1
|
Morris SB, Ocadiz-Ruiz R, Asai N, Malinczak CA, Rasky AJ, Lombardo GK, Velarde EM, Ptaschinski C, Zemans RL, Lukacs NW, Fonseca W. Long-term alterations in lung epithelial cells after EL-RSV infection exacerbate allergic responses through IL-1β-induced pathways. Mucosal Immunol 2024; 17:1072-1088. [PMID: 39069078 PMCID: PMC11610113 DOI: 10.1016/j.mucimm.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
Early-life (EL) respiratory infections increase pulmonary disease risk, especially EL-Respiratory Syncytial Virus (EL-RSV) infections linked to asthma. Mechanisms underlying asthma predisposition remain unknown. In this study, we examined the long-term effects on the lung after four weeks post EL-RSV infection. We identified alterations in the lung epithelial cell, with a rise in the percentage of alveolar type 2 epithelial cells (AT2) and a decreased percentage of cells in the AT1 and AT2-AT1 subclusters, as well as upregulation of Bmp2 and Krt8 genes that are associated with AT2-AT1 trans-differentiation, suggesting potential defects in lung repair processes. We identified persistent upregulation of asthma-associated genes, including Il33. EL-RSV-infected mice allergen-challenged exhibited exacerbated allergic response, with significant upregulation of Il33 in the lung and AT2 cells. Similar long-term effects were observed in mice exposed to EL-IL-1β. Notably, treatment with IL-1ra during acute EL-RSV infection mitigated the long-term alveolar alterations and the allergen-exacerbated response. Finally, epigenetic modifications in the promoter of the Il33 gene were detected in AT2 cells harvested from EL-RSV and EL-IL1β groups, suggesting that long-term alteration in the epithelium after RSV infection is dependent on the IL-1β pathway. This study provides insight into the molecular mechanisms of asthma predisposition after RSV infection.
Collapse
Affiliation(s)
- Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ramon Ocadiz-Ruiz
- Department of Bioengineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nobuhiro Asai
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace K Lombardo
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan M Velarde
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L Zemans
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Eshraghi R, Rafiei M, Hadian Jazi Z, Shafie D, Raisi A, Mirzaei H. MicroRNA-155 and exosomal microRNA-155: Small pieces in the cardiovascular diseases puzzle. Pathol Res Pract 2024; 257:155274. [PMID: 38626659 DOI: 10.1016/j.prp.2024.155274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024]
Abstract
MicroRNAs (miRs, miRNAs) are known to have a part in various human illnesses, such as those related to the heart. One particular miRNA, miR-155, has been extensively studied and has been found to be involved in hematopoietic lineage differentiation, immunity, viral infections, inflammation, as well as vascular remodeling. These processes have all been connected to cardiovascular diseases, including heart failure, diabetic heart disease, coronary artery disease, and abdominal aortic aneurysm. The impacts of miR-155 depend on the type of cell it is acting on and the specific target genes involved, resulting in different mechanisms of disease. Although, the exact part of miR-155 in cardiovascular illnesses is yet not fully comprehended, as some studies have shown it to promote the development of atherosclerosis while others have shown it to prevent it. As a result, to comprehend the underlying processes of miR-155 in cardiovascular disorders, further thorough study is required. It has been discovered that exosomes that could be absorbed by adjacent or distant cells, control post-transcriptional regulation of gene expression by focusing on mRNA. Exosomal miRNAs have been found to have a range of functions, including participating in inflammatory reactions, cell movement, growth, death, autophagy, as well as epithelial-mesenchymal transition. An increasing amount of research indicates that exosomal miRNAs are important for cardiovascular health and have a major role in the development of a number of cardiovascular disorders, including pulmonary hypertension, atherosclerosis, acute coronary syndrome, heart failure, and myocardial ischemia-reperfusion injury. Herein the role of miR-155 and its exosomal form in heart diseases are summarized.
Collapse
Affiliation(s)
- Reza Eshraghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Hadian Jazi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Davood Shafie
- Cardiology/Heart Failure and Transplantation, Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Raisi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
3
|
Tian C, Liu Q, Zhang X, Li Z. Blocking group 2 innate lymphoid cell activation and macrophage M2 polarization: potential therapeutic mechanisms in ovalbumin-induced allergic asthma by calycosin. BMC Pharmacol Toxicol 2024; 25:30. [PMID: 38650035 PMCID: PMC11036756 DOI: 10.1186/s40360-024-00751-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Calycosin, a flavonoid compound extracted from Astragalus membranaceus, has shown anti-asthma benefits in house dust mite-induced asthma. Recent studies have suggested that innate-type cells, including group 2 innate lymphoid cells (ILC2s) and macrophages, serve as incentives for type 2 immunity and targets for drug development in asthma. This work focuses on the effects of calycosin on the dysregulated ILC2s and macrophages in allergic asthma. METHODS In vivo, the asthmatic mouse model was established with ovalbumin (OVA) sensitization and challenge, and calycosin was intraperitoneally administered at doses of 20 and 40 mg/kg. In vivo, mouse primary ILC2s were stimulated with interleukin (IL)-33 and mouse RAW264.7 macrophages were stimulated with IL-4 and IL-13 to establish the cell models. Cells were treated with calycosin at doses of 5 and 10 µM. RESULTS In vivo, we observed significantly reduced numbers of eosinophils, neutrophils, monocyte macrophages and lymphocytes in the bronchoalveolar lavage fluid (BALF) of OVA-exposed mice with 40 mg/kg calycosin. Histopathological assessment showed that calycosin inhibited the airway inflammation and remodeling caused by OVA. Calycosin markedly decreased the up-regulated IL-4, IL-5, IL-13, IL-33, and suppression tumorigenicity 2 (ST2) induced by OVA in BALF and/or lung tissues of asthmatic mice. Calycosin repressed the augment of arginase 1 (ARG1), IL-10, chitinase-like 3 (YM1) and mannose receptor C-type 1 (MRC1) levels in the lung tissues of asthmatic mice. In vivo, calycosin inhibited the IL-33-induced activation as well as the increase of IL-4, IL-5, IL-13 and ST2 in ILC2s. Calycosin also repressed the increase of ARG1, IL-10, YM1 and MRC1 induced by IL-4 and IL-13 in RAW264.7 macrophages. In addition, we found that these changes were more significant in 40 mg/kg calycosin treatment than 20 mg/kg calycosin. CONCLUSIONS Collectively, this study showed that calycosin might attenuate OVA-induced airway inflammation and remodeling in asthmatic mice via preventing ILC2 activation and macrophage M2 polarization. Our study might contribute to further study of asthmatic therapy.
Collapse
Affiliation(s)
- Chunyan Tian
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Graduate, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Liu
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Zhang
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhuying Li
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
4
|
Wang X, Wan W, Zhang J, Lu J, Liu P. Efficient pulmonary fibrosis therapy via regulating macrophage polarization using respirable cryptotanshinone-loaded liposomal microparticles. J Control Release 2024; 366:1-17. [PMID: 38154539 DOI: 10.1016/j.jconrel.2023.12.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Lung inflammation and fibrogenesis are the two main characteristics during the development of pulmonary fibrosis (PF), which are particularly associated with pulmonary macrophages. In this context, whether cryptotanshinone (CTS) could alleviate PF through regulating macrophage polarization were preliminarily demonstrated in vitro. Then the time course of PF and its relationship with macrophage polarization was determined in BLM-induced mice based on cytokine levels in bronchoalveolar lavage fluid (BALF), lung histopathology, flow cytometric analysis, mRNA and protein expression. CTS was loaded into macrophage-targeted and responsively released mannose-modified liposomes (Man-lipo), and the liposomes were then embedded into mannitol microparticles (M-MPs) using spray drying to achieve efficient pulmonary delivery. Afterwards, how CTS regulates macrophage polarization in vivo during different time courses of PF was probed. Furthermore, the molecular mechanisms of CTS against PF by regulating macrophage polarization were elucidated in vivo and in vitro. The full-course therapy group could achieve comparable therapeutic effects compared with the positive control drug PFD group. CTS can alleviate PF through regulating macrophage polarization, mainly by inhibiting NLRP3/TGF-β1 pathway during the inflammation course and modulating MMP-9/TIMP-1 balance during the fibrosis development course, providing new insights into chronic PF treatment.
