1
|
Li J, Wang H, Xia S. Hematopoietic stem and progenitor cells fine-tuning the "sweet" of trained immunity. J Leukoc Biol 2025; 117:qiaf043. [PMID: 40233187 DOI: 10.1093/jleuko/qiaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Recent studies have challenged the traditional view of innate immunity as nonspecific and transient by demonstrating that innate immune cells can develop immune memory in response to various activating factors, a phenomenon known as trained immunity. This process involves epigenetic modifications, such as changes in chromatin accessibility, and metabolic reprogramming, which can provide protection against unrelated pathogens but may also trigger immune-mediated damage. This review summarizes the current understanding of innate immune memory, with a particular focus on recent findings regarding the training of innate immune cells at the hematopoietic stem and progenitor cell stage. We present observations of trained immunity in innate immune cells, summarize key activating factors and underlying mechanisms, and propose potential host-directed immunotherapeutic strategies and preventive measures based on trained immunity. Our aim is to highlight the biological significance of trained immunity and its potential applications in enhancing long-term immunity, improving vaccine efficacy, and preventing immune-related diseases.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| |
Collapse
|
2
|
Creyns B, MacKenzie B, Jannini Sa YAP, Coelho AL, Christensen D, Parimon T, Windsor B, Hogaboam CM. Caveolin Scaffolding Domain (CSD) Peptide LTI-2355 Modulates the Phagocytic and Synthetic Activity of Lung-Derived Myeloid Cells in Idiopathic Pulmonary Fibrosis (IPF) and Post-Acute Sequelae of COVID Fibrosis (PASC-F). Biomedicines 2025; 13:796. [PMID: 40299362 PMCID: PMC12024842 DOI: 10.3390/biomedicines13040796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Rationale: The role of the innate immune system in idiopathic pulmonary fibrosis (IPF) remains poorly understood. However, a functional myeloid compartment is required to remove dying cells and cellular debris, as well as to mediate innate immune responses against pathogens. Aberrant macrophage activity has been described in patients with post-acute sequelae of COVID fibrosis (PASC-F), and caveolin scaffolding domain (CSD) peptides have been found to attenuate inflammation and fibrosis in mouse lung injury models. Therefore, we examined, for the first time, the effects of CSD peptide LTI-2355 on the functional and synthetic properties of human myeloid cells isolated from lung explant tissue of donor lungs as well as IPF and PASC-F lung explant tissue. Methods and Results: CD45+ myeloid cells isolated from lung explant tissue from IPF and PASC-F patients exhibited an impaired capacity to clear autologous dead cells and cellular debris. The uptake of pathogen-coated bioparticles was impaired in myeloid cells from both fibrotic patient groups independent of the type of pathogen, highlighting an intrinsic functional cell impairment. LTI-2355 improved the phagocytic activity of both IPF and PASC-F myeloid cells, and this improvement was paired with decreased proinflammatory and pro-fibrotic synthetic activity. LTI-2355 was also shown to primarily target CD206-expressing IPF and PASC-F myeloid cells. Conclusions: Primary myeloid cells from IPF and PASC-F patients exhibit dysfunctional phagocytic and synthetic properties that are modulated by LTI-2355. LTI-2355 treatment of IPF myeloid cells resulted in significantly reduced sCD163, IFN-α2, IFN-γ, IL-2, IL-10, IL-12p40, and MMP-1 in the cell supernatant. This study highlights an additional mechanism of action of the CSD peptide in the treatment of IPF and progressive fibrotic lung disease.
