101
|
He F, Gao F, Cai N, Jiang M, Wu C. Chlorogenic acid enhances alveolar macrophages phagocytosis in acute respiratory distress syndrome by activating G protein-coupled receptor 37 (GPR 37). PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154474. [PMID: 36194973 DOI: 10.1016/j.phymed.2022.154474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 09/10/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Impaired alveolar macrophages phagocytosis can contribute to pathogenesis of acute respiratory distress syndrome (ARDS) and negatively impacts clinical outcomes. Chlorogenic acid (CGA) is a naturally occurring polyphenolic compound with potential anti-inflammatory and antioxidant bioactivities. Studies have shown that CGA plays a protective role in ARDS, however, the precise protective mechanism of CGA against ARDS, is still unclear. PURPOSE The aim of this study was to investigate whether CGA enhances alveolar macrophages phagocytosis to attenuate lung injury during ARDS. METHODS RAW264.7 cells were stimulated with lipopolysaccharides (100 μg/ml for 24 h) and treated with CGA (100, 200, and 400 μM CGA for 1 h) to measure pro-inflammatory cytokine levels, GPR37 expression and macrophages phagocytosis. Mouse models of ARDS induced by cecal ligation and perforation (CLP) surgery were treated with CGA (100 or 200 mg/kg) to investigate lung inflammatory injury and alveolar macrophages phagocytosis. Computational modeling was performed to examine potential binding sites of G protein-coupled receptor 37 (GPR37) with CGA, and the results were validated by interfering with the binding sites. RESULT In vitro, CGA notably ameliorated inflammatory response and increased phagocytosis in lipopolysaccharides-induced RAW264.7 cells. In vivo, CGA administration significantly alleviated lung inflammatory injury, decreased the bacteria load in the lung, promoted alveolar macrophages phagocytosis and improved the survival rate in mice with CLP-induced ARDS. Moreover, CGA markedly upregulated the expression of GPR37 in vivo and in vitro. However, the protective effect of CGA against ARDS were reversed after silencing the expression of GPR37. CONCLUSION CGA has a protective effect against ARDS and may enhance alveolar macrophages phagocytosis and attenuate lung inflammatory injury by upregulating GPR37 expression.
Collapse
Affiliation(s)
- Fei He
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China..
| | - Fengjuan Gao
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| | - Nan Cai
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| | - Min Jiang
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| | - Chao Wu
- Department of Infectious Disease, Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
102
|
Yang Y, Wei S, Li Q, Chu K, Zhou Y, Xue L, Tian H, Tao S. Vitamin D protects silica particles induced lung injury by promoting macrophage polarization in a KLF4-STAT6 manner. J Nutr Biochem 2022; 110:109148. [PMID: 36049670 DOI: 10.1016/j.jnutbio.2022.109148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/10/2022] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Silicosis is one of the severest occupational diseases worldwide, manifesting as infiltration of inflammatory cells, excessive secretion of pro-inflammatory mediators and pulmonary diffuse fibrosis. Macrophages polarization to M2 is one of the major strategies that attenuates inflammatory response. Our previous study found that vitamin D could protect against silica-induced lung injury by damping the secretion of pro-inflammatory cytokines. Here we further identified that vitamin D attenuated silica particles-induced lung inflammation by regulating macrophage polarization in a KLF4-STAT6 manner. Myeloid-specific Stat6 knockout (cKO) mice were generated for in vivo studies. Primary macrophages purified from bronchoalveolar lavage fluid (BALF) of wildtype or Stat6 cKO mice and differentiated THP-1 cells were used for in vitro studies. Vitamin D was found to promote alveolar macrophage polarizing to M2 phenotype through the STAT6 signaling pathway, as demonstrated by worse lung inflammation and ablated protection of vitamin D in silica particles-instilled Stat6 cKO mice. Mechanismly, vitamin D upregulated KLF4 expression in the alveolar macrophage, which synergistically activated STAT6. Additionally, KLF4 was found to upregulate macrophages autophagy, which protected them from silica particles-induced oxidative stress and cell apoptosis. The protective effects of vitamin D were dismissed by silencing KLF4. Our study demonstrates the potential mechanism of vitamin D-mediated macrophage polarization and reveals the therapeutic application of vitamin D in inflammatory disease.
Collapse
Affiliation(s)
- Youjing Yang
- Chongqing University Central Hospital and Chongqing Emergency Medical Center, Chongqing, China; School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shuhui Wei
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Qianmin Li
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Kaimiao Chu
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yujia Zhou
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Lian Xue
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hailin Tian
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shasha Tao
- Chongqing University Central Hospital and Chongqing Emergency Medical Center, Chongqing, China; School of Public Health, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
103
|
Enomoto N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int J Mol Sci 2022; 23:ijms232315027. [PMID: 36499351 PMCID: PMC9736972 DOI: 10.3390/ijms232315027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Interstitial lung diseases (ILD) are relatively rare and sometimes become life threatening. In particular, rapidly progressive ILD, which frequently presents as acute lung injury (ALI) on lung histopathology, shows poor prognosis if proper and immediate treatments are not initiated. These devastating conditions include acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF), clinically amyopathic dermatomyositis (CADM), epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI)-induced lung injury, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection named coronavirus disease 2019 (COVID-19). In this review, clinical information, physical findings, laboratory examinations, and findings on lung high-resolution computed tomography and lung histopathology are presented, focusing on majorly damaged cells in each disease. Furthermore, treatments that should be immediately initiated in clinical practice for each disease are illustrated to save patients with these diseases.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; ; Tel.: +81-53-435-2263; Fax: +81-53-435-2354
- Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
| |
Collapse
|
104
|
Jin H, Luo R, Li J, Zhao H, Ouyang S, Yao Y, Chen D, Ling Z, Zhu W, Chen M, Liao X, Pi J, Huang G. Inhaled platelet vesicle-decoyed biomimetic nanoparticles attenuate inflammatory lung injury. Front Pharmacol 2022; 13:1050224. [PMID: 36523494 PMCID: PMC9745055 DOI: 10.3389/fphar.2022.1050224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/16/2022] [Indexed: 01/04/2024] Open
Abstract
Acute lung injury (ALI) is an inflammatory response which causes serious damages to alveolar epithelia and vasculature, and it still remains high lethality and mortality with no effective treatment. Based on the inflammatory homing of platelets and cell membrane cloaking nanotechnology, in this study we developed a biomimetic anti-inflammation nanoparticle delivery system for ALI treatment. PM@Cur-RV NPs were designed by combining the poly (lactic-co-glycolic acid) nanoparticles (NPs) coated with platelet membrane vesicles (PM) for the purpose of highly targeting delivery of curcumin (Cur) and resveratrol (RV) to inflammatory lungs. PM@Cur-RV NPs showed good biocompatibility and biosafety both in vitro and in vivo. Accumulation of NPs into lung tract was observed after inhaled NPs. Remarkably, the inhalation of PM@Cur-RV NPs effectively inhibited lung vascular injury evidenced by the decreased lung vascular permeability, and the reduced proinflammatory cytokine burden in an ALI mouse model. The analysis of infiltrated macrophages in the lungs showed that the Cur-RV-modulated macrophage polarized towards M2 phenotype and the decreased histone lactylation might contribute to their anti-inflammation effects. Together, this work highlights the potential of inhalation of biomimetic nanoparticle delivery of curcumin and resveratrol for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Renxing Luo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongxia Zhao
- School of Biomedical and Pharmaceutical Science, Guangdong University of Technology, Guangzhou, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yinlian Yao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Dongyan Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zijie Ling
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Weicong Zhu
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Meijun Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianping Liao
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
105
|
Watanabe T, Watanabe Y, Ikeda N, Aihara M, Yamaguchi Y. Serum levels of
C‐C
motif chemokine ligand 2 and interleukin‐8 as possible biomarkers in patients with toxic epidermal necrolysis accompanied by acute respiratory distress syndrome. J Dermatol 2022; 50:500-510. [PMID: 36419353 DOI: 10.1111/1346-8138.16647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/27/2022]
Abstract
Toxic epidermal necrolysis (TEN) is a fatal cutaneous adverse reaction that occasionally affects multiple organs. Acute respiratory distress syndrome (ARDS) is a rare complication that can cause rapid and potentially fatal pulmonary dysfunction. However, the mechanisms underlying TEN-induced ARDS remain unknown. This retrospective single-center study aimed to identify potential biomarkers for predicting ARDS onset in TEN patients. Pre-treatment serum samples were collected from 16 TEN patients and 16 healthy controls (HCs). The serum levels of cytokines/chemokines were determined using the Luminex Assay Human Premixed Multi-analyte kit. The expression levels of cytokines and chemokines in the skin were examined via immunohistochemistry. The serum levels of C-C motif chemokine ligand 2 (CCL2), interleukin (IL)-6, and IL-8 were significantly higher in TEN patients with ARDS than in those without ARDS and in HCs, whereas those of CCL2 and IL-8 were not significantly different between TEN patients without ARDS and HCs. There was no significant difference in CCL2 and IL-8 expression in the skin between TEN patients with and without ARDS. Interestingly, there were no significant differences in the cytokine/chemokine levels between TEN and other organ damage, other than ARDS and TEN without any organ damage. We further analyzed the changes in cytokine/chemokine levels before and after treatment in two TEN patients with ARDS. CCL2, IL-6, and IL-8 levels decreased after systemic treatment compared to their baseline levels before treatment at an early stage. These results suggest that IL-8 and CCL2 may be involved in the pathogenesis of TEN-induced ARDS and have potential application as predictive markers for ARDS onset.
Collapse
Affiliation(s)
- Tomoya Watanabe
- Department of Environmental Immuno‐Dermatology Yokohama City University School of Medicine Yokohama Japan
| | - Yuko Watanabe
- Department of Environmental Immuno‐Dermatology Yokohama City University School of Medicine Yokohama Japan
| | - Noriko Ikeda
- Department of Environmental Immuno‐Dermatology Yokohama City University School of Medicine Yokohama Japan
| | - Michiko Aihara
- Department of Environmental Immuno‐Dermatology Yokohama City University School of Medicine Yokohama Japan
| | - Yukie Yamaguchi
- Department of Environmental Immuno‐Dermatology Yokohama City University School of Medicine Yokohama Japan
| |
Collapse
|
106
|
Gong H, Chen Y, Chen M, Li J, Zhang H, Yan S, Lv C. Advanced development and mechanism of sepsis-related acute respiratory distress syndrome. Front Med (Lausanne) 2022; 9:1043859. [PMID: 36452899 PMCID: PMC9701739 DOI: 10.3389/fmed.2022.1043859] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2023] Open
Abstract
The introduction of the Sepsis 3.0 guidelines in 2016 improved our understanding of sepsis diagnosis and therapy. Personalized treatment strategies and nursing methods for sepsis patients are recommended in the "Save Sepsis Campaign" in 2021. However, mortality in sepsis patients remains high. Patients with sepsis-related acute respiratory distress syndrome account for around 30% of them, with fatality rates ranging from 30 to 40%. Pathological specimens from individuals with sepsis-related ARDS frequently demonstrate widespread alveolar damage, and investigations have revealed that pulmonary epithelial and pulmonary endothelial injury is the underlying cause. As a result, the purpose of this work is to evaluate the mechanism and research progress of pulmonary epithelial and pulmonary endothelial damage in sepsis-related ARDS, which may provide new directions for future research, diagnosis, and therapy.