Collapse
Affiliation(s)
- Xiuhua Wang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiguo Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Jing Lu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.
| |
Collapse
|
5
|
Du W, Ren N, Xu Y, Chen X. Programmed cell death 4 governs NLRP3-mediated pyroptosis in septic lung disorders. Mol Biol Rep 2024; 51:77. [PMID: 38183433 DOI: 10.1007/s11033-023-08948-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
INTRODUCTION Sepsis is a pathogenic syndrome of prolonged excessive inflammation and immunosuppression produced by invading pathogens. Programmed cell death 4 (PDCD4) may be implicated in a range of inflammatory lesions, and this study aimed to confirm the involvement of PDCD4 in septic lung injury. MATERIALS AND METHODS Mice and bronchial epithelial 16HBE cells were separately subjected to CLP and LPS to generate in vivo and in vitro models. Following the level of PDCD4 was determined, the impacts of PDCD4 knockdown on mouse lung injury degree, inflammation, apoptosis, and pyroptosis levels were evaluated. Afterward, cells were treated with the NLRP3 agonist, and the influences of NLRP3 activation on the regulations of PDCD4 knockdown were determined. RESULTS PDCD4 was elevated following mice developed septic lung injury, PDCD4 knockdown ameliorated septic lung injury and reduced lung inflammation and apoptosis. Moreover, PDCD4 knockdown suppressed NLRP3-mediated pyroptosis, indicating that PDCD4 also mediated pyroptosis. According to cellular models, NLRP3 activation broke the effects of PDCD4 knockdown on cells. CONCLUSIONS The current study reveals that PDCD4 governs NLRP3-mediated pyroptosis in septic lung injury. PDCD4 is not only related to apoptosis and expands the knowledge of PDCD4 regulation of different cell death modes.
Collapse
Affiliation(s)
- Wenjie Du
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266000, Shandong, China
| | - Na Ren
- Department of Emergency Internal Medicine, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, 266033, Shandong, China
| | - Yan Xu
- Quality Control Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, 266033, Shandong, China
| | - Xiao Chen
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
6
|
Xu Y, Lan P, Wang T. The Role of Immune Cells in the Pathogenesis of Idiopathic Pulmonary Fibrosis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1984. [PMID: 38004032 PMCID: PMC10672798 DOI: 10.3390/medicina59111984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown etiology with limited treatment options. The role of the immune system in IPF has received increasing attention. Uncontrolled immune responses drive the onset and progression of IPF. This article provides an overview of the role of innate immune cells (including macrophages, neutrophils, mast cells, eosinophils, dendritic cells, nature killer cells, nature kill cells and γδ T cells) and adaptive immune cells (including Th1 cells, Th2 cells, Th9 cells, Th17 cells, Th22 cells, cytotoxic T cells, B lymphocytes and Treg cells) in IPF. In addition, we review the current status of pharmacological treatments for IPF and new developments in immunotherapy. A deeper comprehension of the immune system's function in IPF may contribute to the development of targeted immunomodulatory therapies that can alter the course of the disease.
Collapse
Affiliation(s)
- Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Khayati S, Dehnavi S, Sadeghi M, Tavakol Afshari J, Esmaeili SA, Mohammadi M. The potential role of miRNA in regulating macrophage polarization. Heliyon 2023; 9:e21615. [PMID: 38027572 PMCID: PMC10665754 DOI: 10.1016/j.heliyon.2023.e21615] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophage polarization is a dynamic process determining the outcome of various physiological and pathological situations through inducing pro-inflammatory responses or resolving inflammation via exerting anti-inflammatory effects. The miRNAs are epigenetic regulators of different biologic pathways that target transcription factors and signaling molecules to promote macrophage phenotype transition and regulate immune responses. Modulating the macrophage activation, differentiation, and polarization by miRNAs is crucial for immune responses in response to microenvironmental signals and under various physiological and pathological conditions. In term of clinical significance, regulating macrophage polarization via miRNAs could be utilized for inflammation control. Also, understanding the role of miRNAs in macrophage polarization can provide insights into diagnostic strategies associated with dysregulated miRNAs and for developing macrophage-centered therapeutic methods. In this case, targeting miRNAs to further regulate of macrophage polarization may become an efficient strategy for treating immune-associated disorders. The current review investigated and categorized various miRNAs directly or indirectly involved in macrophage polarization by targeting different transcription factors and signaling pathways. In addition, prospects for regulating macrophage polarization via miRNA as a therapeutic choice that could be implicated in various pathological conditions, including cancer or inflammation-mediated injuries, were discussed.
Collapse
Affiliation(s)
- Shaho Khayati
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Patrucco F, Solidoro P, Gavelli F, Apostolo D, Bellan M. Idiopathic Pulmonary Fibrosis and Post-COVID-19 Lung Fibrosis: Links and Risks. Microorganisms 2023; 11:microorganisms11040895. [PMID: 37110318 PMCID: PMC10146995 DOI: 10.3390/microorganisms11040895] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is considered the paradigmatic example of chronic progressive fibrosing disease; IPF does not result from a primary immunopathogenic mechanism, but immune cells play a complex role in orchestrating the fibrosing response. These cells are activated by pathogen-associated or danger-associated molecular patterns generating pro-fibrotic pathways or downregulating anti-fibrotic agents. Post-COVID pulmonary fibrosis (PCPF) is an emerging clinical entity, following SARS-CoV-2 infection; it shares many clinical, pathological, and immune features with IPF. Similarities between IPF and PCPF can be found in intra- and extracellular physiopathological pro-fibrotic processes, genetic signatures, as well as in the response to antifibrotic treatments. Moreover, SARS-CoV-2 infection can be a cause of acute exacerbation of IPF (AE-IPF), which can negatively impact on IPF patients’ prognosis. In this narrative review, we explore the pathophysiological aspects of IPF, with particular attention given to the intracellular signaling involved in the generation of fibrosis in IPF and during the SARS-CoV-2 infection, and the similarities between IPF and PCPF. Finally, we focus on COVID-19 and IPF in clinical practice.
Collapse
Affiliation(s)
- Filippo Patrucco
- Respiratory Diseases Unit, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
- Correspondence:
| | - Paolo Solidoro
- Medical Sciences Department, University of Turin, 10126 Turin, Italy
- Respiratory Diseases Unit, Cardiovascular and Thoracic Department, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Francesco Gavelli
- Translational Medicine Department, University of Eastern Piedmont, 28100 Novara, Italy
- Emergency Medicine Department, Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Daria Apostolo
- Translational Medicine Department, University of Eastern Piedmont, 28100 Novara, Italy
| | - Mattia Bellan
- Translational Medicine Department, University of Eastern Piedmont, 28100 Novara, Italy
- Division of Internal Medicine, Medical Department, AOU Maggiore della Carità di Novara, 28100 Novara, Italy
| |
Collapse
|
9
|
Chen Y, He H, Luo L, Liu K, Jiang M, Li S, Zhang X, Yang X, Liu Q. Studying pulmonary fibrosis due to microbial infection via automated microscopic image analysis. Front Microbiol 2023; 14:1176339. [PMID: 37032846 PMCID: PMC10076525 DOI: 10.3389/fmicb.2023.1176339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Pulmonary fibrosis is a consequential complication of microbial infections, which has notably been observed in SARS-CoV-2 infections in recent times. Macrophage polarization, specifically the M2-type, is a significant mechanism that induces pulmonary fibrosis, and its role in the development of Post- COVID-19 Pulmonary Fibrosis is worth investigating. While pathological examination is the gold standard for studying pulmonary fibrosis, manual review is subject to limitations. In light of this, we have constructed a novel method that utilizes artificial intelligence techniques to analyze fibro-pathological images. This method involves image registration, cropping, fibrosis degree classification, cell counting and calibration, and it has been utilized to analyze microscopic images of COVID-19 lung tissue. Methods Our approach combines the Transformer network with ResNet for fibrosis degree classification, leading to a significant improvement over the use of ResNet or Transformer individually. Furthermore, we employ semi-supervised learning which utilize both labeled and unlabeled data to enhance the ability of the classification network in analyzing complex samples. To facilitate cell counting, we applied the Trimap method to localize target cells. To further improve the accuracy of the counting results, we utilized an effective area calibration method that better reflects the positive density of target cells. Results The image analysis method developed in this paper allows for standardization, precision, and staging of pulmonary fibrosis. Analysis of microscopic images of COVID-19 lung tissue revealed a significant number of macrophage aggregates, among which the number of M2-type macrophages was proportional to the degree of fibrosis. Discussion The image analysis method provids a more standardized approach and more accurate data for correlation studies on the degree of pulmonary fibrosis. This advancement can assist in the treatment and prevention of pulmonary fibrosis. And M2-type macrophage polarization is a critical mechanism that affects pulmonary fibrosis, and its specific molecular mechanism warrants further exploration.
Collapse
Affiliation(s)
- Yajie Chen
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Henghui He
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Licheng Luo
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Kangyi Liu
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jiang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqi Li
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqi Zhang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yang
- School of Electronic Information and Communications, Huazhong University of Science and Technology, Wuhan, China
- Xin Yang,
| | - Qian Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Qian Liu,
| |
Collapse
|
10
|
Rasaei R, Tyagi A, Rasaei S, Lee SJ, Yang SR, Kim KS, Ramakrishna S, Hong SH. Human pluripotent stem cell-derived macrophages and macrophage-derived exosomes: therapeutic potential in pulmonary fibrosis. Stem Cell Res Ther 2022; 13:433. [PMID: 36056418 PMCID: PMC9438152 DOI: 10.1186/s13287-022-03136-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/14/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary fibrosis (PF) is a fatal chronic disease characterized by accumulation of extracellular matrix and thickening of the alveolar wall, ultimately leading to respiratory failure. PF is thought to be initiated by the dysfunction and aberrant activation of a variety of cell types in the lung. In particular, several studies have demonstrated that macrophages play a pivotal role in the development and progression of PF through secretion of inflammatory cytokines, growth factors, and chemokines, suggesting that they could be an alternative therapeutic source as well as therapeutic target for PF. In this review, we describe the characteristics, functions, and origins of subsets of macrophages involved in PF and summarize current data on the generation and therapeutic application of macrophages derived from pluripotent stem cells for the treatment of fibrotic diseases. Additionally, we discuss the use of macrophage-derived exosomes to repair fibrotic lung tissue.
