101
|
Arakawa S, Suzukawa M, Watanabe K, Kobayashi K, Matsui H, Nagai H, Nagase T, Ohta K. Secretory immunoglobulin A induces human lung fibroblasts to produce inflammatory cytokines and undergo activation. Clin Exp Immunol 2019; 195:287-301. [PMID: 30570135 DOI: 10.1111/cei.13253] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Immunoglobulin (Ig)A is the most abundant immunoglobulin in humans, and in the airway mucosa secretory IgA (sIgA) plays a pivotal role in first-line defense against invading pathogens and antigens. IgA has been reported to also have pathogenic effects, including possible worsening of the prognosis of idiopathic pulmonary fibrosis (IPF). However, the precise effects of IgA on lung fibroblasts remain unclear, and we aimed to elucidate how IgA activates human lung fibroblasts. We found that sIgA, but not monomeric IgA (mIgA), induced interleukin (IL)-6, IL-8, monocyte chemoattractant protein (MCP)-1 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production by normal human lung fibroblasts (NHLFs) at both the protein and mRNA levels. sIgA also promoted proliferation of NHLFs and collagen gel contraction comparable to with transforming growth factor (TGF)-β, which is involved in fibrogenesis in IPF. Also, Western blot analysis and real-time quantitative polymerase chain reaction (PCR) revealed that sIgA enhanced production of α-smooth muscle actin (α-SMA) and collagen type I (Col I) by NHLFs. Flow cytometry showed that NHLFs bound sIgA, and among the known IgA receptors, NHLFs significantly expressed CD71 (transferrin receptor). Transfection of siRNA targeting CD71 partially but significantly suppressed cytokine production by NHLFs co-cultured with sIgA. Our findings suggest that sIgA may promote human lung inflammation and fibrosis by enhancing production of inflammatory or fibrogenic cytokines as well as extracellular matrix, inducing fibroblast differentiation into myofibroblasts and promoting human lung fibroblast proliferation. sIgA's enhancement of cytokine production may be due partially to its binding to CD71 or the secretory component.
Collapse
Affiliation(s)
- S Arakawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan.,Department of Respiratory Medicine, University of Tokyo, Tokyo, Japan
| | - M Suzukawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - K Watanabe
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan.,Department of Respiratory Medicine, University of Tokyo, Tokyo, Japan
| | - K Kobayashi
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan.,Department of Respiratory Medicine, University of Tokyo, Tokyo, Japan
| | - H Matsui
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - H Nagai
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - T Nagase
- Department of Respiratory Medicine, University of Tokyo, Tokyo, Japan
| | - K Ohta
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| |
Collapse
|
102
|
Hallstrand TS, Leuppi JD, Joos G, Hall GL, Carlsen KH, Kaminsky DA, Coates AL, Cockcroft DW, Culver BH, Diamant Z, Gauvreau GM, Horvath I, de Jongh FHC, Laube BL, Sterk PJ, Wanger J. ERS technical standard on bronchial challenge testing: pathophysiology and methodology of indirect airway challenge testing. Eur Respir J 2018; 52:13993003.01033-2018. [PMID: 30361249 DOI: 10.1183/13993003.01033-2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/20/2018] [Indexed: 12/20/2022]
Abstract
Recently, this international task force reported the general considerations for bronchial challenge testing and the performance of the methacholine challenge test, a "direct" airway challenge test. Here, the task force provides an updated description of the pathophysiology and the methods to conduct indirect challenge tests. Because indirect challenge tests trigger airway narrowing through the activation of endogenous pathways that are involved in asthma, indirect challenge tests tend to be specific for asthma and reveal much about the biology of asthma, but may be less sensitive than direct tests for the detection of airway hyperresponsiveness. We provide recommendations for the conduct and interpretation of hyperpnoea challenge tests such as dry air exercise challenge and eucapnic voluntary hyperpnoea that provide a single strong stimulus for airway narrowing. This technical standard expands the recommendations to additional indirect tests such as hypertonic saline, mannitol and adenosine challenge that are incremental tests, but still retain characteristics of other indirect challenges. Assessment of airway hyperresponsiveness, with direct and indirect tests, are valuable tools to understand and to monitor airway function and to characterise the underlying asthma phenotype to guide therapy. The tests should be interpreted within the context of the clinical features of asthma.
Collapse
Affiliation(s)
- Teal S Hallstrand
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Joerg D Leuppi
- University Clinic of Medicine, Cantonal Hospital Baselland, Liestal, and Medical Faculty University of Basel, Basel, Switzerland
| | - Guy Joos
- Dept of Respiratory Medicine, University of Ghent, Ghent, Belgium
| | - Graham L Hall
- Children's Lung Health, Telethon Kids Institute, School of Physiotherapy and Exercise Science, Curtin University, and Centre for Child Health Research University of Western Australia, Perth, Australia
| | - Kai-Håkon Carlsen
- University of Oslo, Institute of Clinical Medicine, and Oslo University Hospital, Division of Child and Adolescent Medicine, Oslo, Norway
| | - David A Kaminsky
- Pulmonary and Critical Care, University of Vermont College of Medicine, Burlington, VT, USA
| | - Allan L Coates
- Division of Respiratory Medicine, Translational Medicine, Research Institute-Hospital for Sick Children, University of Toronto, ON, Canada
| | - Donald W Cockcroft
- Division of Respirology, Critical Care and Sleep Medicine, Royal University Hospital, Saskatoon, SK, Canada
| | - Bruce H Culver
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Zuzana Diamant
- Dept of Clinical Pharmacy and Pharmacology and QPS-Netherlands, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.,Dept of Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Gail M Gauvreau
- Division of Respirology, Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ildiko Horvath
- Dept of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Frans H C de Jongh
- Dept of Pulmonary Medicine, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Beth L Laube
- Division of Pediatric Pulmonology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter J Sterk
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Jack Wanger
- Pulmonary Function Testing and Clinical Trials Consultant, Rochester, MN, USA
| | | |
Collapse
|
103
|
Debeuf N, Lambrecht BN. Eicosanoid Control Over Antigen Presenting Cells in Asthma. Front Immunol 2018; 9:2006. [PMID: 30233591 PMCID: PMC6131302 DOI: 10.3389/fimmu.2018.02006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022] Open
Abstract
Asthma is a common lung disease affecting 300 million people worldwide. Allergic asthma is recognized as a prototypical Th2 disorder, orchestrated by an aberrant adaptive CD4+ T helper (Th2/Th17) cell immune response against airborne allergens, that leads to eosinophilic inflammation, reversible bronchoconstriction, and mucus overproduction. Other forms of asthma are controlled by an eosinophil-rich innate ILC2 response driven by epithelial damage, whereas in some patients with more neutrophilia, the disease is driven by Th17 cells. Dendritic cells (DCs) and macrophages are crucial regulators of type 2 immunity in asthma. Numerous lipid mediators including the eicosanoids prostaglandins and leukotrienes influence key functions of these cells, leading to either pro- or anti-inflammatory effects on disease outcome. In this review, we will discuss how eicosanoids affect the functions of DCs and macrophages in the asthmatic lung and how this leads to aberrant T cell differentiation that causes disease.
Collapse
Affiliation(s)
- Nincy Debeuf
- Laboratory of Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
104
|
Snodgrass RG, Zezina E, Namgaladze D, Gupta S, Angioni C, Geisslinger G, Lütjohann D, Brüne B. A Novel Function for 15-Lipoxygenases in Cholesterol Homeostasis and CCL17 Production in Human Macrophages. Front Immunol 2018; 9:1906. [PMID: 30197642 PMCID: PMC6117383 DOI: 10.3389/fimmu.2018.01906] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/01/2018] [Indexed: 01/14/2023] Open
Abstract
Arachidonate 15-lipoxygenase (ALOX15) and arachidonate 15-lipoxygenase, type B (ALOX15B) catalyze the dioxygenation of polyunsaturated fatty acids and are upregulated in human alternatively activated macrophages (AAMs) induced by Th2 cytokine interleukin-4 (IL-4) and/or interleukin-13. Known primarily for roles in bioactive lipid mediator synthesis, 15-lipoxygenases (15-LOXs) have been implicated in various macrophage functions including efferocytosis and ferroptosis. Using a combination of inhibitors and siRNAs to suppress 15-LOX isoforms, we studied the role of 15-LOXs in cellular cholesterol homeostasis and immune function in naïve and AAMs. Silencing or inhibiting the 15-LOX isoforms impaired sterol regulatory element binding protein (SREBP)-2 signaling by inhibiting SREBP-2 processing into mature transcription factor and reduced SREBP-2 binding to sterol regulatory elements and subsequent target gene expression. Silencing ALOX15B reduced cellular cholesterol and the cholesterol intermediates desmosterol, lanosterol, 24,25-dihydrolanosterol, and lathosterol as well as oxysterols in IL-4-stimulated macrophages. In addition, attenuating both 15-LOX isoforms did not generally affect IL-4 gene expression but rather uniquely impacted IL-4-induced CCL17 production in an SREBP-2-dependent manner resulting in reduced T cell migration to macrophage conditioned media. In conclusion, we identified a novel role for ALOX15B, and to a lesser extent ALOX15, in cholesterol homeostasis and CCL17 production in human macrophages.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ekaterina Zezina
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dmitry Namgaladze
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Sahil Gupta
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Carlo Angioni
- ZAFES/Pharmazentrum Frankfurt, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- ZAFES/Pharmazentrum Frankfurt, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Frankfurt, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
105
|
Song YD, Li XZ, Wu YX, Shen Y, Liu FF, Gao PP, Sun L, Qian F. Emodin alleviates alternatively activated macrophage and asthmatic airway inflammation in a murine asthma model. Acta Pharmacol Sin 2018; 39:1317-1325. [PMID: 29417945 DOI: 10.1038/aps.2017.147] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
Alternatively activated macrophages (AAMs) are not only associated with asthma but also lead to asthmatic airway inflammation and remodeling. Inhibition of AAMs is an alternative therapeutic strategy for treating asthma. In this study we investigated whether emodin (1,3,8-trihydroxy-6-methylanthraquinone), isolated from the rhizome of Rheum palmatum, alleviated asthmatic airway inflammation and reduced AAM polarization in a murine asthma model. Mice were sensitized with a triple allergen mix containing dust mite, ragweed and aspergillus (DRA). In mice with DRA-induced asthma, asthmatic inflammation was significantly enhanced. Intraperitoneal injection of emodin (20 mg·kg-1·d-1, ip) 1 h prior to DRA challenge on days 12-14 significantly decreased pulmonary eosinophil and lymphocyte infiltration, mucus secretion and serum IgE production, as well as IL-4 and IL-5 production in bronchoalveolar lavage fluid. In response to emodin treatment, activated markers of AAM Ym-1, Fizz-1 and arginase-1 in the lung tissues were remarkably decreased. In mouse bone marrow-derived macrophages (BMDMs) in vitro, emodin (2-50 μmol/L) dose-dependently inhibited IL-4-induced AAM polarization and STAT6 phosphorylation. Collectively, our results suggest that emodin effectively ameliorates asthmatic airway inflammation and AAM polarization, and it may therefore become a potential agent for the treatment of asthma.