Collapse
Affiliation(s)
- Huankai Gong
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yao Chen
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meiling Chen
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Jiankang Li
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hong Zhang
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shijiao Yan
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- School of Public Health, Hainan Medical University, Haikou, China
| | - Chuanzhu Lv
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
107
|
Azapira N, Pourjafar S, Habibi A, Tayebi L, Keshtkar S, Kaviani M. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Promising Treatment for COVID-19 Pandemic. EXP CLIN TRANSPLANT 2022; 20:980-983. [PMID: 33622217 DOI: 10.6002/ect.2020.0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The pandemic of severe acute respiratory syndrome coronavirus-2 infection has prompted the urgent need for novel therapeutic approaches, especially for patients in critically severe conditions. To date, the pathogenesis of COVID-19 is not completely understood, and finding an effective new drug is still inconclusive. Mesenchymal stromal cell-derived extracellular vesicles contain large amounts of proteins, messenger RNA, and microRNAs that act as vehicles that transfer the cargo between cells. These nanotherapeutic materials exert anti-inflammatory effects on the immune system, which are necessary for subsidence of acute inflammation and promotion of tissue repair and regeneration. Therefore, the consideration of mesenchymal stromal cell-derived extracellular vesicles as a new, safe, and effective therapeutic approach in the treatment of COVID-19 pneumonia is suggested.
Collapse
Affiliation(s)
- Negar Azapira
- From the Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | |
Collapse
|
108
|
Bos LDJ, Ware LB. Acute respiratory distress syndrome: causes, pathophysiology, and phenotypes. Lancet 2022; 400:1145-1156. [PMID: 36070787 DOI: 10.1016/s0140-6736(22)01485-4] [Citation(s) in RCA: 301] [Impact Index Per Article: 100.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a common clinical syndrome of acute respiratory failure as a result of diffuse lung inflammation and oedema. ARDS can be precipitated by a variety of causes. The pathophysiology of ARDS is complex and involves the activation and dysregulation of multiple overlapping and interacting pathways of injury, inflammation, and coagulation, both in the lung and systemically. Mechanical ventilation can contribute to a cycle of lung injury and inflammation. Resolution of inflammation is a coordinated process that requires downregulation of proinflammatory pathways and upregulation of anti-inflammatory pathways. The heterogeneity of the clinical syndrome, along with its biology, physiology, and radiology, has increasingly been recognised and incorporated into identification of phenotypes. A precision-medicine approach that improves the identification of more homogeneous ARDS phenotypes should lead to an improved understanding of its pathophysiological mechanisms and how they differ from patient to patient.
Collapse
Affiliation(s)
- Lieuwe D J Bos
- Intensive Care, Amsterdam UMC-location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Lorraine B Ware
- Vanderbilt University School of Medicine, Medical Center North, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
109
|
Shawaf T, Schuberth HJ, Hussen J. Immune cell composition of the bronchoalveolar lavage fluid in healthy and respiratory diseased dromedary camels. BMC Vet Res 2022; 18:353. [PMID: 36131278 PMCID: PMC9490690 DOI: 10.1186/s12917-022-03446-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Background Respiratory diseases are among the most common and expensive to treat diseases in camels with a great economic impact on camel health, welfare, and production. Bronchoalveolar lavage fluid (BALF) has been proven as a valuable sample for investigating the leukocyte populations in the respiratory tract of several species. In the present study, fluorescent antibody labeling and flow cytometry were used to study the immune cell composition of BALF in dromedary camels. Animals with clinical respiratory diseases (n = seven) were compared with apparently healthy animals (n = 10). In addition, blood leukocytes from the same animals were stained in parallel with the same antibodies and analyzed by flow cytometry. Results Camel BALF macrophages, granulocytes, monocytes, and lymphocytes were identified based on their forward and side scatter properties. The expression pattern of the cell markers CD172a, CD14, CD163, and MHCII molecules on BALF cells indicates a similar phenotype for camel, bovine, and porcine BALF myeloid cells. The comparison between camels with respiratory disease and healthy camels regarding cellular composition in their BALF revealed a higher total cell count, a higher fraction of granulocytes, and a lower fraction of macrophages in diseased than healthy camels. Within the lymphocyte population, the percentages of helper T cells and B cells were also higher in diseased than healthy camels. The elevated expression of the activation marker CD11a on helper T cells of diseased camels is an indication of the expansion of helper T cells population due to infection and exposure to respiratory pathogens. The higher abundance of MHCII molecules on BALF macrophages from diseased camels indicates a polarization toward an inflammatory macrophage phenotype (M1) in respiratory diseased camels. No significant differences were observed in the systemic leukogram between healthy and diseased animals. Conclusions Collectively, the current study represents the first report on flow cytometric analysis of immune cell composition of bronchoalveolar lavage fluid (BALF) in dromedary camels.
Collapse
Affiliation(s)
- Turke Shawaf
- Department of Clinical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Hans-Joachim Schuberth
- Institute for Immunology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia.
| |
Collapse
|
110
|
Müller I, Alt P, Rajan S, Schaller L, Geiger F, Dietrich A. Transient Receptor Potential (TRP) Channels in Airway Toxicity and Disease: An Update. Cells 2022; 11:2907. [PMID: 36139480 PMCID: PMC9497104 DOI: 10.3390/cells11182907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Our respiratory system is exposed to toxicants and pathogens from both sides: the airways and the vasculature. While tracheal, bronchial and alveolar epithelial cells form a natural barrier in the airways, endothelial cells protect the lung from perfused toxic compounds, particulate matter and invading microorganism in the vascular system. Damages induce inflammation by our immune response and wound healing by (myo)fibroblast proliferation. Members of the transient receptor potential (TRP) superfamily of ion channel are expressed in many cells of the respiratory tract and serve multiple functions in physiology and pathophysiology. TRP expression patterns in non-neuronal cells with a focus on TRPA1, TRPC6, TRPM2, TRPM5, TRPM7, TRPV2, TRPV4 and TRPV6 channels are presented, and their roles in barrier function, immune regulation and phagocytosis are summarized. Moreover, TRP channels as future pharmacological targets in chronic obstructive pulmonary disease (COPD), asthma, cystic and pulmonary fibrosis as well as lung edema are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), LMU-Munich, Nussbaumstr. 26, 80336 Munich, Germany
| |
Collapse
|
111
|
Jumaniyazova E, Lokhonina A, Dzhalilova D, Kosyreva A, Fatkhudinov T. Immune Cells in Head-and-Neck Tumor Microenvironments. J Pers Med 2022; 12:jpm12091521. [PMID: 36143308 PMCID: PMC9506052 DOI: 10.3390/jpm12091521] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Head-and-neck cancers constitute a heterogeneous group of aggressive tumors with high incidence and low survival rates, collectively being the sixth most prevalent cancer type globally. About 90% of head-and-neck cancers are classified as squamous cell carcinomas (HNSCC). The innate and adaptive immune systems, indispensable for anti-cancer immune surveillance, largely define the rates of HNSCC emergence and progression. HNSCC microenvironments harbor multiple cell types that infiltrate the tumors and interact both with tumor cells and among themselves. Gradually, tumor cells learn to manipulate the immune system, either by adapting their own immunogenicity or through the release of immunosuppressive molecules. These interactions continuously evolve and shape the tumor microenvironment, both structurally and functionally, facilitating angiogenesis, proliferation and metastasis. Our understanding of this evolution is directly related to success in the development of advanced therapies. This review focuses on the key mechanisms that rule HNSCC infiltration, featuring particular immune cell types and their roles in the pathogenesis. A close focus on the tumor-immunity interactions will help identify new immunotherapeutic targets in patients with HNSCC.
Collapse
Affiliation(s)
- Enar Jumaniyazova
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Correspondence: ; Tel.: +7-9254258360
| | - Anastasiya Lokhonina
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Dzhuliia Dzhalilova
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Anna Kosyreva
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Timur Fatkhudinov
- Department of Histology, Cytology and Embryology, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
112
|
Kan Z, Zhao KX, Jiang C, Liu DY, Guo Y, Liu LY, Wang WJ, He ZQ, Zhang ZF, Wang SY. Respiratory exposure to graphene oxide induces pulmonary fibrosis and organ damages in rats involving caspase-1/p38MAPK/TGF-β1 signaling pathways. CHEMOSPHERE 2022; 303:135181. [PMID: 35667501 DOI: 10.1016/j.chemosphere.2022.135181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 05/23/2022] [Accepted: 05/28/2022] [Indexed: 06/15/2023]
Abstract
Numerous studies have shown that graphene oxide (GO) respiratory exposure led to severe lung injury, but whether pulmonary fibrosis caused by GO respiratory exposure is related to the activation of the caspase-1/p38MAPK/TGF-β1 remains unclear. In this study, rats were administrated GO by intratracheal instillation and fed for three months, and the molecular mechanisms of GO on the pulmonary fibrosis and other organ damage caused by GO respiratory exposure were examined. The results showed that the expression of caspase-1/p38MAPK/TGF-β1 pathway-related factors were significantly elevated with the increase of exposure concentrations of GO. Those data proved that the caspase-1/p38MAPK/TGF-β1 signaling pathway was involved in the pulmonary fibrosis caused by GO respiratory exposure. The trends of related factors also proved that the caspase-1/p38MAPK/TGF-β1 pathway was likely to play a dominant role in the sub-acute and sub-chronic stages. The other organ damage examination found that the liver and spleen were damaged initially by the GO respiratory exposure. Meanwhile for the testicle, although the acute injury was severe, signs of recovery were found during the three-month trial period.
Collapse
Affiliation(s)
- Ze Kan
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), Heilongjiang Institute of Labor Hygiene and Occupational Diseases/The Second Hospital of Heilongjiang Province, Harbin, 150028, PR China
| | - Ke-Xin Zhao
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment/School of Environment, Harbin Institute of Technology (HIT), Harbin, 150090, Heilongjiang, China
| | - Chao Jiang
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), Heilongjiang Institute of Labor Hygiene and Occupational Diseases/The Second Hospital of Heilongjiang Province, Harbin, 150028, PR China
| | - Da-Yang Liu
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), Heilongjiang Institute of Labor Hygiene and Occupational Diseases/The Second Hospital of Heilongjiang Province, Harbin, 150028, PR China
| | - Ying Guo
- Guangdong Key Laboratory of Environmental Pollution and Health, And School of Environment, Jinan University, Guangzhou, 510632, China
| | - Li-Yan Liu
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment/School of Environment, Harbin Institute of Technology (HIT), Harbin, 150090, Heilongjiang, China
| | - Wen-Juan Wang
- Heilongjiang Pony Testing Technical Co.,Ltd, Harbin, 150028, Heilongjiang, China
| | - Zhi-Qiang He
- Heilongjiang Pony Testing Technical Co.,Ltd, Harbin, 150028, Heilongjiang, China
| | - Zi-Feng Zhang
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment/School of Environment, Harbin Institute of Technology (HIT), Harbin, 150090, Heilongjiang, China.
| | - Su-Yi Wang
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), Heilongjiang Institute of Labor Hygiene and Occupational Diseases/The Second Hospital of Heilongjiang Province, Harbin, 150028, PR China
| |
Collapse
|
113
|
Su Y, Silva JD, Doherty D, Simpson DA, Weiss DJ, Rolandsson-Enes S, McAuley DF, O'Kane CM, Brazil DP, Krasnodembskaya AD. Mesenchymal stromal cells-derived extracellular vesicles reprogramme macrophages in ARDS models through the miR-181a-5p-PTEN-pSTAT5-SOCS1 axis. Thorax 2022; 78:617-630. [PMID: 35948417 DOI: 10.1136/thoraxjnl-2021-218194] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 06/04/2022] [Indexed: 11/04/2022]
Abstract
RATIONALE A better understanding of the mechanism of action of mesenchymal stromal cells (MSCs) and their extracellular vesicles (EVs) is needed to support their use as novel therapies for acute respiratory distress syndrome (ARDS). Macrophages are important mediators of ARDS inflammatory response. Suppressor of cytokine signalling (SOCS) proteins are key regulators of the macrophage phenotype switch. We therefore investigated whether SOCS proteins are involved in mediation of the MSC effect on human macrophage reprogramming. METHODS Human monocyte-derived macrophages (MDMs) were stimulated with lipopolysaccharide (LPS) or plasma samples from patients with ARDS (these samples were previously classified into hypo-inflammatory and hyper-inflammatory phenotype) and treated with MSC conditioned medium (CM) or EVs. Protein expression was measured by Western blot. EV micro RNA (miRNA) content was determined by miRNA sequencing. In vivo: LPS-injured C57BL/6 mice were given EVs isolated from MSCs in which miR-181a had been silenced by miRNA inhibitor or overexpressed using miRNA mimic. RESULTS EVs were the key component of MSC CM responsible for anti-inflammatory modulation of human macrophages. EVs significantly reduced secretion of tumour necrosis factor-α and interleukin-8 by LPS-stimulated or ARDS plasma-stimulated MDMs and this was dependent on SOCS1. Transfer of miR-181a in EVs downregulated phosphatase and tensin homolog (PTEN) and subsequently activated phosphorylated signal transducer and activator of transcription 5 (pSTAT5) leading to upregulation of SOCS1 in macrophages. In vivo, EVs alleviated lung injury and upregulated pSTAT5 and SOCS1 expression in alveolar macrophages in a miR181-dependent manner. Overexpression of miR-181a in MSCs significantly enhanced therapeutic efficacy of EVs in this model. CONCLUSION miR-181a-PTEN-pSTAT5-SOCS1 axis is a novel pathway responsible for immunomodulatory effect of MSC EVs in ARDS.