Collapse
Affiliation(s)
- Roya Rasaei
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Apoorvi Tyagi
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Shima Rasaei
- Department of Cellular and Molecular Science, Falavarjan Branch, Islamic Azad University, Falavarjan, Iran
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea.
- Institute of Medical Science, Kangwon National University, Chuncheon, 24341, South Korea.
- KW-Bio Co., Ltd, Wonju, South Korea.
| |
Collapse
|
11
|
Peng H, Shi S, Lu Z, Liu L, Peng S, Wei P, Yi T. HOCl-Activated Reactive Organic Selenium Delivery Platform for Alleviation of Inflammation. Bioconjug Chem 2022; 33:1602-1608. [PMID: 36018225 DOI: 10.1021/acs.bioconjchem.2c00349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Selenium plays an important role in the biological system and can be used to treat various types of diseases. However, the current selenium delivery systems face the problems of low activity of released Se-containing compounds or nonspecific toxicity of reactive organic selenium donors in living systems. In response to these problems, we constructed a reactive organic selenium delivery platform by the activation of HOCl. Compared with prodrugs without activation capability, the hypochloroselenoite derivatives released from the present platform after activation displayed higher reactivity and could react with various nucleophiles to participate in specific life processes. Taking the selected compound (DHU-Se1) as an example, we found that it could alleviate the process of inflammation by blocking the polarization of macrophages from M0 to M1. Therefore, the development of this system is of great significance for expanding the application of selenium-containing compounds and treating related diseases.
Collapse
Affiliation(s)
- Hongying Peng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Shi Shi
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhenni Lu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Lingyan Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Shuxin Peng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Peng Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Tao Yi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
12
|
Wu GR, Zhou M, Wang Y, Zhou Q, Zhang L, He L, Zhang S, Yu Q, Xu Y, Zhao J, Xiong W, Wang CY. Blockade of Mbd2 by siRNA-loaded liposomes protects mice against OVA-induced allergic airway inflammation via repressing M2 macrophage production. Front Immunol 2022; 13:930103. [PMID: 36090987 PMCID: PMC9453648 DOI: 10.3389/fimmu.2022.930103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo address the role of methyl-CpG-binding domain 2 (MBD2) in the pathogenesis of asthma and its potential as a target for the asthmatic therapy.MethodsStudies were conducted in asthmatic patients and macrophage-specific Mbd2 knockout mice to dissect the role of MBD2 in asthma pathogenesis. Additionally, RNAi-based therapy with Mbd2 siRNA-loaded liposomes was conducted in an ovalbumin (OVA)-induced allergic airway inflammation mouse model.ResultsAsthmatic patients and mice challenged with OVA exhibited upregulated MBD2 expression in macrophages, especially in alternatively activated (M2) macrophages. In particular, macrophage-specific knockout of Mbd2 protected mice from OVA-induced allergic airway inflammation and suppressed the M2 program. Notably, intratracheal administration of liposomes carrying Mbd2 siRNA decreased the expression of Mbd2 and prevented OVA-induced allergic airway inflammation in mice, as indicated by the attenuated airway inflammation and mucus production.ConclusionsThe above data indicate that Mbd2 implicates in the pathogenesis of asthma predominantly by regulating the polarization of M2 macrophages, which supports that Mbd2 could be a viable target for treatment of asthma in clinical settings.
Collapse
Affiliation(s)
- Guo-Rao Wu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Lei Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Long He
- Department of Clinical Laboratory, Shanghai East Hospital; School of Medicine, Tongji University, Shanghai, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
- Department of Respiratory and Critical Care Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Cong-Yi Wang, ; Weining Xiong,
| | - Cong-Yi Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
- *Correspondence: Cong-Yi Wang, ; Weining Xiong,
| |
Collapse
|
13
|
Liang ZY, Xu XJ, Rao J, Yang ZL, Wang CH, Chen CM. Mesenchymal Stem Cell-Derived Exosomal MiRNAs Promote M2 Macrophages Polarization: Therapeutic Opportunities for Spinal Cord Injury. Front Mol Neurosci 2022; 15:926928. [PMID: 35903172 PMCID: PMC9319398 DOI: 10.3389/fnmol.2022.926928] [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: 04/23/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) is an enormous public health concern affecting approximately 250,000–500,000 people worldwide each year. It is mostly irreversible considering the limitations of currently available treatments, and its prevention and management have been the prime focus of many studies. Mesenchymal stem cell (MSC) transplantation is one of the most promising treatments for SCI. The role of MSCs in SCI has been studied extensively, and MSCs have been shown to have many limitations. Moreover, the therapeutic effects of MSCs are more likely related to paracrine effects. In SCIs, macrophages from peripheral sources differentiate into M1 macrophages, promoting inflammation and aggravating neuronal damage; however, studies have shown that MSC-derived exosomes can induce the polarization of macrophages from the M1 to the M2 phenotype, thereby promoting nerve function recovery in patients with SCI. In this review, we discussed the research progress of MSC-derived exosomal miRNAs in promoting M2 macrophage differentiation in the SCI, and introduced some exosomal miRNAs that can regulate the differentiation of M2 macrophages in non-SCI; it is hoped that the regulatory role of these exosome-derived miRNAs can be confirmed in SCI.
Collapse
Affiliation(s)
- Ze-Yan Liang
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| | | | | | | | - Chun-Hua Wang
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| | - Chun-Mei Chen
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| |
Collapse
|
14
|
Du X, Osoro EK, Chen Q, Yan X, Gao D, Wu L, Ren J, Feng L, Wu N, Lu K, Yang X, Zhong B, Han Y, Zhang F, Li D, Lan X, Lu S. Pdcd4 promotes lipid deposition by attenuating PPARα-mediated fatty acid oxidation in hepatocytes. Mol Cell Endocrinol 2022; 545:111562. [PMID: 35051553 DOI: 10.1016/j.mce.2022.111562] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes. The involvement of programmed cell death 4 (Pdcd4) in inflammation and metabolic diseases has been widely reported. However, the precise regulatory role of Pdcd4 in hepatocytic lipid metabolism and NAFLD is not well known. RESEARCH DESIGN AND METHODS We established a high-fat diet-induced NAFLD (HFD-NAFLD) rat model and a free fatty acids (FFAs)-treated cell model, and analyzed the expression and distribution of PDCD4. The lentivirus for Pdcd4 knockout and the vector for Pdcd4 overexpression were used to alter Pdcd4 expression in BRL 3A cells. Thereafter, lipid accumulation, FA metabolic gene expression, and peroxisome proliferator-activated receptor alpha (Pparα)-dependent peroxisomal β-oxidation-related gene expression, especially that of the critical transcription factors and enzymes acyl-CoA oxidases 1-3 (Acox1-3), were detected both at the mRNA and protein levels. RESULTS PDCD4 expression increased and it was mainly distributed in hepatocyte nuclei of the HFD-NAFLD rats. as well as the FFAs-treated CBRH-7919 and BRL 3A cell lines. Pdcd4 knockout significantly suppressed FFAs-induced lipid accumulation, and Pdcd4 overexpression accelerated FFAs-induced lipid accumulation in hepatocytes. Mechanistically, Pdcd4 negatively regulated the expression Pparα and Acox1-3. In addition, rescue experiments confirmed that Pparα knockdown could attenuate the expression of Acox1-3 in Pdcd4 knockout cells, which ultimately restored lipid deposition to normal levels. PPARα expression decreased in the liver of the HFD-NAFLD rats. The enrichment of PDCD4 in hepatocyte nuclei correlated with lower PPARα expression after FFAs treatment in vitro. CONCLUSION Our results indicate that the abundance of PDCD4 under high-fat conditions facilitates hepatocellular lipid accumulation by decreasing PPARα-dependent FA peroxisomal β-oxidation.