Collapse
|
106
|
Sun B, Zhu L, Tao Y, Sun HX, Li Y, Wang P, Hou Y, Zhao Y, Zhang X, Zhang L, Na N, Zhao Y. Characterization and allergic role of IL-33-induced neutrophil polarization. Cell Mol Immunol 2018; 15:782-793. [PMID: 29503441 PMCID: PMC6141612 DOI: 10.1038/cmi.2017.163] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 01/09/2023] Open
Abstract
Neutrophils are involved in the pathogenesis of allergy. However, the contribution of the different functionally polarized neutrophils in allergy needs to be clarified. We sought to define the characteristics of interleukin (IL)-33-induced neutrophils and the involvement of this subset of polarized neutrophils in allergic pathogenesis. Freshly isolated neutrophils were treated with different cytokines and the cytokine expression levels were detected by real-time PCR. The gene expression profile of IL-33-induced neutrophils was determined by microarray assay. Adoptive transfer assay was used to investigate the function of IL-33-induced neutrophils in an ovalbumin (OVA)-induced allergic asthma model. IL-33-treated neutrophils selectively produced IL-4, IL-5, IL-9 and IL-13 (referred as to N(IL-33) cells) and displayed a distinctive gene expression profile in sharp contrast to resting and lipopolysaccharide (LPS)-treated neutrophils. IL-33-induced neutrophils expressed high Levels of IL-1R2 on cell surface, whereas resting and LPS-treated neutrophils did not, indicating IL-1R2 might be used as a biomarker for N(IL-33) cells. Importantly, N(IL-33) neutrophils exist in the lungs of OVA-induced allergic asthma mice. Adoptive transfer of N(IL-33) neutrophils significantly promotes the severity of the lung pathogenesis in this model. IL-33 induces neutrophil polarization through c-Jun N-terminal kinase- and nuclear factor-κB-dependent pathways. A previously unappreciated neutrophil polarization driven by IL-33 with unique cell surface markers and cytokine/chemokine-producing gene profile was defined. The newly identified N(IL-33) subpopulation may have significant contribution to IL-33-related pathogenesis.
Collapse
Affiliation(s)
- Bo Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Linnan Zhu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yaling Tao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Hai-Xi Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yang Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Peng Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yuzhu Hou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100101, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, 510630, Guangzhou, Guangdong, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
107
|
Allard B, Panariti A, Martin JG. Alveolar Macrophages in the Resolution of Inflammation, Tissue Repair, and Tolerance to Infection. Front Immunol 2018; 9:1777. [PMID: 30108592 PMCID: PMC6079255 DOI: 10.3389/fimmu.2018.01777] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Pathogen persistence in the respiratory tract is an important preoccupation, and of particular relevance to infectious diseases such as tuberculosis. The equilibrium between elimination of pathogens and the magnitude of the host response is a sword of Damocles for susceptible patients. The alveolar macrophage is the first sentinel of the respiratory tree and constitutes the dominant immune cell in the steady state. This immune cell is a key player in the balance between defense against pathogens and tolerance toward innocuous stimuli. This review focuses on the role of alveolar macrophages in limiting lung tissue damage from potentially innocuous stimuli and from infections, processes that are relevant to appropriate tolerance of potential causes of lung disease. Notably, the different anti-inflammatory strategies employed by alveolar macrophages and lung tissue damage control are explored. These two properties, in addition to macrophage manipulation by pathogens, are discussed to explain how alveolar macrophages may drive pathogen persistence in the airways.
Collapse
Affiliation(s)
- Benoit Allard
- Department of Medicine, Meakins Christie Laboratories, Research Institute McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Alice Panariti
- Department of Medicine, Meakins Christie Laboratories, Research Institute McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - James G Martin
- Department of Medicine, Meakins Christie Laboratories, Research Institute McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
108
|
Moon HG, Kim SJ, Jeong JJ, Han SS, Jarjour NN, Lee H, Abboud-Werner SL, Chung S, Choi HS, Natarajan V, Ackerman SJ, Christman JW, Park GY. Airway Epithelial Cell-Derived Colony Stimulating Factor-1 Promotes Allergen Sensitization. Immunity 2018; 49:275-287.e5. [PMID: 30054206 DOI: 10.1016/j.immuni.2018.06.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/25/2018] [Accepted: 06/21/2018] [Indexed: 10/28/2022]
Abstract
Airway epithelial cells (AECs) secrete innate immune cytokines that regulate adaptive immune effector cells. In allergen-sensitized humans and mice, the airway and alveolar microenvironment is enriched with colony stimulating factor-1 (CSF1) in response to allergen exposure. In this study we found that AEC-derived CSF1 had a critical role in the production of allergen reactive-IgE production. Furthermore, spatiotemporally secreted CSF1 regulated the recruitment of alveolar dendritic cells (DCs) and enhanced the migration of conventional DC2s (cDC2s) to the draining lymph node in an interferon regulatory factor 4 (IRF4)-dependent manner. CSF1 selectively upregulated the expression of the chemokine receptor CCR7 on the CSF1R+ cDC2, but not the cDC1, population in response to allergen stimuli. Our data describe the functional specification of CSF1-dependent DC subsets that link the innate and adaptive immune responses in T helper 2 (Th2) cell-mediated allergic lung inflammation.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Seung-Jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jong Jin Jeong
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Seon-Sook Han
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nizar N Jarjour
- Allergy, Pulmonary, and Critical Care Division, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Hyun Lee
- Center for Biomolecular Sciences, and Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Sherry L Abboud-Werner
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sangwoon Chung
- Section of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Center, Columbus, OH, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Viswanathan Natarajan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, and Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - John W Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Center, Columbus, OH, USA
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
109
|
Song Y, Wu Y, Li X, Shen Y, Ding Y, Zhu H, Liu F, Yu K, Sun L, Qian F. Protostemonine attenuates alternatively activated macrophage and DRA-induced asthmatic inflammation. Biochem Pharmacol 2018; 155:198-206. [PMID: 29991449 DOI: 10.1016/j.bcp.2018.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/01/2018] [Indexed: 12/15/2022]
Abstract
Asthma is one of the most common pulmonary diseases that threatens human life because of lack of effective medicines. Protostemonine (PSN), an active alkaloid extracted from the roots of Stemona sesslifolia, has anti-inflammatory effects on acute lung injury and acute liver failure. However, it has not been defined whether PSN alleviates asthmatic inflammation. Here, we reported that PSN inhibits pulmonary eosinophil infiltration, goblet cell hyperplasia, mucus secretion, IgE and Th2 cytokine (IL-4, IL-5, IL-13 and IL-33) production by using DRA (dust mites, ragweed and aspergillus)-induced murine asthma model. Moreover, PSN also attenuated the expression of Arginase-1 (Arg-1), Ym-1 and Fizz-1, markers of AAM (alternatively activated macrophage) polarization, in lung tissues. In addition, PSN attenuated IL-4-induced expression of Arg-1, Ym-1 and Fizz-1 in bone marrow derived macrophages (BMDMs). Treatment with PSN decreased IL-4-induced STAT6 phosphorylation, KLF4 and IRF4 expression in BMDMs. Collectively, our results indicated that PSN ameliorates AAM polarization and asthmatic inflammation and might be a potential agent for treating asthma.