Collapse
Affiliation(s)
- Brecht Creyns
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - BreAnne MacKenzie
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Yago Amigo Pinho Jannini Sa
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Ana Lucia Coelho
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Dale Christensen
- Division of Hematology, Department of Medicine, Duke University, Durham, NC 27708, USA;
| | - Tanyalak Parimon
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Brian Windsor
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Cory M. Hogaboam
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| |
Collapse
|
3
|
Xia S, Gu X, Wang G, Zhong Y, Ma F, Liu Q, Xie J. Regulated Cell Death of Alveolar Macrophages in Acute Lung Inflammation: Current Knowledge and Perspectives. J Inflamm Res 2024; 17:11419-11436. [PMID: 39722732 PMCID: PMC11669335 DOI: 10.2147/jir.s497775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common and serious clinical lung disease characterized by extensive alveolar damage and inflammation leading to impaired gas exchange. Alveolar macrophages (AMs) maintain homeostatic properties and immune defenses in lung tissues. Several studies have reported that AMs are involved in and regulate ALI/ARDS onset and progression via different regulated cell death (RCD) programs, such as pyroptosis, apoptosis, autophagic cell death, and necroptosis. Notably, the effects of RCD in AMs in disease are complex and variable depending on the environment and stimuli. In this review, we provide a comprehensive perspective on how regulated AMs death impacts on ALI/ARDS and assess its potential in new therapeutic development. Additionally, we describe the crosstalk between different RCD types in ALI, and provide new perspectives for the treatment of ALI/ARDS and other severe lung diseases.
Collapse
Affiliation(s)
- Siwei Xia
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyan Gu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fengjie Ma
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qinxue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
4
|
Ku KB, Chae J, Park WH, La J, Lee SS, Lee HK. Assessment of immunopathological responses of a novel non-chemical biocide in C57BL/6 for safe disinfection usage. Lab Anim Res 2024; 40:28. [PMID: 39135094 PMCID: PMC11320990 DOI: 10.1186/s42826-024-00214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Water electrospray technology has been developed and extensively studied for its physical properties and potential application as a non-chemical biocide against airborne pathogens. However, there are still concerns regarding the safety and potential toxicity of inhaling water electrospray (WE) particles. To address these potential hazards and offer insights into the impact of WE on humans, we analyzed the immunopathological response to WE by employing an intranasal challenge C57BL/6 mouse model. This analysis aimed to compare the effects of WE with those of sodium hypochlorite (SH), a well-known biocidal agent. RESULTS The study findings suggest that the WE did not trigger any pathological immune reactions in the intranasal-challenged C57BL/6 mouse model. Mice challenged with WE did not experience body weight loss, and there was no increase in inflammatory cytokine production compared to SH-treated mice. Histopathological analysis revealed that WE did not cause any damage to the lung tissue. In contrast, mice treated with SH exhibited significant lung tissue damage, characterized by the infiltration of neutrophils and eosinophils. Transcriptomic analysis of lung tissue further confirmed the absence of a pathological immune response in mice treated with WE compared to those treated with SH. Upon intranasal challenge with WE, the C57BL/6 mouse model did not show any evidence of immunopathological damage. CONCLUSIONS The results of this study suggest that WE is a safe technology for disinfecting airborne pathogens. It demonstrated little to no effect on immune system activation and pathological outcomes in the intranasal challenge C57BL/6 mouse model. These findings not only support the potential use of WE as an effective and safe method for air disinfection but also highlight the value of the intranasal challenge of the C57BL/6 mouse model in providing significant immunopathological insights for assessing the inhalation of novel materials for potential use.
Collapse
Affiliation(s)
- Keun Bon Ku
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Center for Infectious Disease Vaccine and Diagnosis Innovation, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jihwan Chae
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Won Hyung Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jeongwoo La
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Seung S Lee
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
5
|
Pervizaj-Oruqaj L, Ferrero MR, Matt U, Herold S. The guardians of pulmonary harmony: alveolar macrophages orchestrating the symphony of lung inflammation and tissue homeostasis. Eur Respir Rev 2024; 33:230263. [PMID: 38811033 PMCID: PMC11134199 DOI: 10.1183/16000617.0263-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/20/2024] [Indexed: 05/31/2024] Open
Abstract
Recent breakthroughs in single-cell sequencing, advancements in cellular and tissue imaging techniques, innovations in cell lineage tracing, and insights into the epigenome collectively illuminate the enigmatic landscape of alveolar macrophages in the lung under homeostasis and disease conditions. Our current knowledge reveals the cellular and functional diversity of alveolar macrophages within the respiratory system, emphasising their remarkable adaptability. By synthesising insights from classical cell and developmental biology studies, we provide a comprehensive perspective on alveolar macrophage functional plasticity. This includes an examination of their ontology-related features, their role in maintaining tissue homeostasis under steady-state conditions and the distinct contribution of bone marrow-derived macrophages (BMDMs) in promoting tissue regeneration and restoring respiratory system homeostasis in response to injuries. Elucidating the signalling pathways within inflammatory conditions, the impact of various triggers on tissue-resident alveolar macrophages (TR-AMs), as well as the recruitment and polarisation of macrophages originating from the bone marrow, presents an opportunity to propose innovative therapeutic approaches aimed at modulating the equilibrium between phenotypes to induce programmes associated with a pro-regenerative or homeostasis phenotype of BMDMs or TR-AMs. This, in turn, can lead to the amelioration of disease outcomes and the attenuation of detrimental inflammation. This review comprehensively addresses the pivotal role of macrophages in the orchestration of inflammation and resolution phases after lung injury, as well as ageing-related shifts and the influence of clonal haematopoiesis of indeterminate potential mutations on alveolar macrophages, exploring altered signalling pathways and transcriptional profiles, with implications for respiratory homeostasis.