Collapse
Affiliation(s)
- Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Qian Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Dan Gao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Jiajun Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Lina Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Nan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Kaikai Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Xudong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Bo Zhong
- Department of Pediatrics, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China
| | - Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Beijing, China.
| |
Collapse
|
15
|
Abu Khweek A, Joldrichsen MR, Kim E, Attia Z, Krause K, Daily K, Estfanous S, Hamilton K, Badr A, Anne MNK, Eltobgy M, Corps KN, Carafice C, Zhang X, Gavrilin MA, Boyaka PN, Amer AO. Caspase-11 regulates lung inflammation in response to house dust mites. Cell Immunol 2021; 370:104425. [PMID: 34800762 PMCID: PMC8714054 DOI: 10.1016/j.cellimm.2021.104425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Asthma is an inflammatory lung disorder characterized by mucus hypersecretion, cellular infiltration, and bronchial hyper-responsiveness. House dust mites (HDM) are the most prevalent cause of allergic sensitization. Canonical and noncanonical inflammasomes are multiprotein complexes that assemble in response to pathogen or danger-associated molecular patterns (PAMPs or DAMPs). Murine caspase-11 engages the noncanonical inflammasome. We addressed the role of caspase-11 in mediating host responses to HDM and subsequent allergic inflammation using caspase-11-/- mice, which lack caspase-11 while express caspase-1. We found that HDM induce caspase-11 expression in vitro. The presence of IL-4 and IL-13 promote caspase-11 expression. Additionally, caspase-11-/- macrophages show reduced release of IL-6, IL-12, and KC, and express lower levels of costimulatory molecules (e.g., CD40, CD86 and MHCII) in response to HDM stimulation. Notably, HDM sensitization of caspase-11-/- mice resulted in similar levels of IgE responses and hypothermia in response to nasal HDM challenge compared to WT. However, analysis of cell numbers and cytokines in bronchiolar alveolar lavage fluid (BALF) and histopathology of representative lung segments demonstrate altered inflammatory responses and reduced neutrophilia in the airways of the caspase-11-/- mice. These findings indicate that caspase-11 regulates airway inflammation in response to HDM exposure.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Marisa R Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kylene Daily
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Asmaa Badr
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Midhun N K Anne
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kara N Corps
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus OH 43210, USA
| | - Mikhail A Gavrilin
- Department of Internal Medicine, The Ohio State University, Columbus OH 43210, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA; Infectious Diseases Institute, The Ohio State University, Columbus OH 43210, USA.
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA.
| |
Collapse
|
16
|
Braun C, Katholnig K, Kaltenecker C, Linke M, Sukhbaatar N, Hengstschläger M, Weichhart T. p38 regulates the tumor suppressor PDCD4 via the TSC-mTORC1 pathway. Cell Stress 2021; 5:176-182. [PMID: 34917890 PMCID: PMC8645265 DOI: 10.15698/cst2021.12.260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Programmed cell death protein 4 (PDCD4) exerts critical functions as tumor suppressor and in immune cells to regulate inflammatory processes. The phosphoinositide 3-kinase (PI3K) promotes degradation of PDCD4 via mammalian target of rapamycin complex 1 (mTORC1). However, additional pathways that may regulate PDCD4 expression are largely ill-defined. In this study, we have found that activation of the mitogen-activated protein kinase p38 promoted degradation of PDCD4 in macrophages and fibroblasts. Mechanistically, we identified a pathway from p38 and its substrate MAP kinase-activated protein kinase 2 (MK2) to the tuberous sclerosis complex (TSC) to regulate mTORC1-dependent degradation of PDCD4. Moreover, we provide evidence that TSC1 and TSC2 regulate PDCD4 expression via an additional mechanism independent of mTORC1. These novel data extend our knowledge of how PDCD4 expression is regulated by stress- and nutrient-sensing pathways.
Collapse
Affiliation(s)
- Clarissa Braun
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
- Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karl Katholnig
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Christopher Kaltenecker
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Monika Linke
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Nyamdelger Sukhbaatar
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Li S, Gao S, Jiang Q, Liang Q, Luan J, Zhang R, Zhang F, Ruan H, Li X, Li X, Zhou H, Yang C. Clevudine attenuates bleomycin-induced early pulmonary fibrosis via regulating M2 macrophage polarization. Int Immunopharmacol 2021; 101:108271. [PMID: 34700113 DOI: 10.1016/j.intimp.2021.108271] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022]
Abstract
Pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease. It is a growing clinical problem which can result in breathlessness or respiratory failure and has an average life expectancy of 3 years from diagnosis. Predominantly accumulation of M2 macrophages accelerates fibrosis progression by secreting multiple cytokines that promote fibroblast to myofibroblast transition and aberrant wound healing of epithelial cells. Targeting activated macrophages to inhibit the pro-fibrotic phenotype is considered as an approach for the potential treatment of PF. Clevudine is s a purine nucleoside analogue which in an oral formulation is approved for treatment of patients with hepatitis B virus (HBV). Here, we found that clevudine is capable of suppressing pro-fibrotic phenotype (i.e., CD206, Arg1 and YM1) of M2 macrophages while enhancing anti-fibrotic phenotype (i.e., CD86, IL-6 and IL-10) by inhibiting PI3K/Akt signaling pathway. This effect further alleviates M2-induced myofibroblast activation and epithelial-to-mesenchymal transition (EMT), thus resulting in a decline of collagen deposition, pro-fibrotic cytokines secretion, with a concomitant recover ofpulmonary functions in vivo. Less infiltration of M2 macrophages between α-SMA + cells was also found in clevudine treated mice. Our findings indicate a potential anti-fibrotic effect of clevudine by regulating macrophage polarization and might be meaningful in clinical settings.
Collapse
Affiliation(s)
- Shuangling Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Qiuyan Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Qing Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Jiaoyan Luan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Ruiqin Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Fangxia Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Hao Ruan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| |
Collapse
|
18
|
Sharma RK, Goswami B, Das Mandal S, Guha A, Willard B, Ray PS. Quorum Sensing by Gelsolin Regulates Programmed Cell Death 4 Expression and a Density-Dependent Phenotype in Macrophages. THE JOURNAL OF IMMUNOLOGY 2021; 207:1250-1264. [PMID: 34362832 DOI: 10.4049/jimmunol.2001392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/28/2021] [Indexed: 01/03/2023]
Abstract
Quorum-sensing mechanisms that sense the density of immune cells at the site of inflammation to initiate inflammation resolution have recently been demonstrated as a major determinant of the inflammatory response. We observed a density-dependent increase in expression of the inflammatory tumor suppressor protein programmed cell death 4 (PDCD4) in mouse macrophage cells. Conditioned medium from high-density cells upregulated PDCD4 expression, revealing the presence of a secreted factor(s) acting as a macrophage quorum sensor. Secreted gelsolin (GSN) was identified as the quorum-sensing autoinducer. Alteration of GSN levels changed PDCD4 expression and the density-dependent phenotype of cells. LPS induced the expression of microRNA miR-21, which downregulated both GSN and PDCD4 expression, and reversed the high-density phenotype. The high-density phenotype was correlated with an anti-inflammatory gene expression program, which was counteracted by inflammatory stimulus. Together, our observations establish the miR-21-GSN-PDCD4 regulatory network as a crucial mediator of a macrophage quorum-sensing mechanism for the control of inflammatory responses.
Collapse
Affiliation(s)
- Reshma Kumari Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, India
| | - Binita Goswami
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, India
| | - Sukhen Das Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, India
| | - Abhishek Guha
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, India.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL; and
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Partho Sarothi Ray
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, India;
| |
Collapse
|
19
|
Chen G, Li PH, He JY, Su YL, Chen HJ, Dong JD, Huang YH, Huang XH, Jiang YF, Qin QW, Sun HY. Molecular cloning, inducible expression with SGIV and Vibrio alginolyticus challenge, and function analysis of Epinephelus coioides PDCD4. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104013. [PMID: 33465381 DOI: 10.1016/j.dci.2021.104013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Programmed cell death 4 (PDCD4) in mammals, a gene closely associated with apoptosis, is involved in many biological processes, such as cell aging, differentiation, regulation of cell cycle, and inflammatory response. In this study, grouper Epinephelus coioides PDCD4, EcPDCD4-1 and EcPDCD4-2, were obtained. The open reading frame (ORF) of EcPDCD4-1 is 1413 bp encoding 470 amino acids with a molecular mass of 52.39 kDa and a theoretical pI of 5.33. The ORF of EcPDCD4-2 is 1410 bp encoding 469 amino acids with a molecular mass of 52.29 kDa and a theoretical pI of 5.29. Both EcPDCD4-1 and EcPDCD4-2 proteins contain two conserved MA3 domains, and their mRNA were detected in all eight tissues of E. coioides by quantitative real-time PCR (qRT-PCR) with the highest expression in liver. The expressions of two EcPDCD4s were significantly up-regulated after Singapore grouper iridovirus (SGIV) or Vibrio alginolyticus infection. In addition, over-expression of EcPDCD4-1 or EcPDCD4-2 can inhibit the activity of the nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), and regulate SGIV-induced apoptosis. The results demonstrated that EcPDCD4s might play important roles in E. coioides tissues during pathogen-caused inflammation.
