plasma membrane, which process is still poorly understood. Therefore, the course of the LPS-induced pro-inflammatory responses depends strictly on the rates of TLR4 endocytosis and trafficking through the endo-lysosomal compartment. Notably, prolonged activation of TLR4 is linked with several hereditary human diseases, neurodegeneration and also with autoimmune diseases and cancer. Recent studies have provided ample data on the role of diverse proteins regulating the functions of early, late, and recycling endosomes in the TLR4-induced inflammation caused by LPS or phagocytosis of E. coli. In this review, we focus on the mechanisms of the internalization and intracellular trafficking of TLR4 and CD14, and also of LPS, in immune cells and discuss how dysregulation of the endo-lysosomal compartment contributes to the development of diverse human diseases.
Collapse
Affiliation(s)
- Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland.
| | - Marta Matyjek
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| |
Collapse
|
32
|
Tuominen I, Fuqua BK, Pan C, Renaud N, Wroblewski K, Civelek M, Clerkin K, Asaryan A, Haroutunian SG, Loureiro J, Borawski J, Roma G, Knehr J, Carbone W, French S, Parks BW, Hui ST, Mehrabian M, Magyar C, Cantor RM, Ukomadu C, Lusis AJ, Beaven SW. The Genetic Architecture of Carbon Tetrachloride-Induced Liver Fibrosis in Mice. Cell Mol Gastroenterol Hepatol 2020; 11:199-220. [PMID: 32866618 PMCID: PMC7674618 DOI: 10.1016/j.jcmgh.2020.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis is a multifactorial trait that develops in response to chronic liver injury. Our aim was to characterize the genetic architecture of carbon tetrachloride (CCl4)-induced liver fibrosis using the Hybrid Mouse Diversity Panel, a panel of more than 100 genetically distinct mouse strains optimized for genome-wide association studies and systems genetics. METHODS Chronic liver injury was induced by CCl4 injections twice weekly for 6 weeks. Four hundred thirty-seven mice received CCl4 and 256 received vehicle, after which animals were euthanized for liver histology and gene expression. Using automated digital image analysis, we quantified fibrosis as the collagen proportionate area of the whole section, excluding normal collagen. RESULTS We discovered broad variation in fibrosis among the Hybrid Mouse Diversity Panel strains, demonstrating a significant genetic influence. Genome-wide association analyses revealed significant and suggestive loci underlying susceptibility to fibrosis, some of which overlapped with loci identified in mouse crosses and human population studies. Liver global gene expression was assessed by RNA sequencing across the strains, and candidate genes were identified using differential expression and expression quantitative trait locus analyses. Gene set enrichment analyses identified the underlying pathways, of which stellate cell involvement was prominent, and coexpression network modeling identified modules associated with fibrosis. CONCLUSIONS Our results provide a rich resource for the design of experiments to understand mechanisms underlying fibrosis and for rational strain selection when testing antifibrotic drugs.
Collapse
Affiliation(s)
- Iina Tuominen
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Brie K Fuqua
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Calvin Pan
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Nicole Renaud
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Kevin Wroblewski
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Mete Civelek
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Kara Clerkin
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ashot Asaryan
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sara G Haroutunian
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Joseph Loureiro
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jason Borawski
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | | - Samuel French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Brian W Parks
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Simon T Hui
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Margarete Mehrabian
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Clara Magyar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Rita M Cantor
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Chinweike Ukomadu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Aldons J Lusis
- Departments of Medicine, Microbiology and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Simon W Beaven
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases at UCLA and Pfleger Liver Institute, David Geffen School of Medicine at UCLA, Los Angeles, California.