Collapse
Affiliation(s)
- Yunduan Song
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Yaxian Wu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Xiaozong Li
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Yao Shen
- Department of Respiratory Medicine, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Yunhe Ding
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Hongbo Zhu
- Department of Pathology, Shanghai Pudong Hospital, Fudan University ,Shanghai 201399, PR China
| | - Fangfang Liu
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Kaikai Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Lei Sun
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233003, PR China.
| |
Collapse
|
110
|
Sharma N, Akkoyunlu M, Rabin RL. Macrophages-common culprit in obesity and asthma. Allergy 2018; 73:1196-1205. [PMID: 29178573 DOI: 10.1111/all.13369] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2017] [Indexed: 12/29/2022]
Abstract
Macrophages are essential innate immune cells that also regulate local metabolism. Endogenous or exogenous stimuli may polarize macrophages toward phenotypes that serve distinct innate immunological metabolic functions. IFN-γ or lipopolysaccharide (LPS) polarizes macrophages toward the M1, or "classically activated" phenotype that participates in defense against intracellular pathogens. IL-4, IL-13, or chitin polarizes macrophages toward the M2, or "alternatively activated" phenotype, which defends against multicellular nematodes and fungi. As macrophages polarize in local environments, M1 and M2 macrophages may coexist in different organs and may differentially affect asthma and obesity, two comorbid diseases where polarized macrophages contribute to their pathogenesis. While M1 macrophages are considered beneficial in asthma and contribute to the pathology of obesity, M2 macrophages contribute to the pathology of asthma, but limit metabolic syndrome associated with obesity. Here, we discuss the roles for M1 and M2 macrophages in asthma and obesity, and propose a model by which M1-mediated inflammation in adipose tissue enhances M2-mediated inflammation in the asthmatic lung.
Collapse
Affiliation(s)
- N. Sharma
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - M. Akkoyunlu
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - R. L. Rabin
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| |
Collapse
|
111
|
Rajput C, Walsh MP, Eder BN, Metitiri EE, Popova AP, Hershenson MB. Rhinovirus infection induces distinct transcriptome profiles in polarized human macrophages. Physiol Genomics 2018. [PMID: 29521598 DOI: 10.1152/physiolgenomics.00122.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Infections with rhinovirus (RV) cause asthma exacerbations. Recent studies suggest that macrophages play a role in asthmatic airway inflammation and the innate immune response to RV infection. Macrophages exhibit phenotypes based on surface markers and gene expression. We hypothesized that macrophage polarization state alters gene expression in response to RV infection. Cells were derived from human peripheral blood derived monocytes. M1 and M2 polarization was carried out by using IFN-γ and IL-4, respectively, and RNA was extracted for Affymetrix Human Gene ST2.1 exon arrays. Selected genes were validated by quantitative (q)PCR. Treatment of nonactivated (M0) macrophages with IFN-γ and IL-4 induced the expression of 252 and 153 distinct genes, respectively, including previously-identified M1 and M2 markers. RV infection of M0 macrophages induced upregulation of 232 genes; pathway analysis showed significant overrepresentation of genes involved in IFN-α/β signaling and cytokine signaling in the immune system. RV infection induced differential expression of 195 distinct genes in M1-like macrophages but only seven distinct genes in M2-like-polarized cells. In a secondary analysis, comparison between M0-, RV-infected, and M1-like-polarized, RV-infected macrophages revealed differential expression of 227 genes including those associated with asthma and its exacerbation. qPCR demonstrated increased expression of CCL8, CXCL10, TNFSF10, TNFSF18, IL6, NOD2, and GSDMD and reduced expression of VNN1, AGO1, and AGO2. Together, these data show that, in contrast to M2-like-polarized macrophages, gene expression of M1-like macrophages is highly regulated by RV.
Collapse
Affiliation(s)
- Charu Rajput
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Megan P Walsh
- Department of Epidemiology, University of Michigan School of Public Health , Ann Arbor, MI
| | - Breanna N Eder
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Ediri E Metitiri
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Antonia P Popova
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan
| |
Collapse
|
112
|
Role of Polyamines in Asthma Pathophysiology. Med Sci (Basel) 2018; 6:medsci6010004. [PMID: 29316647 PMCID: PMC5872161 DOI: 10.3390/medsci6010004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/31/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022] Open
Abstract
Asthma is a complex disease of airways, where the interactions of immune and structural cells result in disease outcomes with airway remodeling and airway hyper-responsiveness. Polyamines, which are small-sized, natural super-cations, interact with negatively charged intracellular macromolecules, and altered levels of polyamines and their interactions have been associated with different pathological conditions including asthma. Elevated levels of polyamines have been reported in the circulation of asthmatic patients as well as in the lungs of a murine model of asthma. In various studies, polyamines were found to potentiate the pathogenic potential of inflammatory cells, such as mast cells and granulocytes (eosinophils and neutrophils), by either inducing the release of their pro-inflammatory mediators or prolonging their life span. Additionally, polyamines were crucial in the differentiation and alternative activation of macrophages, which play an important role in asthma pathology. Importantly, polyamines cause airway smooth muscle contraction and thus airway hyper-responsiveness, which is the key feature in asthma pathophysiology. High levels of polyamines in asthma and their active cellular and macromolecular interactions indicate the importance of the polyamine pathway in asthma pathogenesis; therefore, modulation of polyamine levels could be a suitable approach in acute and severe asthma management. This review summarizes the possible roles of polyamines in different pathophysiological features of asthma.
Collapse
|
113
|
Zhou Y, Do DC, Ishmael FT, Squadrito ML, Tang HM, Tang HL, Hsu MH, Qiu L, Li C, Zhang Y, Becker KG, Wan M, Huang SK, Gao P. Mannose receptor modulates macrophage polarization and allergic inflammation through miR-511-3p. J Allergy Clin Immunol 2018; 141:350-364.e8. [PMID: 28629744 PMCID: PMC5944850 DOI: 10.1016/j.jaci.2017.04.049] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 03/10/2017] [Accepted: 04/24/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mannose receptor (MRC1/CD206) has been suggested to mediate allergic sensitization and asthma to multiple glycoallergens, including cockroach allergens. OBJECTIVE We sought to determine the existence of a protective mechanism through which MRC1 limits allergic inflammation through its intronic miR-511-3p. METHODS We examined MRC1-mediated cockroach allergen uptake by lung macrophages and lung inflammation using C57BL/6 wild-type (WT) and Mrc1-/- mice. The role of miR-511-3p in macrophage polarization and cockroach allergen-induced lung inflammation in mice transfected with adeno-associated virus (AAV)-miR-511-3p (AAV-cytomegalovirus-miR-511-3p-enhanced green fluorescent protein) was analyzed. Gene profiling of macrophages with or without miR-511-3p overexpression was also performed. RESULTS Mrc1-/- lung macrophages showed a significant reduction in cockroach allergen uptake compared with WT mice, and Mrc1-/- mice had an exacerbated lung inflammation with increased levels of cockroach allergen-specific IgE and TH2/TH17 cytokines in a cockroach allergen-induced mouse model compared with WT mice. Macrophages from Mrc1-/- mice showed significantly reduced levels of miR-511-3 and an M1 phenotype, whereas overexpression of miR-511-3p rendered macrophages to exhibit a M2 phenotype. Furthermore, mice transfected with AAV-miR-511-3p showed a significant reduction in cockroach allergen-induced inflammation. Profiling of macrophages with or without miR-511-3p overexpression identified 729 differentially expressed genes, wherein expression of prostaglandin D2 synthase (Ptgds) and its product PGD2 were significantly downregulated by miR-511-3p. Ptgds showed a robust binding to miR-511-3p, which might contribute to the protective effect of miR-511-3p. Plasma levels of miR-511-3p were significantly lower in human asthmatic patients compared with nonasthmatic subjects. CONCLUSION These studies support a critical but previously unrecognized role of MRC1 and miR-511-3p in protection against allergen-induced lung inflammation.
Collapse
Affiliation(s)
- Yufeng Zhou
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Md; Children's Hospital and the Institute of Biomedical Sciences and, Fudan University, and Key Laboratory of Neonatal Diseases, Ministry of Health, Shanghai, China
| | - Danh C Do
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Faoud T Ishmael
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pa
| | - Mario Leonardo Squadrito
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Ho Lam Tang
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md
| | - Man-Hsun Hsu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pa
| | - Lipeng Qiu
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Changjun Li
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Yongqing Zhang
- Gene Expression & Genomics Unit, National Institute on Aging, National Institutes of Health, Baltimore, Md
| | - Kevin G Becker
- Gene Expression & Genomics Unit, National Institute on Aging, National Institutes of Health, Baltimore, Md
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shau-Ku Huang
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Md; National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Lou-Hu Hospital, Shen-Zhen University, Shen-Zhen, China.