Collapse
Affiliation(s)
- Guo Chen
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China; Hainan Key Laboratory of Tropical Marine Biotechnology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, PR China; Department of Laboratory, Jining No.1 People's Hospital; Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Shandong, 272111, PR China; Life Sciences Institute, Zhejiang University, Zhejiang Province, 310058, PR China
| | - Pin-Hong Li
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jia-Yang He
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Yu-Ling Su
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - He-Jia Chen
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jun-De Dong
- Hainan Key Laboratory of Tropical Marine Biotechnology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, PR China
| | - You-Hua Huang
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xiao-Hong Huang
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Yu-Feng Jiang
- Department of Laboratory, Jining No.1 People's Hospital; Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Shandong, 272111, PR China.
| | - Qi-Wei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| | - Hong-Yan Sun
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| |
Collapse
|
20
|
The μ-opioid receptor induces miR-21 expression and is ERK/PKCμ-dependent. J Neuroimmunol 2021; 356:577585. [PMID: 33940234 DOI: 10.1016/j.jneuroim.2021.577585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022]
Abstract
Micro RNA-21 (miR-21) is believed to perform an important role in the transition from inflammation to resolution in the innate immune response. The biochemical basis for the induction of miR-21 remains uncertain. However, the activation of the μ-opioid receptor (MOR) induces the expression of miR-21. Our results show that human monocytes treated with μ-opioid agonists exhibit a significant increase in miR-21 expression. We found that MOR-induction of miR-21 requires the activation of the Ras-Raf-MEK-ERK signaling cascade, and to our surprise, the activation of PKCμ (PKD1). These results are significant given the role of miR-21 in the sensitivity to pain.
Collapse
|
21
|
Osoro EK, Du X, Liang D, Lan X, Farooq R, Huang F, Zhu W, Ren J, Sadiq M, Tian L, Yang X, Li D, Lu S. Induction of PDCD4 by albumin in proximal tubule epithelial cells potentiates proteinuria-induced dysfunctional autophagy by negatively targeting Atg5. Biochem Cell Biol 2021; 99:617-628. [PMID: 33831322 DOI: 10.1139/bcb-2021-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The precise molecular mechanism of autophagy dysfunction in type 1 diabetes is not known. Herein, the role of programmed cell death 4 (PDCD4) in autophagy regulation in the pathogenesis of diabetic kidney disease (DKD) in vivo and in vitro was described. It was found that Pdcd4 mRNA and protein was upregulated in the streptozotocin (STZ)-induced DKD rats. In addition, a unilateral ureteral obstruction mouse model displayed an upregulation of PDCD4 in the disease group. kidney biopsy samples of human DKD patients showed an upregulation of PDCD4. Furthermore, western blotting of the STZ-induced DKD rat tissues displayed a low microtubule-associated protein 1A/1B-light chain 3 (LC3)-II, as compared to the control. It was found that albumin overload in cultured PTEC could upregulate the expression of PDCD4 and p62, and decrease the expression of LC3-II and autophagy-related 5 (Atg5) proteins. The knockout of Pdcd4 in cultured PTECs could lessen albumin-induced dysfunctional autophagy as evidenced by the recovery of Atg5 and LC3-II protein. The forced expression of PDCD4 could further suppress the expression of crucial autophagy-related gene Atg5. Herein, endogenous PDCD4 was shown to promote proteinuria-induced dysfunctional autophagy by negatively regulating Atg5. PDCD4 might therefore be a potential therapeutic target in DKD.
Collapse
Affiliation(s)
- Ezra Kombo Osoro
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Xiaojuan Du
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Dong Liang
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Xi Lan
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Riaz Farooq
- Xi'an Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Fumeng Huang
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Wenhua Zhu
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Jiajun Ren
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Muhammad Sadiq
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Lifang Tian
- Xi'an Jiaotong University, 12480, Department of Nephrology, the Second Affiliated Hospital, Xi'an, Shaanxi, China;
| | - Xudong Yang
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Dongmin Li
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| | - Shemin Lu
- Xian Jiaotong University, 12480, Biochemistry and Molecular Biology, Xi'an, Shaanxi, China.,Xi'an Jiaotong University, 12480, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, Shaanxi, China;
| |
Collapse
|
22
|
Nurrahmah QI, Madhyastha R, Madhyastha H, Purbasari B, Maruyama M, Nakajima Y. Retinoic acid abrogates LPS-induced inflammatory response via negative regulation of NF-kappa B/miR-21 signaling. Immunopharmacol Immunotoxicol 2021; 43:299-308. [PMID: 33757404 DOI: 10.1080/08923973.2021.1902348] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CONTEXT Macrophages are essential components of the immune system, with significant roles in inflammation modulation. They can be activated into pro-inflammatory M1 or anti-inflammatory M2 phenotypes, depending on their micro-environment. Molecular factors that modulate macrophage polarization are hot targets for therapeutic strategies to counter chronic inflammatory pathological conditions. OBJECTIVE The current study aimed to elucidate the molecular mechanisms by which Retinoic acid (RA), a potent immunomodulator, suppresses LPS-induced inflammatory response in macrophages. MATERIALS AND METHODS RAW 264.7 macrophages were treated with RA and/or LPS, and analyzed for inflammatory genes and miR-21 by PCR. The roles of miR-21 and NF-ĸB signaling pathway were also assessed by knock-down experiments, immunofluorescence, and ChIP assays. RESULTS Pretreatment with RA quenched the LPS-induced inflammatory responses, including phagocytosis, ROS generation, and NO production. RA shifted the polarization away from the M1 state by negative regulation of IKKα/β, p65, and miR-21. RA hindered the phosphorylation of IKKα/β, translocation of p65 into the nucleus, and the subsequent upregulation of miR-21. Knock-in and knock-down experiments showed that miR-21 is central for the polarization shift toward the pro-inflammatory M1 state. CONCLUSION miR-21 is involved in the LPS-induced pro-inflammatory profile of macrophages and that RA negatively regulates the inflammatory response by targeting NF-ĸB/miR-21 signaling. Our data exposes RA's potential as a pharmacological agent to manipulate miR-21 and counteract hyper-inflammatory response.
Collapse
Affiliation(s)
- Queen Intan Nurrahmah
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| | - Radha Madhyastha
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| | - Harishkumar Madhyastha
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| | - Bethasiwi Purbasari
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| | - Masugi Maruyama
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| | - Yuichi Nakajima
- Faculty of Medicine, Department of Applied Physiology, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
23
|
Yan C, Lv H, Peng Z, Yang D, Shen P, Yu J, Tong C, Wang X. Analysis of miRNA expression changes in bovine endometrial stromal cells treated with lipopolysaccharide. Theriogenology 2021; 167:85-93. [PMID: 33784501 DOI: 10.1016/j.theriogenology.2021.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022]
Abstract
After parturition, bovine uterine stromal cells are often exposed to complex bacterial and viral stimuli owing to epithelial cell rupture, resulting in an inflammatory response. In this study, we used an in vitro model to study the response of bovine endometrial stromal cells to inflammatory mediators and the associated regulated microRNAs in response to lipopolysaccharide. Lipopolysaccharide (LPS) is a bacterial wall component in gram-negative bacteria that causes inflammation upon immune recognition, which is used to create in vitro inflammation models. Thus, we used high-throughput RNA sequencing to identify miRNAs that may have an anti-inflammatory role in the LPS-induced inflammatory response. Two groups of bovine uterine cells were treated with phosphate buffer saline (PBS) and LPS, respectively. Compared with the control (PBS) group, the LPS-treated group had 219 differentially expressed miRNAs, of which 113 were upregulated, and 106 were downregulated. Gene ontology enrichment analysis revealed that the target genes of differentially expressed miRNAs were significantly enriched in several activities, such as transferase activity, small molecule binding, and protein binding. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that the target genes of differential miRNAs were significantly enriched in fluid shear stress and atherosclerosis, MAPK signaling pathway, TNF signaling pathway. By analyzing differentially expressed miRNAs, we found that miR-200c, miR-1247-3p, and let-7b are directly related to the inflammatory response. For instance, miR-200c target genes (MAP3K1, MAP4K3, MAPKAPK5, MAP3K8, MAP3K5) and let-7b target genes (CASP3, IL13, MAPK8, CXCL10) were significantly enriched in the MAPK and IL-17 signaling pathways, respectively. In summary, our research provides insight into the molecular mechanism underlying LPS-induced inflammation in vitro, which may unveil new targets for the treatment of endometritis.
Collapse
Affiliation(s)
- Chenbo Yan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Haimiao Lv
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Zhan Peng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Dexin Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Puxiu Shen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Jingcheng Yu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China
| | - Chao Tong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China; Wuhu Overseas Students Pioneer Park, Wuhu, 241006, PR China.
| | - Xinzhuang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
24
|
Siddiqui S, Johansson K, Joo A, Bonser LR, Koh KD, Le Tonqueze O, Bolourchi S, Bautista RA, Zlock L, Roth TL, Marson A, Bhakta NR, Ansel KM, Finkbeiner WE, Erle DJ, Woodruff PG. Epithelial miR-141 regulates IL-13-induced airway mucus production. JCI Insight 2021; 6:139019. [PMID: 33682796 PMCID: PMC8021117 DOI: 10.1172/jci.insight.139019] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
IL-13-induced goblet cell metaplasia contributes to airway remodeling and pathological mucus hypersecretion in asthma. miRNAs are potent modulators of cellular responses, but their role in mucus regulation is largely unexplored. We hypothesized that airway epithelial miRNAs play roles in IL-13-induced mucus regulation. miR-141 is highly expressed in human and mouse airway epithelium, is altered in bronchial brushings from asthmatic subjects at baseline, and is induced shortly after airway allergen exposure. We established a CRISPR/Cas9-based protocol to target miR-141 in primary human bronchial epithelial cells that were differentiated at air-liquid-interface, and goblet cell hyperplasia was induced by IL-13 stimulation. miR-141 disruption resulted in decreased goblet cell frequency, intracellular MUC5AC, and total secreted mucus. These effects correlated with a reduction in a goblet cell gene expression signature and enrichment of a basal cell gene expression signature defined by single cell RNA sequencing. Furthermore, intranasal administration of a sequence-specific mmu-miR-141-3p inhibitor in mice decreased Aspergillus-induced secreted mucus and mucus-producing cells in the lung and reduced airway hyperresponsiveness without affecting cellular inflammation. In conclusion, we have identified a miRNA that regulates pathological airway mucus production and is amenable to therapeutic manipulation through an inhaled route.