| |
Collapse
|
33
|
Ma Xing Shi Gan Decoction Protects against PM2.5-Induced Lung Injury through Suppression of Epithelial-to-Mesenchymal Transition (EMT) and Epithelial Barrier Disruption. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7176589. [PMID: 32655666 PMCID: PMC7317335 DOI: 10.1155/2020/7176589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
This research was designed to explore the effect of Ma Xing Shi Gan decoction (MXD) in alleviating particulate matter less than 2.5 μm in diameter (PM2.5) induced lung injury from the perspective of epithelial barrier protection and inhibition of epithelial-to-mesenchymal transition (EMT). Rats were exposed to PM2.5 to establish a lung injury model in vivo, and a PM2.5-stimulated primary cultured type II alveolar epithelial cell model was introduced in vitro. Our results indicated that MXD alleviated the weight loss and pathologic changes and improved the epithelial barrier dysfunction. MXD also significantly inhibited the TGF-β/Smad3 pathway, increased the level of ZO-1 and claudin-5, and reversed the EMT process. Notably, the protection of MXD was abolished by TGF-β in vitro. Our results indicated that MXD has a protection against PM2.5-induced lung injury. The proposed mechanism is reversing PM2.5-induced EMT through inhibiting TGF-β/Smad3 pathway and then upregulating the expression of tight-junction proteins.
Collapse
|
34
|
HSPA12A attenuates lipopolysaccharide-induced liver injury through inhibiting caspase-11-mediated hepatocyte pyroptosis via PGC-1α-dependent acyloxyacyl hydrolase expression. Cell Death Differ 2020; 27:2651-2667. [PMID: 32332915 PMCID: PMC7429872 DOI: 10.1038/s41418-020-0536-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/05/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
Liver dysfunction is strongly associated with poor survival of sepsis patients. Cytosolic lipopolysaccharide (LPS) sensing by Caspase-4/5/11 for pyroptosis activation is a major driver of the development of sepsis. Studies in macrophages and endothelial cells have demonstrated that LPS is inactivated by acyloxyacyl hydrolase (AOAH) and leading to desensitizing Caspase-4/5/11 to LPS. However, little is known about the cytosolic LPS-induced pyroptosis in hepatocytes during sepsis. Heat shock protein 12A (HSPA12A) is a novel member of the HSP70 family. Here, we report that LPS increased HSPA12A nuclear translocation in hepatocytes, while knockout of HSPA12A (Hspa12a−/−) in mice promoted LPS-induced acute liver injury. We also noticed that the LPS-induced Caspase-11 activation and its cleavage of gasdermin D (GSDMD) to produce the membrane pore-forming GSDMDNterm (markers of pyroptosis) were greater in livers of Hspa12a−/− mice compared with its wild type controls. Loss- and gain-of-function studies showed that HSPA12A deficiency promoted, whereas HSPA12A overexpression inhibited, cytosolic LPS accumulation, Caspase-11 activation and GSDMDNterm generation in primary hepatocytes following LPS incubation. Notably, LPS-induced AOAH expression was suppressed by HSPA12A deficiency, whereas AOAH overexpression reversed the HSPA12A deficiency-induced promotion of LPS-evoked and Caspase-11-mediated pyroptosis of hepatocytes. In-depth molecular analysis showed that HSPA12A interacted directly with peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and increased its nuclear translocation, thereby inducing AOAH expression for cytosolic LPS inactivation, which ultimately leading to inhibition of the Caspase-11 mediated pyroptosis of hepatocytes. Taken together, these findings revealed HSPA12A as a novel player against LPS-induced liver injury by inhibiting cytosolic LPS-induced hepatocyte pyroptosis via PGC-1α-mediated AOAH expression. Therefore, targeting hepatocyte HSPA12A represents a viable strategy for the management of liver injury in sepsis patients.