| | - Peisong Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
114
|
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: 280] [Impact Index Per Article: 40.0] [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
|
115
|
Liu MC, Xiao HQ, Breslin LM, Bochner BS, Schroeder JT. Enhanced antigen presenting and T cell functions during late-phase allergic responses in the lung. Clin Exp Allergy 2017; 48:334-342. [PMID: 29105205 DOI: 10.1111/cea.13054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Allergic inflammation is a common feature of asthma and may contribute to both development and perpetuation of disease. The interaction of antigen-presenting cells (APC) with sensitized helper T lymphocytes (TC) producing Th2 cytokines may determine the inflammatory response. Recruitment of APC and TC to the lung during allergic responses has been demonstrated, but functional studies in humans have been limited. OBJECTIVE This study examined the function of APC and TC accumulating at sites of inflammation after segmental allergen challenge (SAC). METHODS Fifteen allergic patients underwent SAC, and cells from bronchoalveolar lavage (BAL) were collected after 24 hours. APC and TC from the blood and BAL were purified based on expression of the monocyte marker, CD14; the plasmacytoid dendritic cell (pDC) marker, BDCA4, identifying neuropilin-1 (NRP1); and the helper T cell marker, CD4. Functional activity was assessed using allergen-induced T cell proliferation. Flow cytometry identified cells expressing CD14 and NRP1. RESULTS SAC resulted in a 12-fold increase in mononuclear cells having the morphologic appearance of blood monocytes. Most of these cells co-expressed CD14 and NRP1. After saline challenge, BAL mononuclear cells demonstrated little APC function. Following SAC, BAL mononuclear cells showed function equal to pDC from blood and greater than blood monocytes. Purified NRP1+ cells from BAL had even greater function than pDC cells from blood (P = .008). Using consistent sources of APC, enhanced proliferation of TC from lung compared to blood was also demonstrated (P = .002). CONCLUSIONS The marked increase in APC function for allergen-specific TC proliferation during allergic inflammation is largely due to the recruitment of monocytes and dendritic cells. There is also an enhanced response in the lung TC population, consistent with recruitment of allergen-specific T cells. Interactions between recruited APC and TC may occur as an early event promoting allergic airway inflammation.
Collapse
Affiliation(s)
- M C Liu
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - H Q Xiao
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - L M Breslin
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - B S Bochner
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - J T Schroeder
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
116
|
Ishida Y, Kimura A, Nosaka M, Kuninaka Y, Hemmi H, Sasaki I, Kaisho T, Mukaida N, Kondo T. Essential involvement of the CX3CL1-CX3CR1 axis in bleomycin-induced pulmonary fibrosis via regulation of fibrocyte and M2 macrophage migration. Sci Rep 2017; 7:16833. [PMID: 29203799 PMCID: PMC5714949 DOI: 10.1038/s41598-017-17007-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023] Open
Abstract
The potential role of macrophages in pulmonary fibrosis (PF) prompted us to evaluate the roles of CX3CR1, a chemokine receptor abundantly expressed in macrophages during bleomycin (BLM)-induced PF. Intratracheal BLM injection induced infiltration of leukocytes such as macrophages into the lungs, which eventually resulted in fibrosis. CX3CR1 expression was mainly detected in the majority of macrophages and in a small portion of α-smooth muscle actin-positive cells in the lungs, while CX3CL1 was expressed in macrophages. BLM-induced fibrotic changes in the lungs were reduced without any changes in the number of leukocytes in Cx3cr1 -/- mice, as compared with those in the wild-type (WT) mice. However, intrapulmonary CX3CR1+ macrophages displayed pro-fibrotic M2 phenotypes; lack of CX3CR1 skewed their phenotypes toward M1 in BLM-challenged lungs. Moreover, fibrocytes expressed CX3CR1, and were increased in BLM-challenged WT lungs. The number of intrapulmonary fibrocytes was decreased in Cx3cr1 -/- mice. Thus, locally-produced CX3CL1 can promote PF development primarily by attracting CX3CR1-expressing M2 macrophages and fibrocytes into the lungs.
Collapse
Affiliation(s)
- Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Izumi Sasaki
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
117
|
Keselman A, Fang X, White PB, Heller NM. Estrogen Signaling Contributes to Sex Differences in Macrophage Polarization during Asthma. THE JOURNAL OF IMMUNOLOGY 2017; 199:1573-1583. [PMID: 28760880 DOI: 10.4049/jimmunol.1601975] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 06/30/2017] [Indexed: 11/19/2022]
Abstract
Allergic asthma is a chronic Th2 inflammation in the lungs that constricts the airways and presents as coughing and wheezing. Asthma mostly affects boys in childhood and women in adulthood, suggesting that shifts in sex hormones alter the course of the disease. Alveolar macrophages have emerged as major mediators of allergic lung inflammation in animal models as well as humans. Whether sex differences exist in macrophage polarization and the molecular mechanism(s) that drive differential responses are not well understood. We found that IL-4-stimulated bone marrow-derived and alveolar macrophages from female mice exhibited greater expression of M2 genes in vitro and after allergen challenge in vivo. Alveolar macrophages from female mice exhibited greater expression of the IL-4Rα and estrogen receptor (ER) α compared with macrophages from male mice following allergen challenge. An ERα-specific agonist enhanced IL-4-induced M2 gene expression in macrophages from both sexes, but more so in macrophages from female mice. Furthermore, IL-4-stimulated macrophages from female mice exhibited more transcriptionally active histone modifications at M2 gene promoters than did macrophages from male mice. We found that supplementation of estrogen into ovariectomized female mice enhanced M2 polarization in vivo upon challenge with allergen and that macrophage-specific deletion of ERα impaired this M2 polarization. The effects of estrogen are long-lasting; bone marrow-derived macrophages from ovariectomized mice implanted with estrogen exhibited enhanced IL-4-induced M2 gene expression compared with macrophages from placebo-implanted littermates. Taken together, our findings suggest that estrogen enhances IL-4-induced M2 gene expression and thereby contributes to sex differences observed in asthma.
Collapse
Affiliation(s)
- Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Xi Fang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Preston B White
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
118
|
Abstract
The mucosal surfaces of the human body are typically colonized by polymicrobial communities seeded in infancy and are continuously shaped by environmental exposures. These communities interact with the mucosal immune system to maintain homeostasis in health, but perturbations in their composition and function are associated with lower airway diseases, including asthma, a developmental and heterogeneous chronic disease with various degrees and types of airway inflammation. This review will summarize recent studies examining airway microbiota dysbioses associated with asthma and their relationship with the pathophysiology of this disease.
Collapse
|
119
|
Draijer C, Peters-Golden M. Alveolar Macrophages in Allergic Asthma: the Forgotten Cell Awakes. Curr Allergy Asthma Rep 2017; 17:12. [PMID: 28233154 DOI: 10.1007/s11882-017-0681-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The role of alveolar macrophages in innate immune responses has long been appreciated. Here, we review recent studies evaluating the participation of these cells in allergic inflammation. RECENT FINDINGS Immediately after allergen exposure, monocytes are rapidly recruited from the bloodstream and serve to promote acute inflammation. By contrast, resident alveolar macrophages play a predominantly suppressive role in an effort to restore homeostasis. As inflammation becomes established after repeated exposures, alveolar macrophages can polarize across a continuum of activation phenotypes, losing their suppressive functions and gaining pathogenic functions. Future research should focus on the diverse roles of monocytes/macrophages during various types and phases of allergic inflammation. These properties could lead us to new therapeutic opportunities.
Collapse
Affiliation(s)
- Christina Draijer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
120
|
Liu Y, Gunsten SP, Sultan DH, Luehmann HP, Zhao Y, Blackwell TS, Bollermann-Nowlis Z, Pan JH, Byers DE, Atkinson JJ, Kreisel D, Holtzman MJ, Gropler RJ, Combadiere C, Brody SL. PET-based Imaging of Chemokine Receptor 2 in Experimental and Disease-related Lung Inflammation. Radiology 2017; 283:758-768. [PMID: 28045644 PMCID: PMC5452886 DOI: 10.1148/radiol.2016161409] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose To characterize a chemokine receptor type 2 (CCR2)-binding peptide adapted for use as a positron emission tomography (PET) radiotracer for noninvasive detection of lung inflammation in a mouse model of lung injury and in human tissues from subjects with lung disease. Materials and Methods The study was approved by institutional animal and human studies committees. Informed consent was obtained from patients. A 7-amino acid CCR2 binding peptide (extracellular loop 1 inverso [ECL1i]) was conjugated to tetraazacyclododecane tetraacetic acid (DOTA) and labeled with copper 64 (64Cu) or fluorescent dye. Lung inflammation was induced with intratracheal administration of lipopolysaccharide (LPS) in wild-type (n = 19) and CCR2-deficient (n = 4) mice, and these mice were compared with wild-type mice given control saline (n = 5) by using PET performed after intravenous injection of 64Cu-DOTA-ECL1i. Lung immune cells and those binding fluorescently labeled ECL1i in vivo were detected with flow cytometry. Lung inflammation in tissue from subjects with nondiseased lungs donated for lung transplantation (n = 11) and those with chronic obstructive pulmonary disease (COPD) who were undergoing lung transplantation (n = 16) was evaluated for CCR2 with immunostaining and autoradiography (n = 6, COPD) with 64Cu-DOTA-ECL1i. Groups were compared with analysis of variance, the Mann-Whitney U test, or the t test. Results Signal on PET images obtained in mouse lungs after injury with LPS was significantly greater than that in the saline control group (mean = 4.43% of injected dose [ID] per gram of tissue vs 0.99% of injected dose per gram of tissue; P < .001). PET signal was significantly diminished with blocking studies using nonradiolabeled ECL1i in excess (mean = 0.63% ID per gram of tissue; P < .001) and in CCR2-deficient mice (mean = 0.39% ID per gram of tissue; P < .001). The ECL1i signal was associated with an elevated level of mouse lung monocytes. COPD lung tissue displayed significantly elevated CCR2 levels compared with nondiseased tissue (median = 12.8% vs 1.2% cells per sample; P = .002), which was detected with 64Cu-DOTA-ECL1i by using autoradiography. Conclusion 64Cu-DOTA-ECL1i is a promising tool for PET-based detection of CCR2-directed inflammation in an animal model and in human tissues as a step toward clinical translation. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Yongjian Liu
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Sean P. Gunsten
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Deborah H. Sultan
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Hannah P. Luehmann
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Yongfeng Zhao
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - T. Scott Blackwell
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Zachary Bollermann-Nowlis
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Jie-hong Pan
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Derek E. Byers
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Jeffrey J. Atkinson
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Daniel Kreisel
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Michael J. Holtzman
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Robert J. Gropler
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Christophe Combadiere
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Steven L. Brody
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| |
Collapse
|
121
|
Lechner AJ, Driver IH, Lee J, Conroy CM, Nagle A, Locksley RM, Rock JR. Recruited Monocytes and Type 2 Immunity Promote Lung Regeneration following Pneumonectomy. Cell Stem Cell 2017; 21:120-134.e7. [PMID: 28506464 DOI: 10.1016/j.stem.2017.03.024] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 02/09/2017] [Accepted: 03/27/2017] [Indexed: 12/24/2022]
Abstract
To investigate the role of immune cells in lung regeneration, we used a unilateral pneumonectomy model that promotes the formation of new alveoli in the remaining lobes. Immunofluorescence and single-cell RNA sequencing found CD115+ and CCR2+ monocytes and M2-like macrophages accumulating in the lung during the peak of type 2 alveolar epithelial stem cell (AEC2) proliferation. Genetic loss of function in mice and adoptive transfer studies revealed that bone marrow-derived macrophages (BMDMs) traffic to the lung through a CCL2-CCR2 chemokine axis and are required for optimal lung regeneration, along with Il4ra-expressing leukocytes. Our data suggest that these cells modulate AEC2 proliferation and differentiation. Finally, we provide evidence that group 2 innate lymphoid cells are a source of IL-13, which promotes lung regeneration. Together, our data highlight the potential for immunomodulatory therapies to stimulate alveologenesis in adults.