Collapse
Affiliation(s)
- Yue Su
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Johnatas Dutra Silva
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Declan Doherty
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - David A Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sara Rolandsson-Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Derek P Brazil
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
114
|
Heydarian M, Schulz C, Stoeger T, Hilgendorff A. Association of immune cell recruitment and BPD development. Mol Cell Pediatr 2022; 9:16. [PMID: 35917002 PMCID: PMC9346035 DOI: 10.1186/s40348-022-00148-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
In the neonatal lung, exposure to both prenatal and early postnatal risk factors converge into the development of injury and ultimately chronic disease, also known as bronchopulmonary dysplasia (BPD). The focus of many studies has been the characteristic inflammatory responses provoked by these exposures. Here, we review the relationship between immaturity and prenatal conditions, as well as postnatal exposure to mechanical ventilation and oxygen toxicity, with the imbalance of pro- and anti-inflammatory regulatory networks. In these conditions, cytokine release, protease activity, and sustained presence of innate immune cells in the lung result in pathologic processes contributing to lung injury. We highlight the recruitment and function of myeloid innate immune cells, in particular, neutrophils and monocyte/macrophages in the BPD lung in human patients and animal models. We also discuss dissimilarities between the infant and adult immune system as a basis for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christian Schulz
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Tobias Stoeger
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany. .,Center for Comprehensive Developmental Care (CDeCLMU) at the interdisciplinary Social Pediatric Center, (iSPZ), University Hospital Ludwig-Maximilian University, Munich, Germany.
| |
Collapse
|
115
|
Tao Q, Zhang ZD, Qin Z, Liu XW, Li SH, Bai LX, Ge WB, Li JY, Yang YJ. Aspirin eugenol ester alleviates lipopolysaccharide-induced acute lung injury in rats while stabilizing serum metabolites levels. Front Immunol 2022; 13:939106. [PMID: 35967416 PMCID: PMC9372404 DOI: 10.3389/fimmu.2022.939106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aspirin eugenol ester (AEE) was a novel drug compound with aspirin and eugenol esterified. AEE had various pharmacological activities, such as anti-inflammatory, antipyretic, analgesic, anti-oxidative stress and so on. In this study, it was aimed to investigate the effect of AEE on the acute lung injury (ALI) induced by lipopolysaccharide (LPS) in rats. In vitro experiments evaluated the protective effect of AEE on the LPS-induced A549 cells. The tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) were measured in the cell supernatant. The Wistar rats were randomly divided into five groups (n = 8): control group, model group (LPS group), LPS + AEE group (AEE, 54 mg·kg-1), LPS + AEE group (AEE, 108 mg·kg-1), LPS + AEE group (AEE, 216 mg·kg-1). The lung wet-to-dry weight (W/D) ratio and immune organ index were calculated. WBCs were counted in bronchoalveolar lavage fluid (BALF) and total protein concentration was measured. Hematoxylin-Eosin (HE) staining of lung tissue was performed. Glutathione (GSH), glutathione peroxidase (GPx), catalase (CAT), antioxidant superoxide dismutase (SOD), total antioxidant capacity (T-AOC), lactate dehydrogenase (LDH), C-reactive protein (CRP), myeloperoxidase (MPO), malondialdehyde (MDA), macrophage mobility inhibitory factor (MIF), TNF-α, IL-6, and IL-1β activity were measured. The metabolomic analysis of rat serum was performed by UPLC-QTOF-MS/MS. From the results, compared with LPS group, AEE improved histopathological changes, reduced MDA, CRP, MPO, MDA, and MIF production, decreased WBC count and total protein content in BALF, pro-inflammatory cytokine levels, immune organ index and lung wet-dry weight (W/D), increased antioxidant enzyme activity, in a dose-dependent manner. The results of serum metabolomic analysis showed that the LPS-induced ALI caused metabolic disorders and oxidative stress in rats, while AEE could ameliorate it to some extent. Therefore, AEE could alleviate LPS-induced ALI in rats by regulating abnormal inflammatory responses, slowing down oxidative stress, and modulating energy metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
116
|
Assessment of diagnostic utility of serum hemeoxygenase-1 measurement for acute exacerbation of interstitial pneumonias. Sci Rep 2022; 12:12935. [PMID: 35902685 PMCID: PMC9334264 DOI: 10.1038/s41598-022-17290-0] [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: 02/27/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
The present study aimed to evaluate whether serum heme oxygenase (HO)-1 could be a reliable blood biomarker for diagnosing acute exacerbations (AEs) of both idiopathic interstitial pneumonia (IIP) and secondary interstitial pneumonia (SIP). Serum HO-1 levels of newly diagnosed patients with IP were measured, and the relationships between serum HO-1 and other serum biomarkers and high-resolution CT scores, were evaluated. Blood samples were collected from 90 patients with IIP, including 32 having an AE, and 32 with SIP, including 9 having an AE. The patients having an AE had significantly higher HO-1 levels than those not having an AE (35.2 ng/mL vs. 16.4 ng/mL; p < 0.001). On receiver operating characteristics (ROC) curve analysis for serum HO-1 ability to detect an AE, the area under the ROC curve (AUC) was 0.87 in patients with IIPs and 0.86 in those with SIPs. Also, in patients with both IIPs and SIPs, the combination of the serum HO-1 level and the GGO score showed favorable AUCs (IIPs: 0.92, SIPs: 0.83), though HO-1-not-including model (combination of LDH and GGO) also showed acceptable AUCs. Serum HO-1 could be a clinically useful biomarker for the accurate diagnosis of patients with AEs.
Collapse
|
117
|
Oatis D, Simon-Repolski E, Balta C, Mihu A, Pieretti G, Alfano R, Peluso L, Trotta MC, D’Amico M, Hermenean A. Cellular and Molecular Mechanism of Pulmonary Fibrosis Post-COVID-19: Focus on Galectin-1, -3, -8, -9. Int J Mol Sci 2022; 23:8210. [PMID: 35897786 PMCID: PMC9332679 DOI: 10.3390/ijms23158210] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary fibrosis is a consequence of the pathological accumulation of extracellular matrix (ECM), which finally leads to lung scarring. Although the pulmonary fibrogenesis is almost known, the last two years of the COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its post effects added new particularities which need to be explored. Many questions remain about how pulmonary fibrotic changes occur within the lungs of COVID-19 patients, and whether the changes will persist long term or are capable of resolving. This review brings together existing knowledge on both COVID-19 and pulmonary fibrosis, starting with the main key players in promoting pulmonary fibrosis, such as alveolar and endothelial cells, fibroblasts, lipofibroblasts, and macrophages. Further, we provide an overview of the main molecular mechanisms driving the fibrotic process in connection with Galactin-1, -3, -8, and -9, together with the currently approved and newly proposed clinical therapeutic solutions given for the treatment of fibrosis, based on their inhibition. The work underlines the particular pathways and processes that may be implicated in pulmonary fibrosis pathogenesis post-SARS-CoV-2 viral infection. The recent data suggest that galectin-1, -3, -8, and -9 could become valuable biomarkers for the diagnosis and prognosis of lung fibrosis post-COVID-19 and promising molecular targets for the development of new and original therapeutic tools to treat the disease.
Collapse
Affiliation(s)
- Daniela Oatis
- Department of Infectious Disease, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Doctoral School of Biology, Vasile Goldis Western University of Arad, 310414 Arad, Romania
| | - Erika Simon-Repolski
- Doctoral School of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Department of Pneumology, Arad Clinical Emergency Hospital, 310031 Arad, Romania
| | - Cornel Balta
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania;
| | - Alin Mihu
- Department of Microbiology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Roberto Alfano
- Department of Advanced Medical and Surgical Sciences “DAMSS”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luisa Peluso
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Michele D’Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania;
- Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania
| |
Collapse
|
118
|
Liu C, Xiao K, Xie L. Advances in the Regulation of Macrophage Polarization by Mesenchymal Stem Cells and Implications for ALI/ARDS Treatment. Front Immunol 2022; 13:928134. [PMID: 35880175 PMCID: PMC9307903 DOI: 10.3389/fimmu.2022.928134] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common condition with high mortality. ALI/ARDS is caused by multiple etiologies, and the main clinical manifestations are progressive dyspnea and intractable hypoxemia. Currently, supportive therapy is the main ALI/ARDS treatment, and there remains a lack of targeted and effective therapeutic strategies. Macrophages are important components of innate immunity. M1 macrophages are pro-inflammatory, while M2 macrophages are anti-inflammatory and promote tissue repair. Mesenchymal stem cells (MSCs) are stem cells with broad application prospects in tissue regeneration due to their multi-directional differentiation potential along with their anti-inflammatory and paracrine properties. MSCs can regulate the balance of M1/M2 macrophage polarization to improve the prognosis of ALI/ARDS. In this paper, we review the mechanisms by which MSCs regulate macrophage polarization and the signaling pathways associated with polarization. This review is expected to provide new targets for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Kun Xiao
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| | - Lixin Xie
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Kun Xiao, ; Lixin Xie,
| |
Collapse
|
119
|
Qian Z, Wang Q, Qiu Z, Li D, Zhang C, Xiong X, Zheng Z, Ruan Q, Guo Y, Guo J. Protein nanoparticle-induced osmotic pressure gradients modify pulmonary edema through hyperpermeability in acute respiratory distress syndrome. J Nanobiotechnology 2022; 20:314. [PMID: 35794575 PMCID: PMC9257569 DOI: 10.1186/s12951-022-01519-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/21/2022] [Indexed: 01/14/2023] Open
Abstract
AbstractAcute respiratory distress syndrome (ARDS), caused by noncardiogenic pulmonary edema (PE), contributes significantly to Coronavirus 2019 (COVID-19)-associated morbidity and mortality. We explored the effect of transmembrane osmotic pressure (OP) gradients in PE using a fluorescence resonance energy transfer-based Intermediate filament (IF) tension optical probe. Angiotensin-II- and bradykinin-induced increases in intracellular protein nanoparticle (PN)-OP were associated with inflammasome production and cytoskeletal depolymerization. Intracellular protein nanoparticle production also resulted in cytomembrane hyperpolarization and L-VGCC-induced calcium signals, which differed from diacylglycerol-induced calcium increment via TRPC6 activation. Both pathways involve voltage-dependent cation influx and OP upregulation via SUR1-TRPM4 channels. Meanwhile, intra/extracellular PN-induced OP gradients across membranes upregulated pulmonary endothelial and alveolar barrier permeability. Attenuation of intracellular PN, calcium signals, and cation influx by drug combinations effectively relieved intracellular OP and pulmonary endothelial nonselective permeability, and improved epithelial fluid absorption and PE. Thus, PN-OP is pivotal in pulmonary edema in ARDS and COVID-19, and transmembrane OP recovery could be used to treat pulmonary edema and develop new drug targets in pulmonary injury.