Collapse
|
35
|
Murray SM, Zhang Y, Douek DC, Sekaly RP. Myeloid Cells Enriched for a Dendritic Cell Population From People Living With HIV Have Altered Gene Expression Not Restored by Antiretroviral Therapy. Front Immunol 2020; 11:261. [PMID: 32194550 PMCID: PMC7064632 DOI: 10.3389/fimmu.2020.00261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/31/2020] [Indexed: 12/22/2022] Open
Abstract
Antiretroviral therapy (ART) for human immunodeficiency virus (HIV) infections has been designed to optimize CD4 T-cell survival and limit HIV replication. Cell types other than CD4 T cells such as monocytes/macrophage, dendritic cells, and granulocytes (collectively known as myeloid cells), are generally not considered in the development of ART protocols. Myeloid dendritic cells (mDCs) are the most potent inducers of CD4 T-cell activation and central to the regulation of immune responses. mDCs in the blood are decreased in number, altered in function, and implicated in promoting HIV latency in people living with HIV (PLWH). We found that cells enriched for mDC in PLWH had transcriptional changes compared to mDC from HIV uninfected individuals, some of which were not completely restored by ART. In contrast, other mDC functions such as interleukin-1 signaling and type I interferon pathways were restored by ART. Some of the transcriptional changes in mDC not completely reversed by ART were enriched in genes that are classically associated with cells of the monocyte/macrophage lineage, but new single-cell RNA sequencing studies show that they are also expressed by a subset of mDC. A cellular enzyme, acyloxyacyl hydrolase (AOAH), important for lipopolysaccharide (LPS) detoxification, had increased transcription in mDC of PLWH, not restored by ART. It is possible that one reason ART is not completely successful in PLWH is the failure to phenotypically change the mDCs. Thus, inability of ART to be completely effective might involve myeloid cells and the failure to restore mDC function as measured by gene transcription. We suggest that mDC and myeloid cells should be considered in future combination ART development.
Collapse
Affiliation(s)
- Shannon M Murray
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yuwei Zhang
- Vaccine and Gene Therapy Institute Florida, Port St. Lucie, FL, United States
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rafick P Sekaly
- Vaccine and Gene Therapy Institute Florida, Port St. Lucie, FL, United States
| |
Collapse
|
36
|
The Antitumor Efficacy of β-Elemene by Changing Tumor Inflammatory Environment and Tumor Microenvironment. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6892961. [PMID: 32149121 PMCID: PMC7054771 DOI: 10.1155/2020/6892961] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
Inflammatory mediators and inflammatory cells in the inflammatory microenvironment promote the transformation of normal cells to cancer cells in the early stage of cancer, promote the growth and development of cancer cells, and induce tumor immune escape. The monomeric active ingredient β-elemene is extracted from the traditional Chinese medicine Curcuma wenyujin and has been proven to have good anti-inflammatory and antitumor activities in clinical applications for more than 20 years in China. Recent studies have found that this traditional Chinese medicine plays a vital role in macrophage infiltration and M2 polarization, as well as in regulating immune disorders, and it even regulates the transcription factors NF-κB and STAT3 to alter inflammation, tumorigenesis, and development. In addition, β-elemene regulates not only different inflammatory factors (such as TNF-α, IFN, TGF-β, and IL-6/10) but also oxidative stress in vivo and in vitro. The excellent anti-inflammatory and antitumor effects of β-elemene and its ability to alter the inflammatory microenvironment of tumors have been gradually elaborated. Although the study of monomeric active ingredients in traditional Chinese medicines is insufficient in terms of quality and quantity, the pharmacological effects of more active ingredients of traditional Chinese medicines will be revealed after β-elemene.
Collapse
|
37
|
Amornphimoltham P, Yuen PST, Star RA, Leelahavanichkul A. Gut Leakage of Fungal-Derived Inflammatory Mediators: Part of a Gut-Liver-Kidney Axis in Bacterial Sepsis. Dig Dis Sci 2019; 64:2416-2428. [PMID: 30863955 DOI: 10.1007/s10620-019-05581-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/06/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening response to systemic infection. In addition to frank gastrointestinal (GI) rupture/puncture, sepsis can also be exacerbated by translocation of pathogen-associated molecular patterns (PAMPs) from the GI tract to the systemic circulation (gut origin of sepsis). In the human gut, Gram-negative bacteria and Candida albicans are abundant, along with their major PAMP components, endotoxin (LPS) and (1 → 3)-β-D-glucan (BG). Whereas the influence of LPS in bacterial sepsis has been studied extensively, exploration of the role of BG in bacterial sepsis is limited. Post-translocation, PAMPs enter the circulation through lymphatics and the portal vein, and are detoxified and then excreted via the liver and the kidney. Sepsis-induced liver and kidney injury might therefore affect the kinetics and increase circulating PAMPs. In this article, we discuss the current knowledge of the impact of PAMPs from both gut mycobiota and microbiota, including epithelial barrier function and the "gut-liver-kidney axis," on bacterial sepsis severity.