Collapse
Affiliation(s)
- Andrew J Lechner
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Ian H Driver
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Jinwoo Lee
- Department of Medicine and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Carmen M Conroy
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Abigail Nagle
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Richard M Locksley
- Department of Medicine and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Jason R Rock
- Department of Anatomy, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
122
|
FoxO1 regulates allergic asthmatic inflammation through regulating polarization of the macrophage inflammatory phenotype. Oncotarget 2017; 7:17532-46. [PMID: 27007158 PMCID: PMC4951231 DOI: 10.18632/oncotarget.8162] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/01/2016] [Indexed: 01/18/2023] Open
Abstract
Inflammatory monocyte and tissue macrophages influence the initiation, progression, and resolution of type 2 immune responses, and alveolar macrophages are the most prevalent immune-effector cells in the lung. While we were characterizing the M1- or M2-like macrophages in type 2 allergic inflammation, we discovered that FoxO1 is highly expressed in alternatively activated macrophages. Although several studies have been focused on the fundamental role of FoxOs in hematopoietic and immune cells, the exact role that FoxO1 plays in allergic asthmatic inflammation in activated macrophages has not been investigated. Growing evidences indicate that FoxO1 acts as an upstream regulator of IRF4 and could have a role in a specific inflammatory phenotype of macrophages. Therefore, we hypothesized that IRF4 expression regulated by FoxO1 in alveolar macrophages is required for established type 2 immune mediates allergic lung inflammation. Our data indicate that targeted deletion of FoxO1 using FoxO1-selective inhibitor AS1842856 and genetic ablation of FoxO1 in macrophages significantly decreases IRF4 and various M2 macrophage-associated genes, suggesting a mechanism that involves FoxO1-IRF4 signaling in alveolar macrophages that works to polarize macrophages toward established type 2 immune responses. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, macrophage specific FoxO1 overexpression is associated with an accentuation of asthmatic lung inflammation, whereas pharmacologic inhibition of FoxO1 by AS1842856 attenuates the development of asthmatic lung inflammation. Thus, our study identifies a role for FoxO1-IRF4 signaling in the development of alternatively activated alveolar macrophages that contribute to type 2 allergic airway inflammation.
Collapse
|
123
|
Rowe RK, Pyle DM, Tomlinson AR, Lv T, Hu Z, Gill MA. IgE cross-linking impairs monocyte antiviral responses and inhibits influenza-driven T H1 differentiation. J Allergy Clin Immunol 2017; 140:294-298.e8. [PMID: 28087327 DOI: 10.1016/j.jaci.2016.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 10/08/2016] [Accepted: 11/10/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Regina K Rowe
- Department of Pediatrics, University of Texas Southwestern, Dallas, Tex
| | - David M Pyle
- Department of Pediatrics, University of Texas Southwestern, Dallas, Tex; Department of Immunology, University of Texas Southwestern, Dallas, Texas
| | - Andrew R Tomlinson
- Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas
| | - Tinghong Lv
- Department of Pediatrics, University of Texas Southwestern, Dallas, Tex
| | - Zheng Hu
- Department of Pediatrics, University of Texas Southwestern, Dallas, Tex
| | - Michelle A Gill
- Department of Pediatrics, University of Texas Southwestern, Dallas, Tex; Department of Immunology, University of Texas Southwestern, Dallas, Texas; Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas.
| |
Collapse
|
124
|
Abstract
Macrophages (MPs) are one of the most prominent leukocyte populations in the lung and, in many ways, a forgotten player in asthma pathogenesis. Diverse functions in asthma initiation and maintenance in chronic disease have been demonstrated, which has led to confusion as to if pulmonary MPs are agents of good or evil in asthma. Much of this is due to the wide diversity of MP populations in the lung, many of which are inaccessible experimentally in most clinical studies. This review frames lung MP biology in the context of location, phenotype, function, and response phase in asthma pathogenesis. It also assesses new findings regarding MP diversity that have challenged old dogmas and generates new ways to understand how MPs function.
Collapse
|
125
|
Lambrecht BN, Persson EK, Hammad H. Myeloid Cells in Asthma. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0053-2016. [PMID: 28102118 PMCID: PMC11687443 DOI: 10.1128/microbiolspec.mchd-0053-2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Indexed: 12/24/2022] Open
Abstract
Asthma is a heterogeneous chronic inflammatory disorder of the airways, and not surprisingly, many myeloid cells play a crucial role in pathogenesis. Antigen-presenting dendritic cells are the first to recognize the allergens, pollutants, and viruses that are implicated in asthma pathogenesis, and subsequently initiate the adaptive immune response by migrating to lymph nodes. Eosinophils are the hallmark of type 2 inflammation, releasing toxic compounds in the airways and contributing to airway remodeling. Mast cells and basophils control both the early- and late-phase allergic response and contribute to alterations in smooth muscle reactivity. Finally, relatively little is known about neutrophils and macrophages in this disease. Although many of these myeloid cells respond well to treatment with inhaled steroids, there is now an increasing armamentarium of targeted biologicals that can specifically eliminate only one myeloid cell population, like eosinophils. It is only with those new tools that we will be able to fully understand the role of myeloid cells in chronic asthma in humans.
Collapse
Affiliation(s)
- Bart N Lambrecht
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Emma K Persson
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
| | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| |
Collapse
|
126
|
Park GY, Christman JW. Hidden in Plain Sight: The Overlooked Role of Pulmonary Macrophages in the Pathogenesis of Asthma. Am J Respir Cell Mol Biol 2016; 55:465-466. [PMID: 27689794 DOI: 10.1165/rcmb.2016-0188ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Gye Young Park
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy University of Illinois at Chicago Chicago, Illinois and
| | - John W Christman
- 2 Division of Pulmonary, Allergy, Critical Care and Sleep The Ohio State University Columbus, Ohio
| |
Collapse
|
127
|
Girodet PO, Nguyen D, Mancini JD, Hundal M, Zhou X, Israel E, Cernadas M. Alternative Macrophage Activation Is Increased in Asthma. Am J Respir Cell Mol Biol 2016; 55:467-475. [PMID: 27248771 DOI: 10.1165/rcmb.2015-0295oc] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The immune responses of type 2 T helper cells (Th2) play an important role in asthma and promote the differentiation of alternatively activated (M2) macrophages. M2 macrophages have been increasingly understood to contribute to Th2 immunity. We hypothesized that M2 macrophages are altered in asthma and modulate Th2 responses. The aim of this study was to characterize the phenotype and function of human monocyte-derived M2 and bronchoalveolar lavage fluid (BALF) macrophages from healthy control subjects and subjects with asthma. Phenotypic characteristics and effector function of M2 macrophages were examined using monocyte-derived and BALF macrophages obtained from subjects with asthma (n = 28) and healthy volunteers (n = 9) by flow cytometry and quantitative PCR. Resting monocyte-derived (M0) and M2 macrophages were generated by the addition of macrophage colony-stimulating factor or macrophage colony-stimulating factor plus IL-4, respectively. M2 macrophage cytokine expression and their impact on dendritic and CD4+ T cell activation were examined in vitro. High levels of CD206 and major histocompatibility complex class II expression identify macrophages with an M2 phenotype that are increased 2.9-fold in the BALF of subjects with asthma compared with control subjects. M2 macrophages have elevated IL-6, IL-10, and IL-12p40 production compared with conventional macrophages and modulate dendritic and CD4+ T cell interactions. Histamine receptor 1 and E-cadherin expression identify M2 macrophage subsets associated with increased airflow obstruction. M2 macrophages have a distinct cell surface and effector phenotype and are found in increased numbers in subjects with asthma. These findings suggest that M2 macrophages may play an important role in allergic asthma through their bidirectional interactions with immune and structural cells, and inflammatory mediators.