Graphical Abstract
Collapse
|
120
|
Ogoshi T, Fukunaga M, Suzuki Y, Taura Y, Yatera K. Percutaneous Left Atrial Appendage Closure in a Patient with Diffuse Alveolar Hemorrhaging Associated with Anticoagulant Therapy and Atrial Fibrillation. Intern Med 2022; 61:2045-2050. [PMID: 34840230 PMCID: PMC9334249 DOI: 10.2169/internalmedicine.8551-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Diffuse alveolar hemorrhaging (DAH) due to oral anticoagulation (OAC) is a life-threatening condition that leads to severe respiratory failure. There is a clinical dilemma in that OAC-induced DAH often forces the discontinuation of OAC therapy and the administration of high-dose corticosteroids, which increases the risk of stroke and cardiovascular events. We herein report the first case of OAC-induced DAH and atrial fibrillation (AF) in a patient who completely discontinued OAC therapy and high-dose corticosteroids after experiencing percutaneous left atrial appendage (LAA) occlusion. This case suggests that percutaneous LAA closure may aid in the management of OAC-induced DAH and AF.
Collapse
Affiliation(s)
- Takaaki Ogoshi
- Department of Respiratory, Kokura Memorial Hospital, Japan
| | - Masato Fukunaga
- Department of Cardiovascular Medicine, Kokura Memorial Hospital, Japan
| | - Yu Suzuki
- Department of Respiratory, Kokura Memorial Hospital, Japan
| | - Yusuke Taura
- Department of Respiratory, Kokura Memorial Hospital, Japan
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
121
|
Ma S, Zhang J, Liu H, Li S, Wang Q. The Role of Tissue-Resident Macrophages in the Development and Treatment of Inflammatory Bowel Disease. Front Cell Dev Biol 2022; 10:896591. [PMID: 35721513 PMCID: PMC9199005 DOI: 10.3389/fcell.2022.896591] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn’s disease and ulcerative colitis, is a refractory disease with many immune abnormalities and pathologies in the gastrointestinal tract. Because macrophages can distinguish innocuous antigens from potential pathogens to maintain mucosa barrier functions, they are essential cells in the intestinal immune system. With numerous numbers in the intestinal tract, tissue-resident macrophages have a significant effect on the constant regeneration of intestinal epithelial cells and maintaining the immune homeostasis of the intestinal mucosa. They also have a significant influence on IBD through regulating pro-(M1) or anti-inflammatory (M2) phenotype polarization according to different environmental cues. The disequilibrium of the phenotypes and functions of macrophages, disturbed by intracellular or extracellular stimuli, influences the progression of disease. Further investigation of macrophages’ role in the progression of IBD will facilitate deciphering the pathogenesis of disease and exploring novel targets to develop novel medications. In this review, we shed light on the origin and maintenance of intestinal macrophages, as well as the role of macrophages in the occurrence and development of IBD. In addition, we summarize the interaction between gut microbiota and intestinal macrophages, and the role of the macrophage-derived exosome. Furthermore, we discuss the molecular and cellular mechanisms participating in the polarization and functions of gut macrophages, the potential targeted strategies, and current clinical trials for IBD.
Collapse
Affiliation(s)
- Shengjie Ma
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Chang Chun, China
| | - Jiaxin Zhang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Chang Chun, China
| | - Heshi Liu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Chang Chun, China
| | - Shuang Li
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Chang Chun, China
| | - Quan Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Chang Chun, China
| |
Collapse
|
122
|
Stevens J, Steinmeyer S, Bonfield M, Peterson L, Wang T, Gray J, Lewkowich I, Xu Y, Du Y, Guo M, Wynn JL, Zacharias W, Salomonis N, Miller L, Chougnet C, O’Connor DH, Deshmukh H. The balance between protective and pathogenic immune responses to pneumonia in the neonatal lung is enforced by gut microbiota. Sci Transl Med 2022; 14:eabl3981. [PMID: 35704600 PMCID: PMC10032669 DOI: 10.1126/scitranslmed.abl3981] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although modern clinical practices such as cesarean sections and perinatal antibiotics have improved infant survival, treatment with broad-spectrum antibiotics alters intestinal microbiota and causes dysbiosis. Infants exposed to perinatal antibiotics have an increased likelihood of life-threatening infections, including pneumonia. Here, we investigated how the gut microbiota sculpt pulmonary immune responses, promoting recovery and resolution of infection in newborn rhesus macaques. Early-life antibiotic exposure interrupted the maturation of intestinal commensal bacteria and disrupted the developmental trajectory of the pulmonary immune system, as assessed by single-cell proteomic and transcriptomic analyses. Early-life antibiotic exposure rendered newborn macaques more susceptible to bacterial pneumonia, concurrent with increases in neutrophil senescence and hyperinflammation, broad inflammatory cytokine signaling, and macrophage dysfunction. This pathogenic reprogramming of pulmonary immunity was further reflected by a hyperinflammatory signature in all pulmonary immune cell subsets coupled with a global loss of tissue-protective, homeostatic pathways in the lungs of dysbiotic newborns. Fecal microbiota transfer was associated with partial correction of the broad immune maladaptations and protection against severe pneumonia. These data demonstrate the importance of intestinal microbiota in programming pulmonary immunity and support the idea that gut microbiota promote the balance between pathways driving tissue repair and inflammatory responses associated with clinical recovery from infection in infants. Our results highlight a potential role for microbial transfer for immune support in these at-risk infants.
Collapse
Affiliation(s)
- Joseph Stevens
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shelby Steinmeyer
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Madeline Bonfield
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Laura Peterson
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Timothy Wang
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jerilyn Gray
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ian Lewkowich
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yan Xu
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yina Du
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Minzhe Guo
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - James L. Wynn
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - William Zacharias
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa Miller
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
- California National Primate Research Center, Davis, CA 95616, USA
| | - Claire Chougnet
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Dennis Hartigan O’Connor
- California National Primate Research Center, Davis, CA 95616, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Hitesh Deshmukh
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Corresponding author.
| |
Collapse
|
123
|
Diao Y, Ding Q, Xu G, Li Y, Li Z, Zhu H, Zhu W, Wang P, Shi Y. Qingfei Litan Decoction Against Acute Lung Injury/Acute Respiratory Distress Syndrome: The Potential Roles of Anti-Inflammatory and Anti-Oxidative Effects. Front Pharmacol 2022; 13:857502. [PMID: 35677439 PMCID: PMC9168533 DOI: 10.3389/fphar.2022.857502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is an acute respiratory failure syndrome characterized by progressive arterial hypoxemia and dyspnea. Qingfei Litan (QFLT) decoction, as a classic prescription for the treatment of acute respiratory infections, is effective for the treatment of ALI/ARDS. In this study, the compounds, hub targets, and major pathways of QFLT in ALI/ARDS treatment were analyzed using Ultra high performance liquid chromatography coupled with mass spectrometry (UHPLC-MS) and systemic pharmacology strategies. UHPLC-MS identified 47 main components of QFLT. To explore its anti-inflammatory and anti-oxidative mechanisms, gene ontology (Go) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and network pharmacological analysis were conducted based on the main 47 components. KEGG enrichment analysis showed that TNF signaling pathway and Toll-like receptor signaling pathway may be the key pathways of ALI/ARDS. We explored the anti-inflammatory and anti-oxidative pharmacological effects of QFLT in treatment of ALI/ARDS in vivo and in vitro. QFLT suppressed the levels of proinflammatory cytokines and alleviated oxidative stress in LPS-challenged mice. In vitro, QFLT decreased the levels of TNF-α, IL-6, IL-1β secreted by LPS-activated macrophages, increased GSH level and decreased the LPS-activated reactive oxygen species (ROS) in lung epithelial A549 cells. This study suggested that QFLT may have anti-inflammatory and anti-oxidative effects on ALI/ARDS, combining in vivo and in vitro experiments with systemic pharmacology, providing a potential therapeutic strategy option.
Collapse
Affiliation(s)
- Yirui Diao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Gonghao Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yadong Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenqiu Li
- Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanping Zhu
- Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenxiang Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Peng Wang
- Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuanyuan Shi
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.,School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
124
|
Zou F, Zhuang ZB, Zou SS, Wang B, Zhang ZH. BML-111 alleviates inflammatory response of alveolar epithelial cells via miR-494/Slit2/Robo4 signalling axis to improve acute lung injury. Autoimmunity 2022; 55:318-327. [PMID: 35656971 DOI: 10.1080/08916934.2022.2065671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Fang Zou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Zhong-Bao Zhuang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Shuang-Shuang Zou
- Guangzhou Liwan Stomatological Hospital, Guangzhou, Guangdong Province, P.R. China
| | - Bu Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Zhi-Hua Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| |
Collapse
|
125
|
Wu X, Yao J, Hu Q, Kang H, Miao Y, Zhu L, Li C, Zhao X, Li J, Wan M, Tang W. Emodin Ameliorates Acute Pancreatitis-Associated Lung Injury Through Inhibiting the Alveolar Macrophages Pyroptosis. Front Pharmacol 2022; 13:873053. [PMID: 35721108 PMCID: PMC9201345 DOI: 10.3389/fphar.2022.873053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
Objective: To investigate the protective effect of emodin in acute pancreatitis (AP)-associated lung injury and the underlying mechanisms. Methods: NaT-AP model in rats was constructed using 3.5% sodium taurocholate, and CER+LPS-AP model in mice was constructed using caerulein combined with Lipopolysaccharide. Animals were divided randomly into four groups: sham, AP, Ac-YVAD-CMK (caspase-1 specific inhibitor, AYC), and emodin groups. AP-associated lung injury was assessed with H&E staining, inflammatory cytokine levels, and myeloperoxidase activity. Alveolar macrophages (AMs) pyroptosis was evaluated by flow cytometry. In bronchoalveolar lavage fluid, the levels of lactate dehydrogenase and inflammatory cytokines were measured by enzyme-linked immunosorbent assay. Pyroptosis-related protein expressions were detected by Western Blot. Results: Emodin, similar to the positive control AYC, significantly alleviated pancreas and lung damage in rats and mice. Additionally, emodin mitigated the pyroptotic process of AMs by decreasing the level of inflammatory cytokines and lactate dehydrogenase. More importantly, the protein expressions of NLRP3, ASC, Caspase1 p10, GSDMD, and GSDMD-NT in AMs were significantly downregulated after emodin intervention. Conclusion: Emodin has a therapeutic effect on AP-associated lung injury, which may result from the inhibition of NLRP3/Caspase1/GSDMD-mediated AMs pyroptosis signaling pathways.
Collapse
Affiliation(s)
- Xiajia Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongxin Kang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yifan Miao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lv Zhu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Cong Li
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, China
| | - Xianlin Zhao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Li
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
126
|
Mirchandani AS, Jenkins SJ, Bain CC, Sanchez-Garcia MA, Lawson H, Coelho P, Murphy F, Griffith DM, Zhang A, Morrison T, Ly T, Arienti S, Sadiku P, Watts ER, Dickinson RS, Reyes L, Cooper G, Clark S, Lewis D, Kelly V, Spanos C, Musgrave KM, Delaney L, Harper I, Scott J, Parkinson NJ, Rostron AJ, Baillie JK, Clohisey S, Pridans C, Campana L, Lewis PS, Simpson AJ, Dockrell DH, Schwarze J, Hirani N, Ratcliffe PJ, Pugh CW, Kranc K, Forbes SJ, Whyte MKB, Walmsley SR. Hypoxia shapes the immune landscape in lung injury and promotes the persistence of inflammation. Nat Immunol 2022; 23:927-939. [PMID: 35624205 PMCID: PMC9174051 DOI: 10.1038/s41590-022-01216-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/18/2022] [Indexed: 12/30/2022]
Abstract
Hypoxemia is a defining feature of acute respiratory distress syndrome (ARDS), an often-fatal complication of pulmonary or systemic inflammation, yet the resulting tissue hypoxia, and its impact on immune responses, is often neglected. In the present study, we have shown that ARDS patients were hypoxemic and monocytopenic within the first 48 h of ventilation. Monocytopenia was also observed in mouse models of hypoxic acute lung injury, in which hypoxemia drove the suppression of type I interferon signaling in the bone marrow. This impaired monopoiesis resulted in reduced accumulation of monocyte-derived macrophages and enhanced neutrophil-mediated inflammation in the lung. Administration of colony-stimulating factor 1 in mice with hypoxic lung injury rescued the monocytopenia, altered the phenotype of circulating monocytes, increased monocyte-derived macrophages in the lung and limited injury. Thus, tissue hypoxia altered the dynamics of the immune response to the detriment of the host and interventions to address the aberrant response offer new therapeutic strategies for ARDS.