Collapse
Affiliation(s)
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Asada Leelahavanichkul
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, 10330, Thailand. .,Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
38
|
Wu K, Xiu Y, Zhou P, Qiu Y, Li Y. A New Use for an Old Drug: Carmofur Attenuates Lipopolysaccharide (LPS)-Induced Acute Lung Injury via Inhibition of FAAH and NAAA Activities. Front Pharmacol 2019; 10:818. [PMID: 31379583 PMCID: PMC6659393 DOI: 10.3389/fphar.2019.00818] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/24/2019] [Indexed: 01/10/2023] Open
Abstract
Acute lung injury (ALI), characterized by a severe inflammatory process, is a complex syndrome that can lead to multisystem organ failure. Fatty acid amide hydrolase (FAAH) and N-acylethanolamine acid amidase (NAAA) are two potential therapeutic targets for inflammation-related diseases. Herein, we identified carmofur, a 5-fluorouracil-releasing drug and clinically used as a chemotherapeutic agent, as a dual FAAH and NAAA inhibitor. In Raw264.7 macrophages, carmofur effectively reduced the mRNA expression of pro-inflammatory factors, including IL-1β, IL-6, iNOS, and TNF-α, and down-regulated signaling proteins of the nuclear transcription factor κB (NF-κB) pathway. Furthermore, carmofur significantly ameliorated the inflammatory responses and promoted resolution of pulmonary injury in lipopolysaccharide (LPS)-induced ALI mice. The pharmacological effects of carmofur were partially blocked by peroxisome proliferator-activated receptor-α (PPARα) antagonist MK886 and cannabinoid receptor 2 (CB2) antagonist SR144528, indicating that carmofur attenuated LPS-induced ALI in a PPARα- and CB2-dependent mechanism. Our study suggested that carmofur might be a novel therapeutic agent for ALI, and drug repurposing may provide us effective therapeutic strategies for ALI.
Collapse
Affiliation(s)
- Kangni Wu
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yanghui Xiu
- Eye Institute & Affiliated Xiamen Eye Center, Xiamen University, Xiamen, China
| | - Pan Zhou
- Eye Institute & Affiliated Xiamen Eye Center, Xiamen University, Xiamen, China.,Institute of Hematology, Medical College of Xiamem University, Xiamen, China
| | - Yan Qiu
- Eye Institute & Affiliated Xiamen Eye Center, Xiamen University, Xiamen, China.,Institute of Hematology, Medical College of Xiamem University, Xiamen, China
| | - Yuhang Li
- Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian, China.,CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian, China
| |
Collapse
|
39
|
CXCL16 Induces the Progression of Pulmonary Fibrosis through Promoting the Phosphorylation of STAT3. Can Respir J 2019; 2019:2697376. [PMID: 31379980 PMCID: PMC6652085 DOI: 10.1155/2019/2697376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/18/2019] [Accepted: 03/26/2019] [Indexed: 01/21/2023] Open
Abstract
Aim The transmembrane chemokine (C-X-C motif) ligand 16 (CXCL16) plays a vital role in the pathogenesis of organ fibrosis, including the liver and kidney. However, the detailed biological function of CXCL16 is still not fully understood in the progression of pulmonary fibrosis (PF). The aim of present study is to examine the function of CXCL16 in PF. Materials and Methods In this study, we constructed the PF model on mouse by using bleomycin and analyzed the effect of the mouse recombinant protein CXCL16 on mouse lung fibroblast L929 (LF) as well. To further examine the connection between CXCL16 and STAT3 in mouse LF cells, the STAT3 inhibitor AG490 was utilized to inhibit the expression of STAT3. Meanwhile, lipopolysaccharide was used to enhance the phosphorylation of STAT3 (p-STAT3) in mouse LF cells. Results Our results indicated that the level of CXCL16/CXCR6 was significantly upregulated in the mouse PF model. Moreover, the level of p-STAT3 was also promoted. In addition, the mouse recombinant protein CXCL16 not only contributed to the proliferation of mouse LF cells but also induced the expression of p-STAT3 in LF cells. However, the effect of CXCL16 was deeply abolished by the STAT3 inhibitor AG490 in LF cells. Meanwhile, the antibody of CXCL16 deeply reduced the phosphorylation of STAT3 in lipopolysaccharide (LPS) cultured cells. Conclusions All these results demonstrated that CXCL16 promoted the phosphorylation of STAT3 and further demonstrated that STAT3 was a critical component in CXCL16/CXCR6 signaling pathway. This research not only enhanced the comprehension of CXCL16 but also indicated its potential value as a target in the treatment for human PF.