Collapse
Affiliation(s)
- Pierre-Olivier Girodet
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,2 University Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; and
| | - Daniel Nguyen
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - John Dominic Mancini
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,3 Channing Division of Network Medicine, Harvard Medical School, Boston, Massachusetts
| | - Mandeep Hundal
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Xiaobo Zhou
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,3 Channing Division of Network Medicine, Harvard Medical School, Boston, Massachusetts
| | - Elliot Israel
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Manuela Cernadas
- 1 Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
128
|
Kobayashi K, Koyama K, Suzukawa M, Igarashi S, Hebisawa A, Nagase T, Ohta K. Epithelial-mesenchymal transition promotes reactivity of human lung adenocarcinoma A549 cells to CpG ODN. Allergol Int 2016; 65 Suppl:S45-52. [PMID: 27475623 DOI: 10.1016/j.alit.2016.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/05/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is reported to promote airway remodeling in asthmatics, which is the main histological change that causes complex and severe symptoms in asthmatics. However, little is known about whether EMT also plays a role in acute exacerbations of asthma evoked by respiratory tract infections. METHODS A human lung adenocarcinoma line, A549, was incubated with TGF-β1 at 10 ng/ml to induce EMT. Then the cells were stimulated with CpG ODN. Expression of surface and intracellular molecules was analyzed by flow cytometry. IL-6, IL-8 and MCP-1 in the culture supernatant were measured by Cytometric Bead Assay, and the expression of mRNA was quantitated by real-time PCR. CpG ODN uptake was analyzed by flow cytometry. RESULTS The culture supernatant levels of IL-6, IL-8 and MCP-1 and the expression of mRNA for these cytokines in CpG ODN-stimulated A549 cells that had undergone EMT was significantly higher compared to those that had not. Addition of ODN H154, a TLR9-inhibiting DNA, significantly suppressed the CpG ODN-induced production of those cytokines. However, flow cytometry found the level of TLR9 expression to be slightly lower in A549 cells that had undergone EMT compared to those that had not. On the other hand, CpG ODN uptake was increased in cells that had undergone EMT. CONCLUSIONS EMT induction of A549 cells enhanced CpG ODN uptake and CpG ODN-induced production of IL-6, IL-8 and MCP-1. These results suggest that EMT plays an important role in exacerbation in asthmatics with airway remodeling by enhancing sensitivity to extrinsic pathogens.
Collapse
Affiliation(s)
- Koichi Kobayashi
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Kazuya Koyama
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Maho Suzukawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan.
| | - Sayaka Igarashi
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Akira Hebisawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, University of Tokyo, Tokyo, Japan
| | - Ken Ohta
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| |
Collapse
|
129
|
Liu CL, Zhang JY, Shi GP. Interaction between allergic asthma and atherosclerosis. Transl Res 2016; 174:5-22. [PMID: 26608212 PMCID: PMC4826642 DOI: 10.1016/j.trsl.2015.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/15/2022]
Abstract
Prior studies have established an essential role of mast cells in allergic asthma and atherosclerosis. Mast cell deficiency or inactivation protects mice from allergen-induced airway hyper-responsiveness and diet-induced atherosclerosis, suggesting that mast cells share pathologic activities in both diseases. Allergic asthma and atherosclerosis are inflammatory diseases that contain similar sets of elevated numbers of inflammatory cells in addition to mast cells in the airway and arterial wall, such as macrophages, monocytes, T cells, eosinophils, and smooth muscle cells. Emerging evidence from experimental models and human studies points to a potential interaction between the 2 seemingly unrelated diseases. Patients or mice with allergic asthma have a high risk of developing atherosclerosis or vice versa, despite the fact that asthma is a T-helper (Th)2-oriented disease, whereas Th1 immunity promotes atherosclerosis. In addition to the preferred Th1/Th2 responses that may differentiate the 2 diseases, mast cells and many other inflammatory cells also contribute to their pathogenesis by more than just T cell immunity. Here, we summarize the different roles of airway and arterial wall inflammatory cells and vascular cells in asthma and atherosclerosis and propose an interaction between the 2 diseases, although limited investigations are available to delineate the molecular and cellular mechanisms by which 1 disease increases the risk of the other. Results from mouse allergic asthma and atherosclerosis models and from human population studies lead to the hypothesis that patients with atherosclerosis may benefit from antiasthmatic medications or that the therapeutic regimens targeting atherosclerosis may also alleviate allergic asthma.
Collapse
Affiliation(s)
- Cong-Lin Liu
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Jin-Ying Zhang
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo-Ping Shi
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
130
|
Saini Y, Wilkinson KJ, Terrell KA, Burns KA, Livraghi-Butrico A, Doerschuk CM, O'Neal WK, Boucher RC. Neonatal Pulmonary Macrophage Depletion Coupled to Defective Mucus Clearance Increases Susceptibility to Pneumonia and Alters Pulmonary Immune Responses. Am J Respir Cell Mol Biol 2016; 54:210-21. [PMID: 26121027 DOI: 10.1165/rcmb.2014-0111oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Resident immune cells (e.g., macrophages [MΦs]) and airway mucus clearance both contribute to a healthy lung environment. To investigate interactions between pulmonary MΦ function and defective mucus clearance, a genetic model of lysozyme M (LysM) promoter-mediated MΦ depletion was generated, characterized, and crossed with the sodium channel β subunit transgenic (Scnn1b-Tg) mouse model of defective mucus clearance. Diphtheria toxin A-mediated depletion of LysM(+) pulmonary MΦs in wild-type mice with normal mucus clearance resulted in lethal pneumonia in 24% of neonates. The pneumonias were dominated by Pasteurella pneumotropica and accompanied by emaciation, neutrophilic inflammation, and elevated Th1 cytokines. The incidence of emaciation and pneumonia reached 51% when LysM(+) MΦ depletion was superimposed on the airway mucus clearance defect of Scnn1b-Tg mice. In LysM(+) MΦ-depleted Scnn1b-Tg mice, pneumonias were associated with a broader spectrum of bacterial species and a significant reduction in airway mucus plugging. Bacterial burden (CFUs) was comparable between Scnn1b-Tg and nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice. However, the nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice exhibited increased airway inflammation, the presence of neutrophilic infiltration, and increased levels of inflammatory cytokines in bronchoalveolar lavage fluid compared with Scnn1b-Tg mice. Collectively, these data identify key MΦ-mucus clearance interactions with respect to both infectious and inflammatory components of muco-obstructive lung disease.
Collapse
Affiliation(s)
- Yogesh Saini
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and.,2 Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Kristen J Wilkinson
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Kristy A Terrell
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Kimberlie A Burns
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Alessandra Livraghi-Butrico
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Claire M Doerschuk
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Wanda K O'Neal
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Richard C Boucher
- 1 Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
131
|
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
|
132
|
CCL2 Mediates Neuron-Macrophage Interactions to Drive Proregenerative Macrophage Activation Following Preconditioning Injury. J Neurosci 2016; 35:15934-47. [PMID: 26631474 DOI: 10.1523/jneurosci.1924-15.2015] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
CNS neurons in adult mammals do not spontaneously regenerate axons after spinal cord injury. Preconditioning peripheral nerve injury allows the dorsal root ganglia (DRG) sensory axons to regenerate beyond the injury site by promoting expression of regeneration-associated genes. We have previously shown that peripheral nerve injury increases the number of macrophages in the DRGs and that the activated macrophages are critical to the enhancement of intrinsic regeneration capacity. The present study identifies a novel chemokine signal mediated by CCL2 that links regenerating neurons with proregenerative macrophage activation. Neutralization of CCL2 abolished the neurite outgrowth activity of conditioned medium obtained from neuron-macrophage cocultures treated with cAMP. The neuron-macrophage interactions that produced outgrowth-promoting conditioned medium required CCL2 in neurons and CCR2/CCR4 in macrophages. The conditioning effects were abolished in CCL2-deficient mice at 3 and 7 d after sciatic nerve injury, but CCL2 was dispensable for the initial growth response and upregulation of GAP-43 at the 1 d time point. Intraganglionic injection of CCL2 mimicked conditioning injury by mobilizing M2-like macrophages. Finally, overexpression of CCL2 in DRGs promoted sensory axon regeneration in a rat spinal cord injury model without harmful side effects. Our data suggest that CCL2-mediated neuron-macrophage interaction plays a critical role for amplification and maintenance of enhanced regenerative capacity by preconditioning peripheral nerve injury. Manipulation of chemokine signaling mediating neuron-macrophage interactions may represent a novel therapeutic approach to promote axon regeneration after CNS injury.