Collapse
Affiliation(s)
- Learta Pervizaj-Oruqaj
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Maximiliano Ruben Ferrero
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
6
|
Zhang J, Liu Y. Epithelial stem cells and niches in lung alveolar regeneration and diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:17-26. [PMID: 38645714 PMCID: PMC11027191 DOI: 10.1016/j.pccm.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Indexed: 04/23/2024]
Abstract
Alveoli serve as the functional units of the lungs, responsible for the critical task of blood-gas exchange. Comprising type I (AT1) and type II (AT2) cells, the alveolar epithelium is continuously subject to external aggressors like pathogens and airborne particles. As such, preserving lung function requires both the homeostatic renewal and reparative regeneration of this epithelial layer. Dysfunctions in these processes contribute to various lung diseases. Recent research has pinpointed specific cell subgroups that act as potential stem or progenitor cells for the alveolar epithelium during both homeostasis and regeneration. Additionally, endothelial cells, fibroblasts, and immune cells synergistically establish a nurturing microenvironment-or "niche"-that modulates these epithelial stem cells. This review aims to consolidate the latest findings on the identities of these stem cells and the components of their niche, as well as the molecular mechanisms that govern them. Additionally, this article highlights diseases that arise due to perturbations in stem cell-niche interactions. We also discuss recent technical innovations that have catalyzed these discoveries. Specifically, this review underscores the heterogeneity, plasticity, and dynamic regulation of these stem cell-niche systems. It is our aspiration that a deeper understanding of the fundamental cellular and molecular mechanisms underlying alveolar homeostasis and regeneration will open avenues for identifying novel therapeutic targets for conditions such as chronic obstructive pulmonary disease (COPD), fibrosis, coronavirus disease 2019 (COVID-19), and lung cancer.
Collapse
Affiliation(s)
- Jilei Zhang
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yuru Liu
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Creyns B, MacKenzie B, Sa Y, Coelho AL, Christensen D, Parimon T, Windsor B, Hogaboam CM. Caveolin scaffolding domain (CSD) peptide LTI-2355 modulates the phagocytic and synthetic activity of lung derived myeloid cells in Idiopathic Pulmonary Fibrosis (IPF) and Post-acute sequelae of COVID-fibrosis (PASC-F). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.01.569608. [PMID: 38654821 PMCID: PMC11037873 DOI: 10.1101/2023.12.01.569608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Rationale The role of the innate immune system in Idiopathic Pulmonary Fibrosis (IPF) remains poorly understood. However, a functional myeloid compartment is required to remove dying cells and cellular debris, and to mediate innate immune responses against pathogens. Aberrant macrophage activity has been described in patients with Post-acute sequelae of COVID fibrosis (PASC-F). Therefore, we examined the functional and synthetic properties of myeloid cells isolated from normal donor lung and lung explant tissue from both IPF and PASC-F patients and explored the effect of LTI-2355, a Caveolin Scaffolding Domain (CSD) peptide, on these cells. Methods & Results CD45 + myeloid cells isolated from lung explant tissue from IPF and PASC-F patients exhibited an impaired capacity to clear autologous dead cells and cellular debris. Uptake of pathogen-coated bioparticles was impaired in myeloid cells from both fibrotic patient groups independent of type of pathogen highlighting a cell intrinsic functional impairment. LTI-2355 improved the phagocytic activity of both IPF and PASC-F myeloid cells, and this improvement was paired with decreased pro-inflammatory and pro-fibrotic synthetic activity. LTI-2355 was also shown to primarily target CD206-expressing IPF and PASC-F myeloid cells. Conclusions Primary myeloid cells from IPF and PASC-F patients exhibit dysfunctional phagocytic and synthetic properties that are reversed by LTI-2355. Thus, these studies highlight an additional mechanism of action of a CSD peptide in the treatment of IPF and progressive fibrotic lung disease.