Collapse
|
40
|
Skirecki T, Cavaillon JM. Inner sensors of endotoxin - implications for sepsis research and therapy. FEMS Microbiol Rev 2019; 43:239-256. [PMID: 30844058 DOI: 10.1093/femsre/fuz004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2025] Open
Abstract
Despite great efforts and numerous clinical trials, there is still a major need for effective therapies for sepsis. Neutralization or elimination of bacterial toxins remains a promising approach. The understanding of the interaction of the endotoxin (lipopolysaccharide, LPS) of Gram-negative bacteria with its cellular receptor, namely the CD14/TLR4/MD2 complex, was a major breakthrough. Unfortunately, clinical trials for sepsis on the neutralization of LPS or on the inhibition of TLR4 signaling failed whereas those on LPS removal remain controversial. Recent discoveries of another class of LPS receptors localized within the cytoplasm, namely caspase-11 in mice and caspases-4/5 in humans, have renewed interest in the field. These provide new potential targets for intervention in sepsis pathogenesis. Since cytoplasmic recognition of LPS induces non-canonical inflammasome pathway, a potentially harmful host response, it is conceivable to therapeutically target this mechanism. However, a great deal of care should be used in the translation of research on the non-canonical inflammasome inhibition due to multiple inter-species differences. In this review, we summarize the knowledge on endotoxin sensing in sepsis with special focus on the intracellular sensing. We also highlight the murine versus human differences and discuss potential therapeutic approaches addressing the newly discovered pathways.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care Medicine, Centre of Postgraduate Medical Education, Marymoncka 99/103 Street, 01-813 Warsaw, Poland
| | - Jean-Marc Cavaillon
- Experimental Neuropathology Unit, Institut Pasteur, 28 rue Dr. Roux, 75015 Paris, France
| |
Collapse
|
41
|
Qu L, Chen C, He W, Chen Y, Li Y, Wen Y, Zhou S, Jiang Y, Yang X, Zhang R, Shen L. Glycyrrhizic acid ameliorates LPS-induced acute lung injury by regulating autophagy through the PI3K/AKT/mTOR pathway. Am J Transl Res 2019; 11:2042-2055. [PMID: 31105816 PMCID: PMC6511780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/07/2019] [Indexed: 06/09/2023]
Abstract
Acute lung injury (ALI) is a major pathological issue characterized by serious inflammatory response, and a major clinically critical illness with high morbidity and mortality. Glycyrrhizic acid (GA) is a major bioactive constituent isolated from traditional Chinese herb licorice, which has been reported to have positive effects on inflammation. Nevertheless, the effects of GA on lipopolysaccharide (LPS)-treated ALI in mice have not been reported. The purpose of our study is to investigate the inhibitory effects of GA on ALI treated by LPS and to elucidate its possible mechanisms. We found that GA significantly attenuated lung injury and decreased the production of inflammatory factors TNF-α, IL-1β, and HMGB1 with LPS treatment. GA induced autophagy which was showed by enhanced number of autophagosomes through upregulating the protein levels of LC3-II/I and Beclin-1 and downregulating SQSTM1/P62. Moreover, pre-treatment of 3-Methyladenine (3-MA), an autophagy inhibitor, reversed the inhibiting effects of GA on the secretion of inflammatory factors in ALI. The PI3K/AKT/mTOR pathway was associated with GA-induced autophagy under ALI induced by LPS. In conclusion, this study indicated that GA inhibited the production of inflammatory factors in LPS-induced ALI by regulating the PI3K/AKT/mTOR pathway related autophagy, which may provide a novel therapeutic perspective of GA in ameliorating ALI.