Collapse
|
133
|
Draijer C, Boorsma CE, Reker-Smit C, Post E, Poelstra K, Melgert BN. PGE2-treated macrophages inhibit development of allergic lung inflammation in mice. J Leukoc Biol 2016; 100:95-102. [PMID: 26931576 DOI: 10.1189/jlb.3mab1115-505r] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/06/2016] [Indexed: 11/24/2022] Open
Abstract
In healthy lungs, many macrophages are characterized by IL-10 production, and few are characterized by expression of IFN regulatory factor 5 (formerly M1) or YM1 and/or CD206 (formerly M2), whereas in asthma, this balance shifts toward few producing IL-10 and many expressing IFN regulatory factor 5 or YM1/CD206. In this study, we tested whether redressing the balance by reinstating IL-10 production could prevent house dust mite-induced allergic lung inflammation. PGE2 was found to be the best inducer of IL-10 in macrophages in vitro. Mice were then sensitized and challenged to house dust mites during a 2 wk protocol while treated with PGE2 in different ways. Lung inflammation was assessed 3 d after the last house dust mite challenge. House dust mite-exposed mice treated with free PGE2 had fewer infiltrating eosinophils in lungs and lower YM1 serum levels than vehicle-treated mice. Macrophage-specific delivery of PGE2 did not affect lung inflammation. Adoptive transfer of PGE2-treated macrophages led to fewer infiltrating eosinophils, macrophages, (activated) CD4(+), and regulatory T lymphocytes in lungs. Our study shows that the redirection of macrophage polarization by using PGE2 inhibits development of allergic lung inflammation. This beneficial effect of macrophage repolarization is a novel avenue to explore for therapeutic purposes.
Collapse
Affiliation(s)
- Christina Draijer
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Carian E Boorsma
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Catharina Reker-Smit
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and
| | - Eduard Post
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and
| | - Klaas Poelstra
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and
| | - Barbro N Melgert
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, The Netherlands and Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, The Netherlands
| |
Collapse
|
134
|
Nagre N, Wang S, Kellett T, Kanagasabai R, Deng J, Nishi M, Shilo K, Oeckler RA, Yalowich JC, Takeshima H, Christman J, Hubmayr RD, Zhao X. TRIM72 modulates caveolar endocytosis in repair of lung cells. Am J Physiol Lung Cell Mol Physiol 2015; 310:L452-64. [PMID: 26637632 DOI: 10.1152/ajplung.00089.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 12/01/2015] [Indexed: 01/11/2023] Open
Abstract
Alveolar epithelial and endothelial cell injury is a major feature of the acute respiratory distress syndrome, in particular when in conjunction with ventilation therapies. Previously we showed [Kim SC, Kellett T, Wang S, Nishi M, Nagre N, Zhou B, Flodby P, Shilo K, Ghadiali SN, Takeshima H, Hubmayr RD, Zhao X. Am J Physiol Lung Cell Mol Physiol 307: L449-L459, 2014.] that tripartite motif protein 72 (TRIM72) is essential for amending alveolar epithelial cell injury. Here, we posit that TRIM72 improves cellular integrity through its interaction with caveolin 1 (Cav1). Our data show that, in primary type I alveolar epithelial cells, lack of TRIM72 led to significant reduction of Cav1 at the plasma membrane, accompanied by marked attenuation of caveolar endocytosis. Meanwhile, lentivirus-mediated overexpression of TRIM72 selectively increases caveolar endocytosis in rat lung epithelial cells, suggesting a functional association between these two. Further coimmunoprecipitation assays show that deletion of either functional domain of TRIM72, i.e., RING, B-box, coiled-coil, or PRY-SPRY, abolishes the physical interaction between TRIM72 and Cav1, suggesting that all theoretical domains of TRIM72 are required to forge a strong interaction between these two molecules. Moreover, in vivo studies showed that injurious ventilation-induced lung cell death was significantly increased in knockout (KO) TRIM72(KO) and Cav1(KO) lungs compared with wild-type controls and was particularly pronounced in double KO mutants. Apoptosis was accompanied by accentuation of gross lung injury manifestations in the TRIM72(KO) and Cav1(KO) mice. Our data show that TRIM72 directly and indirectly modulates caveolar endocytosis, an essential process involved in repair of lung epithelial cells through removal of plasma membrane wounds. Given TRIM72's role in endomembrane trafficking and cell repair, we consider this molecule an attractive therapeutic target for patients with injured lungs.
Collapse
Affiliation(s)
- Nagaraja Nagre
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia; Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shaohua Wang
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Thomas Kellett
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Ragu Kanagasabai
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jing Deng
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Miyuki Nishi
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan; and
| | - Konstantin Shilo
- Division of Pulmonary Pathology, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Jack C Yalowich
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan; and
| | - John Christman
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Rolf D Hubmayr
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia; Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio; Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
135
|
Graham A. Mitochondrial regulation of macrophage cholesterol homeostasis. Free Radic Biol Med 2015; 89:982-92. [PMID: 26416507 DOI: 10.1016/j.freeradbiomed.2015.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/28/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022]
Abstract
This review explores the relationship between mitochondrial structure and function in the regulation of macrophage cholesterol metabolism and proposes that mitochondrial dysfunction contributes to loss of the elegant homeostatic mechanisms which normally maintain cellular sterol levels within defined limits. Mitochondrial sterol 27-hydroxylase (CYP27A1) can generate oxysterol activators of liver X receptors which heterodimerise with retinoid X receptors, enhancing the transcription of ATP binding cassette transporters (ABCA1, ABCG1, and ABCG4), that can remove excess cholesterol via efflux to apolipoproteins A-1, E, and high density lipoprotein, and inhibit inflammation. The activity of CYP27A1 is regulated by the rate of supply of cholesterol substrate to the inner mitochondrial membrane, mediated by a complex of proteins. The precise identity of this dynamic complex remains controversial, even in steroidogenic tissues, but may include steroidogenic acute regulatory protein and the 18 kDa translocator protein, together with voltage-dependent anion channels, ATPase AAA domain containing protein 3A, and optic atrophy type 1 proteins. Certainly, overexpression of StAR and TSPO proteins can enhance macrophage cholesterol efflux to apoA-I and/or HDL, while perturbations in mitochondrial function, or changes in the expression of mitochondrial fusion proteins, alter the efficiency of cholesterol efflux. Molecules which can sustain or improve mitochondrial function or increase the activity of the protein complex involved in cholesterol transfer may have utility in resolving the problem of dysregulated macrophage cholesterol homeostasis, a condition which may contribute to inflammation, atherosclerosis, nonalcoholic steatohepatitis, osteoblastic bone resorption, and some disorders of the central nervous system.
Collapse
Affiliation(s)
- Annette Graham
- Department of Life Sciences, School of Health and Life Sciences, and Institute for Applied Health Research, Glasgow Caledonian University, 70 Cowcaddens Road, Glasgow G4 0BA, United Kingdom.
| |
Collapse
|
136
|
Keselman A, Heller N. Estrogen Signaling Modulates Allergic Inflammation and Contributes to Sex Differences in Asthma. Front Immunol 2015; 6:568. [PMID: 26635789 PMCID: PMC4644929 DOI: 10.3389/fimmu.2015.00568] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/23/2015] [Indexed: 12/19/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease that affects ~300 million people worldwide. It is characterized by airway constriction that leads to wheezing, coughing, and shortness of breath. The most common treatments are corticosteroids and β2-adrenergic receptor antagonists, which target inflammation and airway smooth muscle constriction, respectively. The incidence and severity of asthma is greater in women than in men, and women are more prone to develop corticosteroid-resistant or “hard-to-treat” asthma. Puberty, menstruation, pregnancy, menopause, and oral contraceptives are known to contribute to disease outcome in women, suggesting a role for estrogen and other hormones impacting allergic inflammation. Currently, the mechanisms underlying these sex differences are poorly understood, although the effect of sex hormones, such as estrogen, on allergic inflammation is gaining interest. Asthma presents as a heterogeneous disease. In typical Th2-type allergic asthma, interleukin (IL)-4 and IL-13 predominate, driving IgE production and recruitment of eosinophils into the lungs. Chronic Th2-inflammation in the lung results in structural changes and activation of multiple immune cell types, leading to a deterioration of lung function over time. Most immune cells express estrogen receptors (ERα, ERβ, or the membrane-bound G-protein-coupled ER) to varying degrees and can respond to the hormone. Together these receptors have demonstrated the capacity to regulate a spectrum of immune functions, including adhesion, migration, survival, wound healing, and antibody and cytokine production. This review will cover the current understanding of estrogen signaling in allergic inflammation and discuss how this signaling may contribute to sex differences in asthma and allergy.