Collapse
|
8
|
Zhou H, Peng K, Wang J, Wang Y, Wang JJ, Sun SK, Shi MQ, Chen J, Ji FH, Wang X. Aloe-derived vesicles enable macrophage reprogramming to regulate the inflammatory immune environment. Front Bioeng Biotechnol 2023; 11:1339941. [PMID: 38179130 PMCID: PMC10764618 DOI: 10.3389/fbioe.2023.1339941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction: Bacterial pneumonia poses a significant global public health challenge, where unaddressed pathogens and inflammation can exacerbate acute lung injury and prompt cytokine storms, increasing mortality rates. Alveolar macrophages are pivotal in preserving lung equilibrium. Excessive inflammation can trigger necrosis in these cells, disrupting the delicate interplay between inflammation and tissue repair. Methods: We obtained extracellular vesicle from aloe and tested the biosafety by cell viability and hemolysis assays. Confocal microscopy and flow cytometry were used to detect the uptake and internalization of extracellular vesicle by macrophages and the ability of extracellular vesicle to affect the phenotypic reprogramming of macrophages in vitro. Finally, we conducted a clinical feasibility study employing clinical bronchoalveolar lavage fluid as a representative model to assess the effective repolarization of macrophages influenced by extracellular vesicle. Results: In our study, we discovered the potential of extracellular vesicle nanovesicles derived from aloe in reprograming macrophage phenotypes. Pro-inflammatory macrophages undergo a transition toward an anti-inflammatory immune phenotype through phagocytosing and internalizing these aloe vera-derived extracellular vesicle nanovesicles. This transition results in the release of anti-inflammatory IL-10, effectively curbing inflammation and fostering lung tissue repair. Discussion: These findings firmly establish the immunomodulatory impact of aloe-derived extracellular vesicle nanovesicles on macrophages, proposing their potential as a therapeutic strategy to modulate macrophage immunity in bacterial pneumonia.
Collapse
Affiliation(s)
- Hao Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology and Institute of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jia-Jia Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shi-Kun Sun
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mai-Qing Shi
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Hai Ji
- Department of Anesthesiology and Institute of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
9
|
Lu HY, Wang MY, Zhu SX, Ju HM, Xu SQ, Qiao Y, Wei SJ, Su ZL. ILC2 influence the differentiation of alveolar type II epithelial cells in bronchopulmonary dysplasia mice. J Leukoc Biol 2023; 114:604-614. [PMID: 37647586 DOI: 10.1093/jleuko/qiad092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
Bronchopulmonary dysplasia, a common complication of premature infants, is mainly characterized by blocked alveolarization. Proverbially, the injury of alveolar type II epithelial cells is regarded as the pathologic basis of occurrence and development of bronchopulmonary dysplasia. In the case of alveolar epithelial damage, alveolar type II epithelial cells can also differentiate to alveolar type I epithelial cells as progenitor cells. During bronchopulmonary dysplasia, the differentiation of alveolar type II epithelial cells becomes abnormal. Group 2 innate lymphoid cells can produce type 2 cytokines in response to a variety of stimuli, including the epithelial cytokines IL-25, IL-33, and thymic stromal lymphopoietin. Previous studies have shown that group 2 innate lymphoid cells can inhibit the alveolarization process of bronchopulmonary dysplasia by secreting IL-13. However, whether group 2 innate lymphoid cells can affect the differentiation of alveolar type II epithelial cells in the pathologic process of bronchopulmonary dysplasia remains unclear. In this study, we have shown that IL-13 secreted by group 2 innate lymphoid cells increased during bronchopulmonary dysplasia, which was related to the release of large amounts of IL-33 by impaired alveolar type II epithelial cells. This led to abnormal differentiation of alveolar type II epithelial cells, reduced differentiation to alveolar type I epithelial cells, and increased transdifferentiation to mesenchymal cells through the epithelial-mesenchymal transition. Taken together, our study provides a complementary understanding of the development of bronchopulmonary dysplasia and highlights a novel immune mechanism in the pathogenesis of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Ming-Yan Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Shao-Xuan Zhu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Hui-Min Ju