Collapse
Affiliation(s)
- Ananda S Mirchandani
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| | - Stephen J Jenkins
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Calum C Bain
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Manuel A Sanchez-Garcia
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Hannah Lawson
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Patricia Coelho
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona Murphy
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David M Griffith
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ailiang Zhang
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tyler Morrison
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tony Ly
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Simone Arienti
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Pranvera Sadiku
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Emily R Watts
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rebecca S Dickinson
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Leila Reyes
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - George Cooper
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sarah Clark
- Intensive Care Unit, Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - David Lewis
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Van Kelly
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Christos Spanos
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Kathryn M Musgrave
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Respiratory Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Liam Delaney
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Isla Harper
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jonathan Scott
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Anthony J Rostron
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - J Kenneth Baillie
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Sara Clohisey
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Clare Pridans
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lara Campana
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | - A John Simpson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - David H Dockrell
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jürgen Schwarze
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nikhil Hirani
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Peter J Ratcliffe
- Nuffield Department of Medicine Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Francis Crick Institute, London, UK
| | - Christopher W Pugh
- Nuffield Department of Medicine Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kamil Kranc
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Stuart J Forbes
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Moira K B Whyte
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sarah R Walmsley
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
127
|
Saraiva AL, Justino AB, Franco RR, Silva HCG, Arruda FDS, Klein SG, Celes MRN, Goulart LR, Espindola FS. Polyphenols-Rich Fraction from Annona muricata Linn. Leaves Attenuates Oxidative and Inflammatory Responses in Neutrophils, Macrophages, and Experimental Lung Injury. Pharmaceutics 2022; 14:pharmaceutics14061182. [PMID: 35745755 PMCID: PMC9228609 DOI: 10.3390/pharmaceutics14061182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 02/05/2023] Open
Abstract
Annona muricata Linn. is a common plant found in the warmest regions of South and Central America and its use in traditional medicine has been reported for the treatment of various illnesses. In the current study, we investigate the antioxidant and anti-inflammatory activities of crude extract and fractions from A. muricata L. leaves in isolated murine phagocytic immune cells as well as experimental LPS-induced acute lung injury (ALI). In a luminol-dependent chemiluminescence assay, we showed that ethyl acetate (EtOAc.f) and n-butanol (BuOH.f) fractions—both rich in polyphenols—reduced the generation of reactive oxygen species (ROS) by neutrophils stimulated with opsonized zymosan; similar results were found in culture of bone marrow-derived macrophages (BMDMs). By evaluating anti-inflammatory activity in BMDMs, EtOAc.f and BuOH.f reduced secretion of IL-6 and expression of the co-stimulatory molecule CD40. Furthermore, in LPS-induced ALI, oral administration of EtOAc.f reduced myeloperoxidase (MPO) activity in lung tissue. In addition, on a mechanism dependent on glutathione levels, the oxidative damage was also attenuated. These findings revealed direct antioxidant and anti-inflammatory activities of polyphenols-rich fractions of A. muricata L. leaves on neutrophils and macrophages. Moreover, the reduced oxidative damage and levels of inflammatory markers in experimental ALI suggest that these fractions might be explored for the development of new therapies for inflammatory conditions.
Collapse
Affiliation(s)
- André Lopes Saraiva
- Institute of Biotechnology, Federal University of Uberlândia, Rua Acre s/n, Bloco 2E, Uberlândia 38400-902, MG, Brazil; (A.L.S.); (A.B.J.); (R.R.F.); (H.C.G.S.)
| | - Allisson Benatti Justino
- Institute of Biotechnology, Federal University of Uberlândia, Rua Acre s/n, Bloco 2E, Uberlândia 38400-902, MG, Brazil; (A.L.S.); (A.B.J.); (R.R.F.); (H.C.G.S.)
| | - Rodrigo Rodrigues Franco
- Institute of Biotechnology, Federal University of Uberlândia, Rua Acre s/n, Bloco 2E, Uberlândia 38400-902, MG, Brazil; (A.L.S.); (A.B.J.); (R.R.F.); (H.C.G.S.)
| | - Heitor Cappato Guerra Silva
- Institute of Biotechnology, Federal University of Uberlândia, Rua Acre s/n, Bloco 2E, Uberlândia 38400-902, MG, Brazil; (A.L.S.); (A.B.J.); (R.R.F.); (H.C.G.S.)
| | - Felipe dos Santos Arruda
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235, Setor Leste Universitário, Goiânia 74605-050, GO, Brazil; (F.d.S.A.); (M.R.N.C.)
| | - Sandra Gabriela Klein
- Rodent Vivarium Network (REBIR), Dean of Research and Graduate Studies, Federal University of Uberlândia, Rua Ceará s/n, Bloco 4U, Uberlândia 38405-315, MG, Brazil;
| | - Mara Rúbia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235, Setor Leste Universitário, Goiânia 74605-050, GO, Brazil; (F.d.S.A.); (M.R.N.C.)
| | | | - Foued Salmen Espindola
- Institute of Biotechnology, Federal University of Uberlândia, Rua Acre s/n, Bloco 2E, Uberlândia 38400-902, MG, Brazil; (A.L.S.); (A.B.J.); (R.R.F.); (H.C.G.S.)
- Correspondence: ; Tel.: +55-34-3225-8439
| |
Collapse
|
128
|
Pathogenesis of pneumonia and acute lung injury. Clin Sci (Lond) 2022; 136:747-769. [PMID: 35621124 DOI: 10.1042/cs20210879] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Pneumonia and its sequelae, acute lung injury, present unique challenges for pulmonary and critical care healthcare professionals, and these challenges have recently garnered global attention due to the ongoing Sars-CoV-2 pandemic. One limitation to translational investigation of acute lung injury, including its most severe manifestation (acute respiratory distress syndrome, ARDS) has been heterogeneity resulting from the clinical and physiologic diagnosis that represents a wide variety of etiologies. Recent efforts have improved our understanding and approach to heterogeneity by defining sub-phenotypes of ARDS although significant gaps in knowledge remain. Improving our mechanistic understanding of acute lung injury and its most common cause, infectious pneumonia, can advance our approach to precision targeted clinical interventions. Here, we review the pathogenesis of pneumonia and acute lung injury, including how respiratory infections and lung injury disrupt lung homoeostasis, and provide an overview of respiratory microbial pathogenesis, the lung microbiome, and interventions that have been demonstrated to improve outcomes-or not-in human clinical trials.
Collapse
|
129
|
Chen L, Ma Q, Zhang G, Lei Y, Wang W, Zhang Y, Li T, Zhong W, Ming Y, Song G. Protective effect and mechanism of loganin and morroniside on acute lung injury and pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154030. [PMID: 35279615 DOI: 10.1016/j.phymed.2022.154030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Loganin and morroniside are two iridoid glycosides with anti-inflammatory, antioxidant and anti-tumor effects. Whether they have effect on acute lung injury and pulmonary fibrosis are still unknown. PURPOSE To explore the potential effects of loganin and morroniside against acute lung cancer and pulmonary fibrosis, and the underlying molecular mechanism. STUDY DESIGN AND METHODS Cell and animal models of acute lung injury were established by the induction of LPS. After intervention with loganin and morroniside, the pathological symptom of lung tissue was assessed, pro-inflammatory factors in cells and lung tissues were detected, NF- κB/STAT3 signaling pathway related proteins were detected by western blotting. Mice pulmonary fibrosis model was induced by bleomycin, pathological symptom was assessed by HE and Masson staining. Fibrosis related indicators were detected by qPCR or western blot. CD4+/CD8+ was detected by flow cytometry. RESULTS Loganin and morroniside relieved the pathological symptom of lung tissue in acute lung injury, pro-inflammatory factors such as IL-6, IL-1β, TNF-α mRNA were inhibited. Expression of p-p65 and STAT3 in lung tissues were also downregulated. In addition, loganin and morroniside downregulated the expression of collagen fiber, hydroxyproline and TGF-β1, collagen I and α-SMA mRNA in lung tissues of pulmonary fibrosis model. This study proved that loganin and morroniside have protective effect on acute lung injury and pulmonary fibrosis, and may provide theoretical basis for the development of new clinical drugs.
Collapse
Affiliation(s)
- Lianghua Chen
- Key Laboratory of Fujian Province for physiology and Biochemistry of Subtropical Plant, Fujian Institute of Subtropical Botany, Xiamen, Fujian 361006, China
| | - Qiujuan Ma
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Gongye Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yongbin Lei
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weiwei Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yuqi Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Tingting Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Wei Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yanlin Ming
- Key Laboratory of Fujian Province for physiology and Biochemistry of Subtropical Plant, Fujian Institute of Subtropical Botany, Xiamen, Fujian 361006, China
| | - Gang Song
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
130
|
Elewa YHA, Masum MA, Mohamed SKA, Islam MR, Nakamura T, Ichii O, Kon Y. The Ameliorative Effect of Dexamethasone on the Development of Autoimmune Lung Injury and Mediastinal Fat-Associated Lymphoid Clusters in an Autoimmune Disease Mouse Model. Int J Mol Sci 2022; 23:ijms23084449. [PMID: 35457267 PMCID: PMC9027674 DOI: 10.3390/ijms23084449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
In our previous study, we revealed the ameliorative therapeutic effect of dexamethasone (Dex) for Lupus nephritis lesions in the MRL/MpJ-Fas lpr/lpr (Lpr) mouse model. The female Lpr mice developed a greater number of mediastinal fat-associated lymphoid clusters (MFALCs) and inflammatory lung lesions compared to the male mice. However, the effect of Dex, an immunosuppressive drug, on both lung lesions and the development of MFALCs in Lpr mice has not been identified yet. Therefore, in this study, we compared the development of lung lesions and MFALCs in female Lpr mice that received either saline (saline group “SG”) or dexamethasone (dexamethasone group “DG”) in drinking water as a daily dose along with weekly intraperitoneal injections for 10 weeks. Compared to the SG group, the DG group showed a significant reduction in the levels of serum anti-dsDNA antibodies, the size of MFALCs, the degree of lung injury, the area of high endothelial venules (HEVs), and the number of proliferating and immune cells in both MFALCs and the lungs. A significant positive correlation was observed between the size of MFALCs and the cellular aggregation in the lungs of Lpr mice. Therefore, this study confirmed the ameliorative effect of Dex on the development of lung injury and MFALCs via their regressive effect on both immune cells’ proliferative activity and the development of HEVs. Furthermore, the reprogramming of MFALCs by targeting immune cells and HEVs may provide a therapeutic strategy for autoimmune-disease-associated lung injury.