Collapse
Affiliation(s)
- Lihua Qu
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Chao Chen
- Department of Pathology and Key Laboratory of Cancer Stem Cells and Translational Medicine, Hunan Normal University School of MedicineChangsha 410013, China
| | - Wei He
- Department of Ultrasonography, The Third Xiangya Hospital of Central South UniversityChangsha 410013, China
| | - Yangye Chen
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yi Li
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yi Wen
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Sichun Zhou
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yiqun Jiang
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Xiaoping Yang
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Ran Zhang
- Department of Immunology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Li Shen
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| |
Collapse
|
42
|
Abstract
Lung infections caused by bacteria can induce a spectrum of immune responses, which is in part determined by the level of exposure and the degree of the host response. The host response involves pattern recognition receptors (PRRs) which sense pathogen and damage associated molecular patterns. Therefore, models of acute lung inflammation are necessary for further understanding the role of the innate immune system during bacterial infection in humans. Mice are a widely used model organism for studying important aspects of human lung pathogenesis, including acute and chronic inflammatory diseases. Klebsiella pneumoniae is a gram-negative bacterium that is commonly associated with respiratory infections, especially in a hospital setting. In this protocol, we describe a model of bacteria-mediated lung inflammation using K. pneumoniae. After a single intratracheal administration of K. pneumoniae, mice showed a strong level of Th1-mediated immune activation in the lungs. The model described here, while optimized for K. pneumonia, can be performed using other bacteria, fungi, and viruses as well.
Collapse
|
43
|
Liang Y, Fan C, Yan X, Lu X, Jiang H, Di S, Ma Z, Feng Y, Zhang Z, Feng P, Feng X, Feng J, Jin F. Berberine ameliorates lipopolysaccharide‐induced acute lung injury via the
PERK
‐mediated
Nrf2/HO‐1
signaling axis. Phytother Res 2018; 33:130-148. [PMID: 30346043 DOI: 10.1002/ptr.6206] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/17/2018] [Accepted: 09/08/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Yuan Liang
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
- Department of Respiration Kunming General Hospital of the People's Liberation Army Kunming China
| | - Chongxi Fan
- Department of Biomedical Engineering The Fourth Military Medical University Xi'an China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Xi Lu
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Hua Jiang
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital The Fourth Military Medical University Xi'an China
- Department of Cardiothoracic Surgery The 97th Hospital of PLA Xuzhou China
| | - Zhengbin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Pan Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital The Fourth Military Medical University Xi'an China
| | - Faguang Jin
- Department of Respiration, Tangdu Hospital The Fourth Military Medical University Xi'an China
| |
Collapse
|
44
|
Zhang H, Wang Z, Liu R, Qian T, Liu J, Wang L, Chu Y. Reactive oxygen species stimulated pulmonary epithelial cells mediate the alveolar recruitment of FasL
+
killer B cells in LPS‐induced acute lung injuries. J Leukoc Biol 2018; 104:1187-1198. [DOI: 10.1002/jlb.3a0218-075r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 07/19/2018] [Accepted: 08/14/2018] [Indexed: 11/11/2022] Open
Affiliation(s)
- Hushan Zhang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Zhiming Wang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Ronghua Liu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Tingting Qian
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Jiajing Liu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Luman Wang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
- Biotherapy Research CenterFudan University Shanghai China
| | - Yiwei Chu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
- Biotherapy Research CenterFudan University Shanghai China
| |
Collapse
|
45
|
Liu J, Chen Q, Liu S, Yang X, Zhang Y, Huang F. Sini decoction alleviates E. coli induced acute lung injury in mice via equilibrating ACE-AngII-AT1R and ACE2-Ang-(1-7)-Mas axis. Life Sci 2018; 208:139-148. [DOI: 10.1016/j.lfs.2018.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 11/13/2022]
|
46
|