Collapse
Affiliation(s)
- Sana Siddiqui
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | - Kristina Johansson
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
- Department of Microbiology and Immunology
| | - Alex Joo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | | | - Kyung Duk Koh
- Lung Biology Center
- Cardiovascular Research Institute
| | | | - Samaneh Bolourchi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | - Rodriel A. Bautista
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | | | - Theodore L. Roth
- Department of Microbiology and Immunology
- Biomedical Sciences Graduate Program, and
- Diabetes Center, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, California, USA
| | - Alexander Marson
- Department of Microbiology and Immunology
- Innovative Genomics Institute, University of California, Berkeley, California, USA
- J. David Gladstone Institutes, San Francisco, California, USA
- Department of Medicine, Division of Infectious Diseases, UCSF, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Nirav R. Bhakta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center
- Department of Microbiology and Immunology
| | | | - David J. Erle
- Lung Biology Center
- Cardiovascular Research Institute
| | - Prescott G. Woodruff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
- Cardiovascular Research Institute
| |
Collapse
|
25
|
Yanagihara T, Sato S, Upagupta C, Kolb M. What have we learned from basic science studies on idiopathic pulmonary fibrosis? Eur Respir Rev 2019; 28:28/153/190029. [PMID: 31511255 PMCID: PMC9488501 DOI: 10.1183/16000617.0029-2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/11/2019] [Indexed: 12/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal age-related lung disease characterised by progressive and irreversible scarring of the lung. Although the details are not fully understood, there has been tremendous progress in understanding the pathogenesis of idiopathic pulmonary fibrosis, which has led to the identification of many new potential therapeutic targets. In this review we discuss several of these advances with a focus on genetic susceptibility and cellular senescence primarily affecting epithelial cells, activation of profibrotic pathways, disease-enhancing fibrogenic cell types and the role of the remodelled extracellular matrix. This review provides a summary of the most important findings in basic science investigations in pulmonary fibrosis and how they affect drug development and future patient management.http://bit.ly/2RjGMFZ
Collapse
Affiliation(s)
- Toyoshi Yanagihara
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Dept of Medicine, McMaster University, Hamilton, ON, Canada.,Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Both authors contributed equally
| | - Seidai Sato
- Dept of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Both authors contributed equally
| | - Chandak Upagupta
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Dept of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
26
|
Jiang Y, Gao Q, Wang LY, Ma T, Zhu FL, Wang Q, Gao F, Guo C, Zhang LN. Deficiency of programmed cell death 4 affects the balance of T cell subsets in hyperlipidemic mice. Mol Immunol 2019; 112:387-393. [DOI: 10.1016/j.molimm.2019.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/10/2019] [Accepted: 06/28/2019] [Indexed: 12/18/2022]
|
27
|
Jiang C, Guo Y, Yu H, Lu S, Meng L. Pleiotropic microRNA-21 in pulmonary remodeling: novel insights for molecular mechanism and present advancements. Allergy Asthma Clin Immunol 2019; 15:33. [PMID: 31139230 PMCID: PMC6528201 DOI: 10.1186/s13223-019-0345-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/08/2019] [Indexed: 12/29/2022] Open
Abstract
MicroRNA-21 (miR-21), probably one of the most studied miRNAs to date, is found pleiotropic in various biological events. Its emerging role in pulmonary remodeling has attracted extensive attention. This review summarizes the genomic information of its primary transcript and various transcriptional regulations on its promoter. In addition, the role of miR-21 in pulmonary remodeling related signaling such as transforming growth factor β (TGF-β), bone morphogenetic protein (BMP), epidermal growth factor receptor (EGFR) and Notch signaling is discussed. Various validated miR-21 target genes participate in controlling of the overactive cell accumulation, smooth muscle contraction, inflammatory stress (trigger for lung epithelium damage), extracellular matrix deposition and hypoxia-induced disorders. Moreover, we focus on its particular implication in events including inflammatory stress-driven epithelium damage, epithelial-to-mesenchymal transition (EMT), transdifferentiation of fibroblasts into myofibroblasts, hypoxia stimuli and ROS response, as well as some other pulmonary remodeling related events such as overactive fibroblast (myofibroblast) accumulation, extracellular matrix deposition, and angiogenesis. Here, we summarize the strong potential of miR-21 in pulmonary remodeling and provide novel clues for further research in this area.
Collapse
Affiliation(s)
- Congshan Jiang
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Yuanxu Guo
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Hongchuan Yu
- Department of Respiratory Medicine, Xi'an Children Hospital, Xi'an, Shaanxi People's Republic of China
| | - Shemin Lu
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Liesu Meng
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| |
Collapse
|
28
|
Gui X, Qiu X, Tian Y, Xie M, Li H, Gao Y, Zhuang Y, Cao M, Ding H, Ding J, Zhang Y, Cai H. Prognostic value of IFN-γ, sCD163, CCL2 and CXCL10 involved in acute exacerbation of idiopathic pulmonary fibrosis. Int Immunopharmacol 2019; 70:208-215. [PMID: 30851700 DOI: 10.1016/j.intimp.2019.02.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/24/2019] [Accepted: 02/24/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) is of concern because of its propensity for rapid deterioration and high mortality. Its aetiology and mechanism are still unclear. The aims of this study were to clarify the pathophysiology differences between AE-IPF and stable IPF (S-IPF) by comparing the serum levels of various cytokines and chemokines in the two groups and to identify those involvement in the occurrence of acute exacerbation and associated with mortality. METHODS The study included 28 patients with AE-IPF, 32 patients with S-IPF, and 18 healthy control subjects. We measured the serum cytokine and chemokine levels in all cases by multiplex assay. Serum levels of cytokines and chemokines were compared between AE-IPF and S-IPF subjects. Logistic regression analysis was applied to identify the ability of these variables to predict acute exacerbation. Kaplan-Meier curves were used to analyse survival and Cox proportional hazard regression was used to identify predictors of survival. RESULTS Levels of several cytokines and chemokines were significantly higher in both patient groups with IPF (with the exception of interleukin-2 [IL-2], chemokine cc-motif ligand 3, and RANTES [regulation upon activation normal T-cell express sequence]) than in healthy controls. Serum IL-1β (p = 0.008) and interferon (IFN)-γ (p = 0.007) levels tended to be higher in patients with AE-IPF than in those with S-IPF. The concentration of chemokine cc-motif ligand (CCL) 2 was significantly higher in bronchoalveolar lavage fluid than in serum (p = 0.001). Higher C-reactive protein, lactate dehydrogenase, percent forced vital capacity, percent diffusing capacity of the lung for carbon monoxide, and IFN-γ values in the patients with IPF were correlated with acute exacerbation status, with respective odds ratios of 1.241 (p = 0.011), 1.050 (p = 0.004), 1.043 (p = 0.001), 0.927 (p = 0.014), and 0.929 (p = 0.020). Acute exacerbation status was associated with an increased risk of mortality (hazard ratio 0.107, 95% confidence interval 0.036-0.314; p < 0.001). Univariate Cox regression demonstrated an association of IFN-γ, CCL2, C-X-C motif chemokine 10 (CXCL10) and sCD163 levels with an increased mortality risk (p = 0.015, p = 0.002, p = 0.001, and p = 0.030, respectively). CONCLUSIONS Our data demonstrate that serum levels of some pro-inflammatory cytokines and macrophage chemokines are upregulated during acute exacerbations of IPF and that these exacerbations are associated with the serum IFN-γ level. Chemokines and protein such as sCD163, CCL2, and CXCL10 are associated with activation of macrophages and may have a serious impact on overall survival in patients with IPF.