Collapse
Affiliation(s)
- Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Correspondence: ; Tel.: +81-11-706-5188
| | - Md Abdul Masum
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
| | - Sherif Kh. A. Mohamed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt;
| | - Md Rashedul Islam
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
| | - Teppei Nakamura
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
- Department of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Hokkaido 066-0052, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Hokkaido 060-0818, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; (M.A.M.); (M.R.I.); (T.N.); (O.I.); (Y.K.)
| |
Collapse
|
131
|
The Modulation of Interferon Regulatory Factor-1 via Caspase-1-Mediated Alveolar Macrophage Pyroptosis in Ventilator-Induced Lung Injury. Mediators Inflamm 2022; 2022:1002582. [PMID: 35462787 PMCID: PMC9033353 DOI: 10.1155/2022/1002582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/26/2022] [Accepted: 03/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background To examine the role of interferon regulatory factor-1 (IRF-1) and to explore the potential molecular mechanism in ventilator-induced lung injury. Methods Wild-type C57BL/6 mice and IRF-1 gene knockout mice/caspase-1 knockout mice were mechanically ventilated with a high tidal volume to establish a ventilator-related lung injury model. The supernatant of the alveolar lavage solution and the lung tissues of these mice were collected. The degree of lung injury was examined by hematoxylin and eosin staining. The protein and mRNA expression levels of IRF-1, caspase-1 (p10), and interleukin (IL)-1β (p17) in lung tissues were measured by western blot and quantitative real-time polymerase chain reaction, respectively. Pyroptosis of alveolar macrophages was detected by flow cytometry and western blotting for active caspase-1 and cleaved GSDMD. An enzyme-linked immunosorbent assay was used to measure the levels of IL-1β, IL-18, IL-6, TNF-α, and high mobility group box protein 1 (HMGB-1) in alveolar lavage fluid. Results IRF-1 expression and caspase-1-dependent pyroptosis in lung tissues of wild-type mice were significantly upregulated after mechanical ventilation with a high tidal volume. The degree of ventilator-related lung injury in IRF-1 gene knockout mice and caspase-1 knockout mice was significantly improved compared to that in wild-type mice, and the levels of GSDMD, IL-1β, IL-18, IL-6, and HMGB-1 in alveolar lavage solution were significantly reduced (P < 0.05). The expression levels of caspase-1 (p10), cleaved GSDMD, and IL-1β (p17) proteins in lung tissues of IRF-1 knockout mice with ventilator-related lung injury were significantly lower than those of wild-type mice, and the level of pyroptosis of macrophages in alveolar lavage solution was significantly reduced. Conclusions IRF-1 may aggravate ventilator-induced lung injury by regulating the activation of caspase-1 and the focal death of alveolar macrophages.
Collapse
|
132
|
Parny M, Coste A, Aubouy A, Rahabi M, Prat M, Pipy B, Treilhou M. Differential immunomodulatory effects of six pesticides of different chemical classes on human monocyte-derived macrophage functions. Food Chem Toxicol 2022; 163:112992. [PMID: 35395341 DOI: 10.1016/j.fct.2022.112992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/04/2022] [Accepted: 04/03/2022] [Indexed: 01/19/2023]
Abstract
Exposure to pesticides through eyes, skin, ingestion and inhalation may affects human health by interfering with immune cells, such as macrophages. We evaluated, in vitro, the effect of six pesticides widely used in apple arboriculture on the functions of human monocyte-derived macrophages (hMDMs). hMDMs were cultured for 4 or 24 h with or without pesticides (0.01, 0.1, 1, 10 μmol.L-1). We showed that chlorpyrifos, thiacloprid, thiophanate, boscalid, and captan had little toxic effect at the tested concentrations, while dithianon had low-cytotoxicity at 10 μmol.L-1. While boscalid showed no effect on hMDMs function, thiophanate (0.01 μmol.L-1) stimulated with TPA and thiacloprid (1, 10 μmol.L-1) stimulated with zymosan activated ROS production. Chlorpyrifos, dithianon, and captan inhibited ROS production and TNF-α, IL-1β pro-inflammatory cytokines. We established that dithianon (0.01-1 μmol.L-1) and captan (0.1, 1 μmol.L-1) induced mRNA expression of NQO1 and HMOX1 antioxidant enzymes. Dithianon also induced the mRNA expression of catalase, superoxide dismutase-2 at 10 μmol.L-1. Together, these results show that exposure to chlorpyrifos, dithianon, and captan induce immunomodulatory effects that may influence the disease fighting properties of monocytes/macrophages while pesticides such as thiacloprid, thiophanate and boscalid have little influence.
Collapse
Affiliation(s)
- Melissa Parny
- EA7417, BTSB, Université Fédérale Toulouse Midi-Pyrénées, INU Champollion, Albi, France; PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Agnès Coste
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Agnès Aubouy
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Mouna Rahabi
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Melissa Prat
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Bernard Pipy
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Michel Treilhou
- EA7417, BTSB, Université Fédérale Toulouse Midi-Pyrénées, INU Champollion, Albi, France.
| |
Collapse
|
133
|
Trappetti V, Fazzari J, Fernandez-Palomo C, Smyth L, Potez M, Shintani N, de Breuyn Dietler B, Martin OA, Djonov V. Targeted Accumulation of Macrophages Induced by Microbeam Irradiation in a Tissue-Dependent Manner. Biomedicines 2022; 10:735. [PMID: 35453485 PMCID: PMC9025837 DOI: 10.3390/biomedicines10040735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation therapy (RT) is a vital component of multimodal cancer treatment, and its immunomodulatory effects are a major focus of current therapeutic strategies. Macrophages are some of the first cells recruited to sites of radiation-induced injury where they can aid in tissue repair, propagate radiation-induced fibrogenesis and influence tumour dynamics. Microbeam radiation therapy (MRT) is a unique, spatially fractionated radiation modality that has demonstrated exceptional tumour control and reduction in normal tissue toxicity, including fibrosis. We conducted a morphological analysis of MRT-irradiated normal liver, lung and skin tissues as well as lung and melanoma tumours. MRT induced distinct patterns of DNA damage, reflecting the geometry of the microbeam array. Macrophages infiltrated these regions of peak dose deposition at variable timepoints post-irradiation depending on the tissue type. In normal liver and lung tissue, macrophages clearly demarcated the beam path by 48 h and 7 days post-irradiation, respectively. This was not reflected, however, in normal skin tissue, despite clear DNA damage marking the beam path. Persistent DNA damage was observed in MRT-irradiated lung carcinoma, with an accompanying geometry-specific influx of mixed M1/M2-like macrophage populations. These data indicate the unique potential of MRT as a tool to induce a remarkable accumulation of macrophages in an organ/tissue-specific manner. Further characterization of these macrophage populations is warranted to identify their organ-specific roles in normal tissue sparing and anti-tumour responses.
Collapse
Affiliation(s)
- Verdiana Trappetti
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| | - Jennifer Fazzari
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| | - Cristian Fernandez-Palomo
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| | - Lloyd Smyth
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, University of Melbourne, Melbourne, VIC 3052, Australia;
| | - Marine Potez
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Nahoko Shintani
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| | - Bettina de Breuyn Dietler
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| | - Olga A. Martin
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, 305 Grattan St., Melbourne, VIC 3000, Australia
- Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstarsse 2, 3012 Bern, Switzerland; (V.T.); (J.F.); (C.F.-P.); (M.P.); (N.S.); (B.d.B.D.); (O.A.M.)
| |
Collapse
|
134
|
Wang L, Zhao M. Suppression of NOD-like receptor protein 3 inflammasome activation and macrophage M1 polarization by hederagenin contributes to attenuation of sepsis-induced acute lung injury in rats. Bioengineered 2022; 13:7262-7276. [PMID: 35266443 PMCID: PMC9208453 DOI: 10.1080/21655979.2022.2047406] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acute lung injury (ALI) is a major leading cause of death in sepsis patients. Hederagenin (HG), derived from Hedera helix Linné, has anti-inflammatory effects, while its role in sepsis-induced ALI has not been elucidated. In vivo, rats were subjected to cecal ligation and puncture to induce ALI and then treated with HG (12.5, 25, or 50 mg/kg) by gavage. Administration of HG raised survival rate, ameliorated lung injury, and decreased lung wet/dry ratio and inflammatory cell accumulation in bronchoalveloar lavage fluid (BALF) of ALI rats. HG inhibited macrophage polarization toward the M1 phenotype as evidenced by decreased CD86 expression in rat lung tissues. Moreover, HG decreased the secretion of TNF-α, IL-6 and monocyte chemoattractant protein-1 (MCP-1) in BALF and the levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in lung tissues. In vitro, phorbol-12-myristate-13-acetate (PMA)-differentiated THP-1 macrophages were stimulated with 100 ng/mL lipopolysaccharide. HG treatment inhibited M1 macrophage polarization and the production of M1-related pro-inflammatory mediators (IL-6, MCP-1, iNOS, and COX-2). Mechanistically, HG inhibited NLRP3 inflammasome activation and subsequent release of IL-18 and IL-1β, and suppressed NF-κB signaling pathway both in vivo and in vitro. Notably, HG treatment further emphasized the inhibitory effect of NF-κB inhibitor BAY11-7082 on NLRP3 inflammasome activation and macrophage M1 polarization. Taken together, HG exerts a protective effect against sepsis-induced ALI by reducing the inflammatory response and macrophage M1 polarization, which may involve NF-κB pathway-modulated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Min Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
135
|
Mirata S, Almonti V, Di Giuseppe D, Fornasini L, Raneri S, Vernazza S, Bersani D, Gualtieri AF, Bassi AM, Scarfì S. The Acute Toxicity of Mineral Fibres: A Systematic In Vitro Study Using Different THP-1 Macrophage Phenotypes. Int J Mol Sci 2022; 23:2840. [PMID: 35269982 PMCID: PMC8911508 DOI: 10.3390/ijms23052840] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Alveolar macrophages are the first line of defence against detrimental inhaled stimuli. To date, no comparative data have been obtained on the inflammatory response induced by different carcinogenic mineral fibres in the three main macrophage phenotypes: M0 (non-activated), M1 (pro-inflammatory) and M2 (alternatively activated). To gain new insights into the different toxicity mechanisms of carcinogenic mineral fibres, the acute effects of fibrous erionite, crocidolite and chrysotile in the three phenotypes obtained by THP-1 monocyte differentiation were investigated. The three mineral fibres apparently act by different toxicity mechanisms. Crocidolite seems to exert its toxic effects mostly as a result of its biodurability, ROS and cytokine production and DNA damage. Chrysotile, due to its low biodurability, displays toxic effects related to the release of toxic metals and the production of ROS and cytokines. Other mechanisms are involved in explaining the toxicity of biodurable fibrous erionite, which induces lower ROS and toxic metal release but exhibits a cation-exchange capacity able to alter the intracellular homeostasis of important cations. Concerning the differences among the three macrophage phenotypes, similar behaviour in the production of pro-inflammatory mediators was observed. The M2 phenotype, although known as a cell type recruited to mitigate the inflammatory state, in the case of asbestos fibres and erionite, serves to support the process by supplying pro-inflammatory mediators.
Collapse
Affiliation(s)
- Serena Mirata
- Department Earth, Environment and Life Sciences, University of Genova, 16132 Genova, Italy;
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy; (V.A.); (S.V.); (A.M.B.)
| | - Vanessa Almonti
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy; (V.A.); (S.V.); (A.M.B.)
- Department Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Dario Di Giuseppe
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy; (D.D.G.); (A.F.G.)
| | - Laura Fornasini
- ICCOM-CNR—Institute of Chemistry of OrganoMetallic Compounds, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (L.F.); (S.R.)
| | - Simona Raneri
- ICCOM-CNR—Institute of Chemistry of OrganoMetallic Compounds, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (L.F.); (S.R.)
| | - Stefania Vernazza
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy; (V.A.); (S.V.); (A.M.B.)
- Department Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Danilo Bersani
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parco Area delle Scienze 7/A, 43124 Parma, Italy;
| | - Alessandro F. Gualtieri
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy; (D.D.G.); (A.F.G.)
| | - Anna Maria Bassi
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy; (V.A.); (S.V.); (A.M.B.)