Qian G, Jiang W, Zou B, Feng J, Cheng X, Gu J, Chu T, Niu C, He R, Chu Y, Lu M. LPS inactivation by a host lipase allows lung epithelial cell sensitization for allergic asthma. J Exp Med 2018; 215:2397-2412. [PMID: 30021797 PMCID: PMC6122967 DOI: 10.1084/jem.20172225] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/16/2018] [Accepted: 07/06/2018] [Indexed: 01/17/2023] Open
Abstract
This study provides strong evidence that intestinal commensal LPS desensitizes lung epithelial cells and therefore diminishes allergic responses to inhaled allergens. A host lipase, acyloxyacyl hydrolase (AOAH), prevents the desensitization by inactivating commensal LPS. Allergic asthma is a chronic inflammatory disease primarily mediated by Th2 immune mechanisms. Numerous studies have suggested that early life exposure to lipopolysaccharide (LPS) is negatively associated with allergic asthma. One proposed mechanism invokes desensitization of lung epithelial cells by LPS. We report here that acyloxyacyl hydrolase (AOAH), a host lipase that degrades and inactivates LPS, renders mice more susceptible to house dust mite (HDM)–induced allergic asthma. Lung epithelial cells from Aoah−/− mice are refractory to HDM stimulation, decreasing dendritic cell activation and Th2 responses. Antibiotic treatment that diminished commensal LPS-producing bacteria normalized Aoah−/− responses to HDM, while giving LPS intrarectally ameliorated asthma. Aoah−/− mouse feces, plasma, and lungs contained more bioactive LPS than did those of Aoah+/+ mice. By inactivating commensal LPS, AOAH thus prevents desensitization of lung epithelial cells. An enzyme that prevents severe lung inflammation/injury in Gram-negative bacterial pneumonia has the seemingly paradoxical effect of predisposing to a Th2-mediated airway disease.
Collapse
Affiliation(s)
- Guojun Qian
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Benkun Zou
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jintao Feng
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofang Cheng
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianqing Chu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Niu
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rui He
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mingfang Lu
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Inflammation-restricted anti-inflammatory activities of a N -acylethanolamine acid amidase (NAAA) inhibitor F215. Pharmacol Res 2018; 132:7-14. [DOI: 10.1016/j.phrs.2018.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 11/21/2022]
|
48
|
Bidne KL, Dickson MJ, Ross JW, Baumgard LH, Keating AF. Disruption of female reproductive function by endotoxins. Reproduction 2018; 155:R169-R181. [DOI: 10.1530/rep-17-0406] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
Endotoxemia can be caused by obesity, environmental chemical exposure, abiotic stressors and bacterial infection. Circumstances that deleteriously impact intestinal barrier integrity can induce endotoxemia, and controlled experiments have identified negative impacts of lipopolysaccharide (LPS; an endotoxin mimetic) on folliculogenesis, puberty onset, estrus behavior, ovulation, meiotic competence, luteal function and ovarian steroidogenesis. In addition, neonatal LPS exposures have transgenerational female reproductive impacts, raising concern about early life contacts to this endogenous reproductive toxicant. Aims of this review are to identify physiological stressors causing endotoxemia, to highlight potential mechanism(s) by which LPS compromises female reproduction and identify knowledge gaps regarding how acute and/or metabolic endotoxemia influence(s) female reproduction.
Collapse
|
49
|
Abstract
LPS is a potent bacterial endotoxin that triggers the innate immune system. Proper recognition of LPS by pattern-recognition receptors requires a full complement of typically six acyl chains in the lipid portion. Acyloxyacyl hydrolase (AOAH) is a host enzyme that removes secondary (acyloxyacyl-linked) fatty acids from LPS, rendering it immunologically inert. This activity is critical for recovery from immune tolerance that follows Gram-negative infection. To understand the molecular mechanism of AOAH function, we determined its crystal structure and its complex with LPS. The substrate's lipid moiety is accommodated in a large hydrophobic pocket formed by the saposin and catalytic domains with a secondary acyl chain inserted into a narrow lateral hydrophobic tunnel at the active site. The enzyme establishes dispensable contacts with the phosphate groups of LPS but does not interact with its oligosaccharide portion. Proteolytic processing allows movement of an amphipathic helix possibly involved in substrate access at membranes.
Collapse
|