Collapse
Affiliation(s)
- Xianhua Gui
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Xiaohua Qiu
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Yaqiong Tian
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Miaomiao Xie
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Hui Li
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Yujuan Gao
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Yi Zhuang
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Mengshu Cao
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China
| | - Hui Ding
- Department of Respiratory Medicine, Yixing People Hospital, Affiliated Jiangsu University, No. 75 Tongzhenguan Road, Yixing 214200, Jiangsu, PR China
| | - Jingjing Ding
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China.
| | - Yingwei Zhang
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China.
| | - Hourong Cai
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China.
| |
Collapse
|
29
|
Zhang L, Wang Y, Wu G, Xiong W, Gu W, Wang CY. Macrophages: friend or foe in idiopathic pulmonary fibrosis? Respir Res 2018; 19:170. [PMID: 30189872 PMCID: PMC6127991 DOI: 10.1186/s12931-018-0864-2] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a prototype of lethal, chronic, progressive interstitial lung disease of unknown etiology. Over the past decade, macrophage has been recognized to play a significant role in IPF pathogenesis. Depending on the local microenvironments, macrophages can be polarized to either classically activated (M1) or alternatively activated (M2) phenotypes. In general, M1 macrophages are responsible for wound healing after alveolar epithelial injury, while M2 macrophages are designated to resolve wound healing processes or terminate inflammatory responses in the lung. IPF is a pathological consequence resulted from altered wound healing in response to persistent lung injury. In this review, we intend to summarize the current state of knowledge regarding the process of macrophage polarization and its mediators in the pathogenesis of pulmonary fibrosis. Our goal is to update the understanding of the mechanisms underlying the initiation and progression of IPF, and by which, we expect to provide help for developing effective therapeutic strategies in clinical settings.
Collapse
Affiliation(s)
- Lei Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Guorao Wu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Weining Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Weikuan Gu
- Departments of Orthopaedic Surgery-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
30
|
Saradna A, Do DC, Kumar S, Fu QL, Gao P. Macrophage polarization and allergic asthma. Transl Res 2018; 191:1-14. [PMID: 29066321 PMCID: PMC5776696 DOI: 10.1016/j.trsl.2017.09.002] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Allergic asthma is associated with airway inflammation and airway hyperresponsiveness. Macrophage polarization has been shown to have a profound impact on asthma pathogenesis. On exposure to local microenvironments, recruited macrophages can be polarized into either classically activated (or M1) or alternatively activated (or M2) phenotypes. Macrophage polarization has been heavily associated with development of asthma. The process of regulation of macrophage polarization involves an intricate interplay between various cytokines, chemokines, transcriptional factors, and immune-regulatory cells. Different signals from the microenvironment are controlled by different receptors on the macrophages to initiate various macrophage polarization pathways. Most importantly, there is an increased attention on the epigenetic changes (eg, microRNAs, DNA methylation, and histone modification) that impact macrophage functional responses and M1/M2 polarization through modulating cellular signaling and signature gene expression. Thus, modulation of macrophage phenotypes through molecular intervention by targeting some of those potential macrophage regulators may have therapeutic potential in the treatment of allergic asthma and other allergic diseases. In this review, we will discuss the origin of macrophages, characterization of macrophages, macrophage polarization in asthma, and the underlying mechanisms regarding allergen-induced macrophage polarization with emphasis on the regulation of epigenetics, which will provide new insights into the therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Arjun Saradna
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Department of Internal Medicine, Maimonides Medical Center, Brooklyn, NY
| | - Danh C Do
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shruthi Kumar
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Bangalore Medical College and Research Institute, Bangalore, India
| | - Qing-Ling Fu
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peisong Gao
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
31
|
Li X, Yang H, Wu S, Meng Q, Sun H, Lu R, Cui J, Zheng Y, Chen W, Zhang R, Aschner M, Chen R. Suppression of PTPN6 exacerbates aluminum oxide nanoparticle-induced COPD-like lesions in mice through activation of STAT pathway. Part Fibre Toxicol 2017; 14:53. [PMID: 29233151 PMCID: PMC5728016 DOI: 10.1186/s12989-017-0234-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023] Open
Abstract
Background Inhaled nanoparticles can deposit in the deep lung where they interact with pulmonary cells. Despite numerous studies on pulmonary nanotoxicity, detailed molecular mechanisms of specific nanomaterial-induced lung injury have yet to be identified. Results Using whole-body dynamic inhalation model, we studied the interactions between aluminum oxide nanoparticles (Al2O3 NPs) and the pulmonary system in vivo. We found that seven-day-exposure to Al2O3 NPs resulted in emphysema and small airway remodeling in murine lungs, accompanied by enhanced inflammation and apoptosis. Al2O3 NPs exposure led to suppression of PTPN6 and phosphorylation of STAT3, culminating in increased expression of the apoptotic marker PDCD4. Rescue of PTPN6 expression or application of a STAT3 inhibitor, effectively protected murine lungs from inflammation and apoptosis, as well as, in part, from the induction of chronic obstructive pulmonary disease (COPD)-like effects. Conclusion In summary, our studies show that inhibition of PTPN6 plays a critical role in Al2O3 NPs-induced COPD-like lesions. Electronic supplementary material The online version of this article (10.1186/s12989-017-0234-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Hongbao Yang
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, China
| | - Shenshen Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Qingtao Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Hao Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Runze Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Jian Cui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, 266021, China
| | - Wen Chen
- Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing, 210009, China. .,Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
32
|
Oligonucleotide Therapy for Obstructive and Restrictive Respiratory Diseases. Molecules 2017; 22:molecules22010139. [PMID: 28106744 PMCID: PMC6155767 DOI: 10.3390/molecules22010139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/05/2017] [Accepted: 01/08/2017] [Indexed: 12/21/2022] Open
Abstract
Inhaled oligonucleotide is an emerging therapeutic modality for various common respiratory diseases, including obstructive airway diseases like asthma and chronic obstructive pulmonary disease (COPD) and restrictive airway diseases like idiopathic pulmonary fibrosis (IPF). The advantage of direct accessibility for oligonucleotide molecules to the lung target sites, bypassing systemic administration, makes this therapeutic approach promising with minimized potential systemic side effects. Asthma, COPD, and IPF are common chronic respiratory diseases, characterized by persistent airway inflammation and dysregulated tissue repair and remodeling, although each individual disease has its unique etiology. Corticosteroids have been widely prescribed for the treatment of asthma, COPD, and IPF. However, the effectiveness of corticosteroids as an anti-inflammatory drug is limited by steroid resistance in severe asthma, the majority of COPD cases, and pulmonary fibrosis. There is an urgent medical need to develop target-specific drugs for the treatment of these respiratory conditions. Oligonucleotide therapies, including antisense oligonucleotide (ASO), small interfering RNA (siRNA), and microRNA (miRNA) are now being evaluated both pre-clinically and clinically as potential therapeutics. The mechanisms of action of ASO and siRNA are highly target mRNA specific, ultimately leading to target protein knockdown. miRNA has both biomarker and therapeutic values, and its knockdown by a miRNA antagonist (antagomir) has a broader but potentially more non-specific biological outcome. This review will compile the current findings of oligonucleotide therapeutic targets, verified in various respiratory disease models and in clinical trials, and evaluate different chemical modification approaches to improve the stability and potency of oligonucleotides for the treatment of respiratory diseases.
Collapse
|
33
|
Wang M, Shibamoto T, Kuda Y, Tanida M, Zhang T, Song J, Mukai K, Kurata Y. The Responses of Pulmonary and Systemic Circulation and Airway to Allergic Mediators in Anesthetized Rats. Biol Pharm Bull 2017; 39:556-63. [PMID: 27040628 DOI: 10.1248/bpb.b15-00864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung allergic diseases sometimes accompany pulmonary vaso- and broncho-constriction. Rats are currently used for the experimental study of lung allergies. However, their hemodynamic mechanisms are not fully understood. Therefore the effects of allergic mediators were determined systematically in vivo in rats in terms of pulmonary vascular resistance (PVR), airway pressure (AWP) and total peripheral resistance (TPR). We directly measured pulmonary arterial pressure, left atrial pressure, systemic arterial pressure, central venous pressure and aortic blood flow to determine PVR and TPR, as well as AWP, following injections of platelet-activating factor (PAF), histamine, serotonin, leukotriene (LT) C4, and prostaglandin (PG) D2 in anesthetized open-chest artificially ventilated Sprague-Dawley (SD) rats. PVR was dose-dependently increased by consecutive administration of PAF, LTC4, and PGD2, with the maximal responsiveness being PAF>LTC4>PGD2. However, neither histamine nor serotonin changed PVR. TPR was decreased by all agents except LTC4 which actually increased it. PAF and serotonin, but not the other agents, increased AWP. In conclusion, allergic mediators exert non-uniform actions on pulmonary and systemic circulation and airways in anesthetized SD rats: PAF, LTC4 and PGD2, but not histamine or serotonin, caused substantial pulmonary vasoconstriction; LTC4 yielded systemic vasoconstriction, while the others caused systemic vasodilatation; only two mediators, PAF and serotonin, induce airway constriction.