- Department Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Sonia Scarfì
- Department Earth, Environment and Life Sciences, University of Genova, 16132 Genova, Italy;
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy; (V.A.); (S.V.); (A.M.B.)
| |
Collapse
|
136
|
Corry J, Kettenburg G, Upadhyay AA, Wallace M, Marti MM, Wonderlich ER, Bissel SJ, Goss K, Sturgeon TJ, Watkins SC, Reed DS, Bosinger SE, Barratt-Boyes SM. Infiltration of inflammatory macrophages and neutrophils and widespread pyroptosis in lung drive influenza lethality in nonhuman primates. PLoS Pathog 2022; 18:e1010395. [PMID: 35271686 PMCID: PMC8939778 DOI: 10.1371/journal.ppat.1010395] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 03/22/2022] [Accepted: 02/24/2022] [Indexed: 01/04/2023] Open
Abstract
Severe influenza kills tens of thousands of individuals each year, yet the mechanisms driving lethality in humans are poorly understood. Here we used a unique translational model of lethal H5N1 influenza in cynomolgus macaques that utilizes inhalation of small-particle virus aerosols to define mechanisms driving lethal disease. RNA sequencing of lung tissue revealed an intense interferon response within two days of infection that resulted in widespread expression of interferon-stimulated genes, including inflammatory cytokines and chemokines. Macaques with lethal disease had rapid and profound loss of alveolar macrophages (AMs) and infiltration of activated CCR2+ CX3CR1+ interstitial macrophages (IMs) and neutrophils into lungs. Parallel changes of AMs and neutrophils in bronchoalveolar lavage (BAL) correlated with virus load when compared to macaques with mild influenza. Both AMs and IMs in lethal influenza were M1-type inflammatory macrophages which expressed neutrophil chemotactic factors, while neutrophils expressed genes associated with activation and generation of neutrophil extracellular traps (NETs). NETs were prominent in lung and were found in alveolar spaces as well as lung parenchyma. Genes associated with pyroptosis but not apoptosis were increased in lung, and activated inflammatory caspases, IL-1β and cleaved gasdermin D (GSDMD) were present in bronchoalveolar lavage fluid and lung homogenates. Cleaved GSDMD was expressed by lung macrophages and alveolar epithelial cells which were present in large numbers in alveolar spaces, consistent with loss of epithelial integrity. Cleaved GSDMD colocalized with viral NP-expressing cells in alveoli, reflecting pyroptosis of infected cells. These novel findings reveal that a potent interferon and inflammatory cascade in lung associated with infiltration of inflammatory macrophages and neutrophils, elaboration of NETs and cell death by pyroptosis mediates lethal H5N1 influenza in nonhuman primates, and by extension humans. These innate pathways represent promising therapeutic targets to prevent severe influenza and potentially other primary viral pneumonias in humans. Influenza can cause acute lung injury and death, but the mechanisms resulting in lethal influenza in humans are not well understood. We used a novel model of lethal influenza in nonhuman primates caused by aerosol infection with highly pathogenic avian influenza virus that closely resembles human disease to define how the virus causes severe pneumonia. We found that a potent innate immune response starting with high-level production of interferons and inflammatory factors in the lung drives severe disease. Inflammatory cells including macrophages and neutrophils were recruited into lung because of this early response, which in turn led to release of neutrophil extracellular traps that blocked lung alveoli. In addition, a particularly inflammatory form of cell death known as pyroptosis occurred in lungs during lethal influenza. These new findings show that an intense interferon response leading to an inflammatory cascade of macrophages and neutrophils, release of neutrophil extracellular traps, and cell death by pyroptosis is responsible for acute lung injury in lethal influenza. These innate pathways could be targeted by drugs to prevent lung injury in critically ill influenza patients.
Collapse
Affiliation(s)
- Jacqueline Corry
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (JC); (SMBB)
| | - Gwenddolen Kettenburg
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amit A. Upadhyay
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Megan Wallace
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michelle M. Marti
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth R. Wonderlich
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephanie J. Bissel
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kyndal Goss
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Timothy J. Sturgeon
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Steven E. Bosinger
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Simon M. Barratt-Boyes
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (JC); (SMBB)
| |
Collapse
|
137
|
Effects of electroacupuncture pretreatment on M1 polarization of alveolar macrophages in rats with acute lung injury. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2022. [DOI: 10.1007/s11726-022-1288-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
138
|
Ning Q, Wu D, Wang X, Xi D, Chen T, Chen G, Wang H, Lu H, Wang M, Zhu L, Hu J, Liu T, Ma K, Han M, Luo X. The mechanism underlying extrapulmonary complications of the coronavirus disease 2019 and its therapeutic implication. Signal Transduct Target Ther 2022; 7:57. [PMID: 35197452 PMCID: PMC8863906 DOI: 10.1038/s41392-022-00907-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.
Collapse
Affiliation(s)
- Qin Ning
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Di Wu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Xi
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwu Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Lu
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Ma
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meifang Han
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Luo
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
139
|
Van Slambrouck J, Van Raemdonck D, Vos R, Vanluyten C, Vanstapel A, Prisciandaro E, Willems L, Orlitová M, Kaes J, Jin X, Jansen Y, Verleden GM, Neyrinck AP, Vanaudenaerde BM, Ceulemans LJ. A Focused Review on Primary Graft Dysfunction after Clinical Lung Transplantation: A Multilevel Syndrome. Cells 2022; 11:cells11040745. [PMID: 35203392 PMCID: PMC8870290 DOI: 10.3390/cells11040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
Primary graft dysfunction (PGD) is the clinical syndrome of acute lung injury after lung transplantation (LTx). However, PGD is an umbrella term that encompasses the ongoing pathophysiological and -biological mechanisms occurring in the lung grafts. Therefore, we aim to provide a focused review on the clinical, physiological, radiological, histological and cellular level of PGD. PGD is graded based on hypoxemia and chest X-ray (CXR) infiltrates. High-grade PGD is associated with inferior outcome after LTx. Lung edema is the main characteristic of PGD and alters pulmonary compliance, gas exchange and circulation. A conventional CXR provides a rough estimate of lung edema, while a chest computed tomography (CT) results in a more in-depth analysis. Macroscopically, interstitial and alveolar edema can be distinguished below the visceral lung surface. On the histological level, PGD correlates to a pattern of diffuse alveolar damage (DAD). At the cellular level, ischemia-reperfusion injury (IRI) is the main trigger for the disruption of the endothelial-epithelial alveolar barrier and inflammatory cascade. The multilevel approach integrating all PGD-related aspects results in a better understanding of acute lung failure after LTx, providing novel insights for future therapies.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Cedric Vanluyten
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Elena Prisciandaro
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Pulmonary Circulation Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium;
| | - Michaela Orlitová
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yanina Jansen
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Geert M. Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arne P. Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
- Department of Anesthesiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
140
|
Investigating the Intercellular Communication Network of Immune Cell in Acute Respiratory Distress Syndrome with Sepsis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4586648. [PMID: 35222683 PMCID: PMC8866031 DOI: 10.1155/2022/4586648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/27/2022] [Indexed: 11/17/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is recognized as a serious public health issue that results in respiratory failure and high mortality rates. The syndrome is characterized by immune cell aggregation, communication, activation, and alveolar epithelial damage. To elucidate the complex dynamic process of the immune system's response in ARDS, we construct the intercellular communication network of immune cells in ARDS based on a single-cell RNA sequencing dataset (including three sepsis-induced ARDS patients and four sepsis-only patients). The results show that macrophages relayed most of the intercellular signals (ligand–receptor pairs) in both groups. Many genes related to immune response (IFI44L, ISG, and HLA-DQB1) and biological functions (response to virus, negative regulation of viral life cycle, and response to interferon-beta) were detected via differentially expressed gene analysis of macrophages between the two groups. Deep analysis of the intercellular signals related to the macrophage found that sepsis-induced ARDS harbored distinctive intercellular signals related to chemokine–chemokine receptors (CCL3/4/5−CCR1), which mainly are involved in the disturbance of the STAT family transcription factors (TFs), such as STAT2 and STAT3. These signals and downstream TFs might play key roles in macrophage M1/M2 polarization in the process of sepsis-induced ARDS. This study provides a comprehensive view of the intercellular communication landscape between sepsis and sepsis-induced ARDS and identifies key intercellular communications and TFs involved in sepsis-induced ARDS. We believe that our study provides valuable clues for understanding the immune response mechanisms of ARDS.
Collapse
|
141
|
Chen S, Chen L, Ye L, Jiang Y, Li Q, Zhang H, Zhang R, Li H, Yu D, Zhang R, Niu Y, Zhao Q, Liu J, Ouyang G, Aschner M, Zheng Y, Zhang L, Chen W, Li D. PP2A-mTOR-p70S6K/4E-BP1 axis regulates M1 polarization of pulmonary macrophages and promotes ambient particulate matter induced mouse lung injury. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127624. [PMID: 34740159 DOI: 10.1016/j.jhazmat.2021.127624] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/10/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
To identify key signaling pathways involved in ambient particulate matter (PM)-induced pulmonary injury, we generated a mouse model with myeloid-specific deletion of Ppp2r1a gene (encoding protein phosphatase 2 A (PP2A) A subunit), and conducted experiments in a real-ambient PM exposure system. PP2A Aα-/- homozygote (Aα HO) mice and matched wild-type (WT) littermates were exposed to PM over 3-week and 6-week. The effects of PM exposure on pulmonary inflammation, oxidative stress, and apoptosis were significantly enhanced in Aα HO compared to WT mice. The number of pulmonary macrophages increased by 74.8~88.0% and enhanced M1 polarization appeared in Aα HO mice upon PM exposure. Secretion of M1 macrophage-related inflammatory cytokines was significantly increased in Aα HO vs. WT mice following PM exposure. Moreover, we demonstrated that PP2A-B56α holoenzyme regulated M1 polarization and that the mTOR signaling pathway mediated the persistent M1 polarization upon PM2.5 exposure. Importantly, PP2A-B56α holoenzyme was shown to complex with mTOR/p70S6K/4E-BP1, and suppression of B56α led to enhanced phosphorylation of mTOR, p70S6K, and 4E-BP1. These observations demonstrate that the PP2A-mTOR-p70S6K/4E-BP1 signaling is a critical pathway in mediating macrophage M1 polarization, which contributes to PM-induced pulmonary injury.
Collapse
Affiliation(s)
- Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lizhu Ye
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qiong Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Haiyan Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huiyao Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Dianke Yu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujie Niu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Qun Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian 116023, China
| | - Jianhui Liu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian 116023, China
| | - Gangfeng Ouyang
- KLGHEI of Environment and Energy Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Yuxin Zheng
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Lihua Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian 116023, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
142
|
Zhai Z, Ouyang W, Yao Y, Zhang Y, Zhang H, Xu F, Gao C. Dexamethasone-loaded ROS-responsive poly(thioketal) nanoparticles suppress inflammation and oxidative stress of acute lung injury. Bioact Mater 2022; 14:430-442. [PMID: 35415281 PMCID: PMC8965854 DOI: 10.1016/j.bioactmat.2022.01.047] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is associated with excessive inflammatory response, leading to acute respiratory distress syndrome (ARDS) without timely treatment. A fewer effective drugs are available currently to treat the ALI/ARDS. Herein, a therapeutic nanoplatform with reactive oxygen species (ROS)-responsiveness was developed for the regulation of inflammation. Dexamethasone acetate (Dex) was encapsulated into poly(thioketal) polymers to form polymeric nanoparticles (NPs) (PTKNPs@Dex). The NPs were composed of poly(1,4-phenyleneacetonedimethylene thioketal) (PPADT) and polythioketal urethane (PTKU), in which the thioketal bonds could be cleaved by the high level of ROS at the ALI site. The PTKNPs@Dex could accumulate in the pulmonary inflammatory sites and release the encapsulated payloads rapidly, leading to the decreased ROS level, less generation of pro-inflammatory cytokines, and reduced lung injury and mortality of mice. RNA sequencing (RNA-seq) analysis showed that the therapeutic efficacy of the NPs was associated with the modulation of many immune and inflammation-linked pathways. These findings provide a newly developed nanoplatform for the efficient treatment of ALI/ARDS. A therapeutic nanoplatform with ROS-responsiveness was developed for the regulation of inflammation. NPs composed of low Mw of PPADT and high Mw of PTKU were loaded with dexamethasone to obtain a self-adaptive system. The Dex-loaded NPs significantly decreased lung inflammation, and reduced lung injury and mortality in vivo.