Collapse
Affiliation(s)
- Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Nicola Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
137
|
de Boer JD, Berkhout LC, de Stoppelaar SF, Yang J, Ottenhoff R, Meijers JCM, Roelofs JJTH, van't Veer C, van der Poll T. Effect of the oral thrombin inhibitor dabigatran on allergic lung inflammation induced by repeated house dust mite administration in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L768-75. [PMID: 26320153 DOI: 10.1152/ajplung.00102.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 08/18/2015] [Indexed: 01/08/2023] Open
Abstract
Asthma is a chronic disease of the airways; asthma patients are hampered by recurrent symptoms of dyspnoea and wheezing caused by bronchial obstruction. Most asthma patients suffer from chronic allergic lung inflammation triggered by allergens such as house dust mite (HDM). Coagulation activation in the pulmonary compartment is currently recognized as a feature of allergic lung inflammation, and data suggest that coagulation proteases further drive inflammatory mechanisms. Here, we tested whether treatment with the oral thrombin inhibitor dabigatran attenuates allergic lung inflammation in a recently developed HDM-based murine asthma model. Mice were fed dabigatran (10 mg/g) or placebo chow during a 3-wk HDM airway exposure model. Dabigatran treatment caused systemic thrombin inhibitory activity corresponding with dabigatran levels reported in human trials. Surprisingly, dabigatran did not lead to inhibition of HDM-evoked coagulation activation in the lung as measured by levels of thrombin-antithrombin complexes and D-dimer. Repeated HDM administration caused an influx of eosinophils and neutrophils into the lungs, mucus production in the airways, and a T helper 2 response, as reflected by a rise in bronchoalveolar IL-4 and IL-5 levels and a systemic rise in IgE and HDM-IgG1. Dabigatran modestly improved HDM-induced lung pathology (P < 0.05) and decreased IL-4 levels (P < 0.01), without influencing other HDM-induced responses. Considering the limited effects of dabigatran in spite of adequate plasma levels, these results argue against clinical evaluation of dabigatran in patients with asthma.
Collapse
Affiliation(s)
- Johannes D de Boer
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands.
| | - Lea C Berkhout
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands
| | - Sacha F de Stoppelaar
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands
| | - Jack Yang
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Joost C M Meijers
- Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Cornelis van't Veer
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands
| | - Tom van der Poll
- Academic Medical Center, University of Amsterdam, Center of Infection and Immunity Amsterdam & Center for Experimental and Molecular Medicine, the Netherlands. Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
138
|
Sanfilippo AM, Furuya Y, Roberts S, Salmon SL, Metzger DW. Allergic Lung Inflammation Reduces Tissue Invasion and Enhances Survival from Pulmonary Pneumococcal Infection in Mice, Which Correlates with Increased Expression of Transforming Growth Factor β1 and SiglecF(low) Alveolar Macrophages. Infect Immun 2015; 83:2976-83. [PMID: 25964474 PMCID: PMC4468552 DOI: 10.1128/iai.00142-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/04/2015] [Indexed: 11/20/2022] Open
Abstract
Asthma is generally thought to confer an increased risk for invasive pneumococcal disease (IPD) in humans. However, recent reports suggest that mortality rates from IPD are unaffected in patients with asthma and that chronic obstructive pulmonary disease (COPD), a condition similar to asthma, protects against the development of complicated pneumonia. To clarify the effects of asthma on the subsequent susceptibility to pneumococcal infection, ovalbumin (OVA)-induced allergic lung inflammation (ALI) was induced in mice followed by intranasal infection with A66.1 serotype 3 Streptococcus pneumoniae. Surprisingly, mice with ALI were significantly more resistant to lethal infection than non-ALI mice. The heightened resistance observed following ALI correlated with enhanced early clearance of pneumococci from the lung, decreased bacterial invasion from the airway into the lung tissue, a blunted inflammatory cytokine and neutrophil response to infection, and enhanced expression of transforming growth factor β1 (TGF-β1). Neutrophil depletion prior to infection had no effect on enhanced early bacterial clearance or resistance to IPD in mice with ALI. Although eosinophils recruited into the lung during ALI appeared to be capable of phagocytizing bacteria, neutralization of interleukin-5 (IL-5) to inhibit eosinophil recruitment likewise had no effect on early clearance or survival following infection. However, enhanced resistance was associated with an increase in levels of clodronate-sensitive, phagocytic SiglecF(low) alveolar macrophages within the airways following ALI. These findings suggest that, while the risk of developing IPD may actually be decreased in patients with acute asthma, additional clinical data are needed to better understand the risk of IPD in patients with different asthma phenotypes.
Collapse
Affiliation(s)
- Alan M Sanfilippo
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Yoichi Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sean Roberts
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sharon L Salmon
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Dennis W Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
139
|
Qian F, Deng J, Lee YG, Zhu J, Karpurapu M, Chung S, Zheng JN, Xiao L, Park GY, Christman JW. The transcription factor PU.1 promotes alternative macrophage polarization and asthmatic airway inflammation. J Mol Cell Biol 2015; 7:557-67. [PMID: 26101328 DOI: 10.1093/jmcb/mjv042] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/10/2015] [Indexed: 12/25/2022] Open
Abstract
The transcription factor PU.1 is involved in regulation of macrophage differentiation and maturation. However, the role of PU.1 in alternatively activated macrophage (AAM) and asthmatic inflammation has yet been investigated. Here we report that PU.1 serves as a critical regulator of AAM polarization and promotes the pathological progress of asthmatic airway inflammation. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, conditional PU.1-deficient (PU/ER(T)(+/-)) mice displayed attenuated allergic airway inflammation, including decreased alveolar eosinophil infiltration and reduced production of IgE, which were associated with decreased mucous glands and goblet cell hyperplasia. The reduced asthmatic inflammation in PU/ER(T)(+/-) mice was restored by adoptive transfer of IL-4-induced wild-type (WT) macrophages. Moreover, after treating PU/ER(T)(+/-) mice with tamoxifen to rescue PU.1 function, the allergic asthmatic inflammation was significantly restored. In vitro studies demonstrate that treatment of PU.1-deficient macrophages with IL-4 attenuated the expression of chitinase 3-like 3 (Ym-1) and resistin-like molecule alpha 1 (Fizz-1), two specific markers of AAM polarization. In addition, PU.1 expression in macrophages was inducible in response to IL-4 challenge, which was associated with phosphorylation of signal transducer and activator of transcription 6 (STAT6). Furthermore, DRA challenge in sensitized mice almost abrogated gene expression of Ym-1 and Fizz-1 in lung tissues of PU/ER(T)(+/-) mice compared with WT mice. These data, all together, indicate that PU.1 plays a critical role in AAM polarization and asthmatic inflammation.
Collapse
Affiliation(s)
- Feng Qian
- School of Pharmacy, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| | - Jing Deng
- Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| | - Yong Gyu Lee
- Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| | - Jimmy Zhu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Manjula Karpurapu
- Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| | - Sangwoon Chung
- Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| | - Jun-Nian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, China
| | - Lei Xiao
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, University of Illinois at Chicago, Chicago, USA
| | - Gye Young Park
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, University of Illinois at Chicago, Chicago, USA
| | - John W Christman
- Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, USA
| |
Collapse
|
140
|
Yu L, Wu X, Wei J, Liao Q, Xu L, Luo S, Zeng X, Zhao Y, Lv Z, Wu Z. Preliminary expression profile of cytokines in brain tissue of BALB/c mice with Angiostrongylus cantonensis infection. Parasit Vectors 2015; 8:328. [PMID: 26070790 PMCID: PMC4476182 DOI: 10.1186/s13071-015-0939-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/05/2015] [Indexed: 12/05/2022] Open
Abstract
Background Angiostrongylus cantonensis (A. cantonensis) infection can result in increased risk of eosinophilic meningitis. Accumulation of eosinophils and inflammation can result in the A. cantonensis infection playing an important role in brain tissue injury during this pathological process. However, underlying mechanisms regarding the transcriptomic responses during brain tissue injury caused by A. cantonensis infection are yet to be elucidated. This study is aimed at identifying some genomic and transcriptomic factors influencing the accumulation of eosinophils and inflammation in the mouse brain infected with A. cantonensis. Methods An infected mouse model was prepared based on our laboratory experimental process, and then the mouse brain RNA Libraries were constructed for deep Sequencing with Illumina Genome Analyzer. The raw data was processed with a bioinformatics’ pipeline including Refseq genes expression analysis using cufflinks, annotation and classification of RNAs, lncRNA prediction as well as analysis of co-expression network. The analysis of Refseq data provides the measure of the presence and prevalence of transcripts from known and previously unknown genes. Results This study showed that Cys-Cys (CC) type chemokines such as CCL2, CCL8, CCL1, CCL24, CCL11, CCL7, CCL12 and CCL5 were elevated significantly at the late phase of infection. The up-regulation of CCL2 indicated that the worm of A. cantonensis had migrated into the mouse brain at an early infection phase. CCL2 could be induced in the brain injury during migration and CCL2 might play a major role in the neuropathic pain caused by A. cantonensis infection. The up-regulated expression of IL-4, IL-5, IL-10, and IL-13 showed Th2 cell predominance in immunopathological reactions at late infection phase in response to infection by A. cantonensis. These different cytokines can modulate and inhibit each other and function as a network with the specific potential to drive brain eosinophilic inflammation. The increase of ATF-3 expression at 21 dpi suggested the injury of neuronal cells at late phase of infection. 1217 new potential lncRNA were candidates of interest for further research. Conclusions These cytokine networks play an important role in the development of central nervous system inflammation caused by A. cantonensis infection. Electronic supplementary material The online version of this article (doi:10.1186/s13071-015-0939-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liping Yu
- Department of Preventive Medicine, School of Medicine, Three Gorges University, Yichang, China. .,Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Xiaoying Wu
- Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Jie Wei
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Qi Liao
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China.
| | - Lian Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Siqi Luo
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Xin Zeng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Yi Zhao
- Advanced Computing Research Laboratory, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
| | - Zhiyue Lv
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, The Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|