Collapse
Affiliation(s)
- Mofei Wang
- Department of Physiology II, Kanazawa Medical University
| | | | | | | | | | | | | | | |
Collapse
|
34
|
miR-155 Regulated Inflammation Response by the SOCS1-STAT3-PDCD4 Axis in Atherogenesis. Mediators Inflamm 2016; 2016:8060182. [PMID: 27843203 PMCID: PMC5098093 DOI: 10.1155/2016/8060182] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/01/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
Inflammation response plays a critical role in all phases of atherosclerosis (AS). Increased evidence has demonstrated that miR-155 mediates inflammatory mediators in macrophages to promote plaque formation and rupture. However, the precise mechanism of miR-155 remains unclear in AS. Here, we also found that miR-155 and PDCD4 were elevated in the aortic tissue of atherosclerotic mice and ox-LDL treated RAW264.7 cells. Further studies showed that miR-155 not only directly inhibited SOCS1 expression, but also increased the expression of p-STAT and PDCD4, as well as the production of proinflammation mediators IL-6 and TNF-α. Downregulation of miR-155 and PDCD4 and upregulation of SOCS1 obviously decreased the IL-6 and TNF-α expression. In addition, inhibition of miR-155 levels in atherosclerotic mice could notably reduce the IL-6 and TNF-α level in plasma and aortic tissue, accompanied with increased p-STAT3 and PDCD4 and decreased SOCS1. Thus, miR-155 might mediate the inflammation in AS via the SOCS1-STAT3-PDCD4 axis. These results provide a rationale for intervention of intracellular miR-155 as possible antiatherosclerotic targets.
Collapse
|
35
|
Abstract
Lung macrophages link innate and adaptive immune responses during allergic airway inflammatory responses. Alveolar macrophages (AMs) and interstitial macrophages are two different phenotypes that differentially exert immunological function under physiological and pathological conditions. Exposure to pathogen induces polarization of AM cells into classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells). M1 cells dominantly express proinflammatory cytokines such as TNF-α and IL-1 β and induce lung inflammation and tissue damage. M2 cells are further divided into M2a and M2c subsets. M2a cells dominantly produce allergic cytokines IL-4 and IL-13, but M2c cells dominantly produce anti-inflammatory cytokine IL-10. M2a and M2c cells are differently involved in initiation, inflammation resolution, and tissue remodeling in the different stages of asthma. Microenvironment dynamically influences polarization of AM cells. Cytokines, chemokines, and immune-regulatory cells interplay and affect the balance between the polarization of M1 and M2 cells, subsequently influencing disease progression. Thus, modulation of AM phenotypes through molecular intervention has therapeutic potential in the treatment of asthma and other allergic inflammatory diseases. This review updated recent advances in polarization and functional specialization of these macrophage subtypes with emphasis on modulation of polarization of M2 cells in asthma of human subjects and animal models.
Collapse
Affiliation(s)
- Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
36
|
PDCD4 Deficiency Aggravated Colitis and Colitis-associated Colorectal Cancer Via Promoting IL-6/STAT3 Pathway in Mice. Inflamm Bowel Dis 2016; 22:1107-18. [PMID: 26891257 DOI: 10.1097/mib.0000000000000729] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although programmed cell death (PDCD) 4 is generally considered to be a new tumor suppressor, the consequence of Pdcd4 deficiency in tumorigenesis is not well established. The role of PDCD4 in colitis-associated colorectal carcinoma (CRC) remains unknown. METHODS Experimental colitis and CRC were induced by dextran sodium sulfate and dextran sodium sulfate with azoxymethane, respectively, in wild type and Pdcd4 knockout (Pdcd4(-/-)) mice and were evaluated by clinical examination and histopathology. Levels of cytokines were detected by enzyme-linked immunosorbent assay. Changes in signaling pathways were examined by Western blot and immunofluorescent staining. Cell proliferation was determined by BrdU incorporation and Cell Counting Kit-8 staining. RESULTS Pdcd4 deficiency not only aggravated the dextran sodium sulfate-induced acute colitis but also promoted the development of colitis-induced CRC. Mechanically, Pdcd4 deficiency accelerated epithelial cell proliferation during tumorigenesis, markedly up-regulated the expression of proinflammatory cytokines, such as interleukin (IL)-6, and enhanced the activation of signal transducer and activator of transcription (STAT3), a IL-6 downstream effector. Using purified cells, we found that Pdcd4 deficiency increased IL-6 expression in vitro and the susceptibility to IL-6/STAT3 pathway-mediated cell proliferation significantly. Furthermore, blockade of IL-6/STAT3 pathway through sgp130Fc reversed the promoting effect of Pdcd4 deficiency on colonic epithelial cell proliferation in vivo. CONCLUSION The Pdcd4 deficiency accelerates colitis and colitis-associated CRC presumably through up-regulating IL-6/STAT3 pathway, suggesting that PDCD4 plays a protective role in inflammation-associated carcinoma and might be a potential target for the treatment of CRC.
Collapse
|
37
|
Han M, Chung Y, Young Hong J, Rajput C, Lei J, Hinde JL, Chen Q, Weng SP, Bentley JK, Hershenson MB. Toll-like receptor 2-expressing macrophages are required and sufficient for rhinovirus-induced airway inflammation. J Allergy Clin Immunol 2016; 138:1619-1630. [PMID: 27084403 DOI: 10.1016/j.jaci.2016.01.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 12/29/2015] [Accepted: 01/22/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND We have shown that rhinovirus, a cause of asthma exacerbation, colocalizes with CD68+ and CD11b+ airway macrophages after experimental infection in human subjects. We have also shown that rhinovirus-induced cytokine expression is abolished in Toll-like receptor (TLR2)-/- bone marrow-derived macrophages. OBJECTIVE We hypothesize that TLR2+ macrophages are required and sufficient for rhinovirus-induced airway inflammation in vivo. METHODS Naive and ovalbumin (OVA)-sensitized and challenged C57BL/6 wild-type and TLR2-/- mice were infected with RV1B, followed by IgG or anti-TLR2, to determine the requirement and sufficiency of TLR2 for rhinovirus-induced airway responses. Bone marrow chimera experiments using OVA-treated C57BL/6 and TLR2-/- mice were also performed. Finally, naive TLR2-/- mice underwent intranasal transfer of bone marrow-derived wild-type macrophages. RESULTS RV1B infection of naive wild-type mice induced an influx of airway neutrophils and CD11b+ exudative macrophages, which was reduced in TLR2-/- mice. After allergen exposure, rhinovirus-induced neutrophilic and eosinophilic airway inflammation and hyperresponsiveness were reduced in TLR2-/- and anti-TLR2-treated mice. Transfer of TLR2-/- bone marrow into wild-type, OVA-treated C57BL/6 mice blocked rhinovirus-induced airway responses, whereas transfer of wild-type marrow to TLR2-/- mice restored them. Finally, transfer of wild-type macrophages to naive TLR2-/- mice was sufficient for neutrophilic inflammation after rhinovirus infection, whereas macrophages treated with IL-4 (to induce M2 polarization) were sufficient for eosinophilic inflammation, mucous metaplasia, and airways hyperresponsiveness. CONCLUSIONS TLR2 is required for early inflammatory responses induced by rhinovirus, and TLR2+ macrophages are sufficient to confer airway inflammation to TLR2-/- mice, with the pattern of inflammation depending on the macrophage activation state.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Yutein Chung
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Jun Young Hong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Mich
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Jing Lei
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Joanna L Hinde
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Qiang Chen
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Steven P Weng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - J Kelley Bentley
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Mich; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Mich.
| |
Collapse
|
38
|
Jiang Y, Gao Q, Wang L, Guo C, Zhu F, Wang B, Wang Q, Gao F, Chen Y, Zhang L. Deficiency of programmed cell death 4 results in increased IL-10 expression by macrophages and thereby attenuates atherosclerosis in hyperlipidemic mice. Cell Mol Immunol 2015; 13:524-34. [PMID: 26166769 DOI: 10.1038/cmi.2015.47] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 05/09/2015] [Accepted: 05/09/2015] [Indexed: 01/19/2023] Open
Abstract
Programmed cell death 4 (Pdcd4) is a newly defined inhibitor of transcription and translation and a tumor suppressor. Recent studies have suggested that Pdcd4 may also be involved in some inflammatory diseases. However, its role in atherosclerosis, a chronic inflammation of the arterial wall, remains to be investigated. Here, we found that Pdcd4 deficiency in mice increased the expression of IL-10 in macrophages and decreased the expression of IL-17 in T cells in the presence of an atherosclerosis-associated stimulator in vitro and in high fat-induced atherosclerotic plaques. Importantly, knocking out Pdcd4 led to a decrease in atherosclerotic lesions in Apoe(-/-) mice fed a high fat diet. This effect could be partly reversed by blocking IL-10 with a neutralizing antibody but not by the application of exogenous IL-17. Further mechanistic studies revealed that Pdcd4 negatively regulated the expression of IL-10 in an ERK1/2- and p38-dependent manner. These results demonstrate that Pdcd4 deficiency attenuates atherosclerosis in hyperlipidemic mice in part through the upregulation of the anti-inflammatory cytokine IL-10. This indicates that endogenous Pdcd4 promotes atherosclerosis and therefore represents a potential therapeutic target for patients with atherosclerosis.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China.,Department of Hematology, the Second Hospital of Shandong University, Jinan, Shandong, China
| | - Qi Gao
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China.,Department of Clinical Laboratory, Provincial Hospital affiliated with Shandong University, Jinan, Shandong, China
| | - Liyang Wang
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Chun Guo
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Faliang Zhu
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Bo Wang
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Qun Wang
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Fei Gao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Youhai Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Lining Zhang
- Department of Immunology, School of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|