Collapse
Affiliation(s)
- Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Wei Ouyang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuqi Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haolan Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Corresponding author. Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Corresponding author. MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
143
|
Kim TO, Park KJ, Cho YN, Jin HM, Jo YG, Kim HS, Ju JK, Shin HJ, Kho BG, Kee SJ, Park YW. Altered distribution, activation and increased IL-17 production of mucosal-associated invariant T cells in patients with acute respiratory distress syndrome. Thorax 2022; 77:865-872. [PMID: 35086913 DOI: 10.1136/thoraxjnl-2021-217724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/06/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Mucosal-associated invariant T (MAIT) cells are a subset of innate-like T cells that are engaged in a number of diseases, but their roles in acute respiratory distress syndrome (ARDS) are not fully examined yet. This study aimed to examine levels and functions of MAIT cells in patients with ARDS. METHODS Peripheral blood samples from patients with ARDS (n=50) and healthy controls (HCs, n=50) were collected. Levels of MAIT cells, cytokines, CD69, programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) were measured by flow cytometry. RESULTS Circulating MAIT cell levels were significantly reduced in patients with ARDS than in HCs. MAIT cell levels were inversely correlated with disease severity and mortality. Cytokine production profiles in MAIT cells showed that percentages of interleukin (IL)-17 producing MAIT cell were significantly higher in patients with ARDS than in HCs. Patients with ARDS exhibited higher expression levels of CD69, PD-1 and LAG-3 in circulating MAIT cells. Moreover, levels of MAIT cells and expression levels of CD69, PD-1 and IL-17 in MAIT cells were higher in bronchoalveolar lavage fluid samples than in peripheral blood samples. Our in vitro experiments showed that MAIT cells triggered macrophages to produce proinflammatory cytokines such as tumour necrosis factor-α, IL-1β and IL-8. CONCLUSIONS This study demonstrates that circulating MAIT cells are numerically deficient in patients with ARDS. In addition, MAIT cells were found to be activated, migrate into lung, secrete IL-17 and then stimulate macrophages. These findings suggest that MAIT cells contribute to the worsening of inflammation in the lung of patients with ARDS.
Collapse
Affiliation(s)
- Tae-Ok Kim
- Pulmonology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Ki-Jeong Park
- Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Young-Nan Cho
- Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Hye-Mi Jin
- Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Young-Goun Jo
- Surgery, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Hyo Shin Kim
- Surgery, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Jae Kyun Ju
- Surgery, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Hong-Joon Shin
- Pulmonology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Bo-Gun Kho
- Pulmonology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Seung-Jung Kee
- Laboratory Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Yong-Wook Park
- Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Korea .,Rheumatology, Chonnam National University Bitgoeul Hospital, Gwangju, Korea
| |
Collapse
|
144
|
Zheng F, Pan Y, Yang Y, Zeng C, Fang X, Shu Q, Chen Q. Novel biomarkers for acute respiratory distress syndrome: genetics, epigenetics and transcriptomics. Biomark Med 2022; 16:217-231. [PMID: 35026957 DOI: 10.2217/bmm-2021-0749] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) can be induced by multiple clinical factors, including sepsis, acute pancreatitis, trauma, intestinal ischemia/reperfusion and burns. However, these factors alone may poorly explain the risk and outcomes of ARDS. Emerging evidence suggests that genomic-based or transcriptomic-based biomarkers may hold the promise to establish predictive or prognostic stratification methods for ARDS, and also to help in developing novel therapeutic targets for ARDS. Notably, genetic/epigenetic variations correlated with susceptibility and prognosis of ARDS and circulating microRNAs have emerged as potential biomarkers for diagnosis or prognosis of ARDS. Although limited by sample size, ethnicity and phenotypic heterogeneity, ongoing genetic/transcriptomic research contributes to the characterization of novel biomarkers and ultimately helps to develop innovative therapeutics for ARDS patients.
Collapse
Affiliation(s)
- Fei Zheng
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Yihang Pan
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Yang Yang
- Department of Intensive Care Medicine, The Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Congli Zeng
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Qiang Shu
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Qixing Chen
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| |
Collapse
|
145
|
Ishikawa S, Miyazawa M, Tanaka C, Uesawa R, Nishizawa J, Uemura R, Kobayashi I, Hobo S. Interferon gamma, lipopolysaccharide, and modified-live viral vaccines stimulation alter the mRNA expression of tumor necrosis factor α, inducible nitric oxide synthase, and interferon β in bovine alveolar macrophages. Vet Immunol Immunopathol 2022; 244:110378. [PMID: 34999416 DOI: 10.1016/j.vetimm.2021.110378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/27/2021] [Accepted: 12/31/2021] [Indexed: 10/19/2022]
Abstract
To understand the pathogenesis of bovine respiratory disease (BRD), it is necessary to elucidate the mechanisms of alveolar macrophage regulation by cytokines and pathogen-associated molecular patterns (PAMPs). Moreover, "non-specific effects (NSEs)" an innate immune regulatory mechanism in response to vaccines containing PAMPs, has recently attracted attention. It may be applied to BRD control, but there is limited knowledge in bovine. To investigate this, we stimulated alveolar macrophages in vitro with lipopolysaccharide (LPS), polyinosinic-polycytidylic acid sodium salt (Poly I:C), interferon gamma (IFN-γ), and modified-live viral (MLV) vaccines, respectively, and analyzed changes in tumor necrosis factor alpha (TNF-α), inducible nitric oxide synthase (iNOS), and interferon beta (IFN-β) mRNA expression levels. mRNA expression levels of TNF-α, iNOS, and IFN-β were significantly increased in bovine alveolar macrophages stimulated by IFN-γ and MLV vaccine; LPS, IFN-γ, and MLV vaccine; and MLV vaccine only, respectively. Additionally, all MLV vaccine-stimulated mRNA expression increases were observed in a concentration-dependent manner. These results revealed in part, the mechanism of bovine alveolar macrophage regulation by cytokines and PAMPs. Understanding the regulatory mechanisms of alveolar macrophages will contribute to understanding the pathogenesis of BRD and preventive and therapeutic BRD management based on NSEs.
Collapse
Affiliation(s)
- Shingo Ishikawa
- Division of Veterinary Science, Graduate School of Life and Environmental Biosciences, Osaka Prefecture University, Izumi-Sano, Osaka, 598-8531, Japan; Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Masataka Miyazawa
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Chiho Tanaka
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Ryoma Uesawa
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Juri Nishizawa
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan
| | - Ryoko Uemura
- Center for Animal Disease Control, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Ikuo Kobayashi
- Center for Animal Disease Control, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Seiji Hobo
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.
| |
Collapse
|
146
|
ISHIKAWA S, MIYAZAWA M, ZIBIKI Y, KAMIKAKIMOTO R, HOBO S. Flow cytometric analysis of bronchoalveolar lavage fluid immune dynamics in calves. J Vet Med Sci 2022; 84:548-557. [PMID: 35153256 PMCID: PMC9096045 DOI: 10.1292/jvms.21-0522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Understanding the immune dynamics in the respiratory mucosa of calves is necessary for a
good management of bovine respiratory disease. Immune dynamics in the respiratory mucosa
in humans and experimental animals has been assessed by flow cytometric analysis of
bronchoalveolar lavage fluid (BALF); however, few reports have addressed this subject in
calves. The aim of this study was to establish a universal method to analyze
bronchoalveolar lavage fluid (BALF) by flow cytometry and to obtain basic knowledge of
bovine respiratory mucosal immune dynamics. We investigated the immune cell populations in
BALF and evaluated the surface antigen expression of alveolar macrophages in calves using
flow cytometer. To further analyze the surface antigen variation observed in alveolar
macrophages in detail, stimulation assays were performed in vitro. BALF
cells were separated into three distinct populations based on their light scatter plot,
which were considered to be macrophages, lymphocytes, and neutrophils. In most
individuals, most of the BALF immune cells were alveolar macrophages, but an increased
proportion of lymphocytes and neutrophils was observed in some individuals. Analysis of
each surface antigen expression in alveolar macrophages showed that CD21 and MHC class II
expression changed in response to changes in the leukocyte population. Moreover, when
alveolar macrophages were stimulated with interferon-γ in vitro, the
expression of CD21 was drastically reduced and MHC class II was increased, suggesting that
functional changes in alveolar macrophages themselves are involved in the immune
dynamics.
Collapse
Affiliation(s)
- Shingo ISHIKAWA
- Division of Veterinary Science, Graduate School of Life and Environmental Biosciences, Osaka Prefecture University
| | | | | | | | - Seiji HOBO
- Joint Faculty of Veterinary Medicine, Kagoshima University
| |
Collapse
|
147
|
Jia Y, Li C, Yin M, Lin J, Zhang L, Li N, Jiang N, Xu Q, Wang Q, Gu L, Yu B, Zhao G. Kaempferol ameliorate the prognosis of Aspergillus fumigatus keratitis by reducing fungal load and inhibiting the Dectin-1 and p38 MAPK pathway. Exp Eye Res 2022; 216:108960. [DOI: 10.1016/j.exer.2022.108960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/04/2022]
|
148
|
Tissue-resident immunity in the lung: a first-line defense at the environmental interface. Semin Immunopathol 2022; 44:827-854. [PMID: 36305904 PMCID: PMC9614767 DOI: 10.1007/s00281-022-00964-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
The lung is a vital organ that incessantly faces external environmental challenges. Its homeostasis and unimpeded vital function are ensured by the respiratory epithelium working hand in hand with an intricate fine-tuned tissue-resident immune cell network. Lung tissue-resident immune cells span across the innate and adaptive immunity and protect from infectious agents but can also prove to be pathogenic if dysregulated. Here, we review the innate and adaptive immune cell subtypes comprising lung-resident immunity and discuss their ontogeny and role in distinct respiratory diseases. An improved understanding of the role of lung-resident immunity and how its function is dysregulated under pathological conditions can shed light on the pathogenesis of respiratory diseases.
Collapse
|
149
|
Sun S, Yao Y, Huang C, Xu H, Zhao Y, Wang Y, Zhu Y, Miao Y, Feng X, Gao X, Zheng J, Zhang Q. CD36 regulates LPS-induced acute lung injury by promoting macrophages M1 polarization. Cell Immunol 2022; 372:104475. [PMID: 35063904 DOI: 10.1016/j.cellimm.2021.104475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/02/2021] [Accepted: 12/31/2021] [Indexed: 01/11/2023]
|
150
|
Grau-Expósito J, Perea D, Suppi M, Massana N, Vergara A, Soler MJ, Trinite B, Blanco J, García-Pérez J, Alcamí J, Serrano-Mollar A, Rosado J, Falcó V, Genescà M, Buzon MJ. Evaluation of SARS-CoV-2 entry, inflammation and new therapeutics in human lung tissue cells. PLoS Pathog 2022; 18:e1010171. [PMID: 35025963 PMCID: PMC8791477 DOI: 10.1371/journal.ppat.1010171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/26/2022] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.
Collapse
Affiliation(s)
- Judith Grau-Expósito
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Marina Suppi
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Núria Massana
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Ander Vergara
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria José Soler
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Benjamin Trinite
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier García-Pérez
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Clinic HIV Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Anna Serrano-Mollar
- Experimental Pathology Department, Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Joel Rosado
- Thoracic Surgery and Lung Transplantation Department, Vall d’Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d’Hebron, VHIR Task Force COVID-19, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria J. Buzon
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| |
Collapse
|