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Su-Qing Xu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Yu Qiao
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Shan-Jie Wei
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Zhao-Liang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
- Institute for medical Immunology, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| |
Collapse
|
10
|
Chakraborty S, Singh A, Wang L, Wang X, Sanborn MA, Ye Z, Maienschein-Cline M, Mukhopadhyay A, Ganesh BB, Malik AB, Rehman J. Trained immunity of alveolar macrophages enhances injury resolution via KLF4-MERTK-mediated efferocytosis. J Exp Med 2023; 220:e20221388. [PMID: 37615937 PMCID: PMC10450795 DOI: 10.1084/jem.20221388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 05/19/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023] Open
Abstract
Recent studies suggest that training of innate immune cells such as tissue-resident macrophages by repeated noxious stimuli can heighten host defense responses. However, it remains unclear whether trained immunity of tissue-resident macrophages also enhances injury resolution to counterbalance the heightened inflammatory responses. Here, we studied lung-resident alveolar macrophages (AMs) prechallenged with either the bacterial endotoxin or with Pseudomonas aeruginosa and observed that these trained AMs showed greater resilience to pathogen-induced cell death. Transcriptomic analysis and functional assays showed greater capacity of trained AMs for efferocytosis of cellular debris and injury resolution. Single-cell high-dimensional mass cytometry analysis and lineage tracing demonstrated that training induces an expansion of a MERTKhiMarcohiCD163+F4/80low lung-resident AM subset with a proresolving phenotype. Reprogrammed AMs upregulated expression of the efferocytosis receptor MERTK mediated by the transcription factor KLF4. Adoptive transfer of these trained AMs restricted inflammatory lung injury in recipient mice exposed to lethal P. aeruginosa. Thus, our study has identified a subset of tissue-resident trained macrophages that prevent hyperinflammation and restore tissue homeostasis following repeated pathogen challenges.
Collapse
Affiliation(s)
- Sreeparna Chakraborty
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Abhalaxmi Singh
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Li Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Xinge Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois College of Medicine, Chicago, IL, USA
| | - Mark A. Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zijing Ye
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Balaji B. Ganesh
- Research Resources Center, University of Illinois Chicago, Chicago, Illinois, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois College of Medicine, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
11
|
Wang X, Peng Z, Wang L, Zhang J, Zhang K, Guo Z, Xu G, Li J. Cordyceps militaris Solid Medium Extract Alleviates Lipoteichoic Acid-Induced MH-S Inflammation by Inhibiting TLR2/NF-κB/NLRP3 Pathways. Int J Mol Sci 2023; 24:15519. [PMID: 37958501 PMCID: PMC10648577 DOI: 10.3390/ijms242115519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/19/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023] Open
Abstract
The aim of this study was to investigate the inhibitory effects of Cordyceps militaris solid medium extract (CME) and cordycepin (COR) on LTA-induced inflammation in MH-S cells and their mechanisms of action. In this study, the establishment of an LTA-induced MH-S inflammation model was determined, the CCK-8 method was used to determine the safe concentration range for a drug for COR and CME, the optimal concentration of COR and CME to exert anti-inflammatory effects was further selected, and the expression of inflammatory factors of TNF-α, IL-1β, IL-18, and IL-6 was detected using ELISA. The relative expression of TNF-α, IL-1β, IL-18, IL-6, IL-10, TLR2 and MyD88 mRNA was detected using RT-PCR, and the IL-1β, IL-18, TLR2, MyD88, NF-κB p-p65, NLRP3, pro-caspase-1, Caspase-1 and ASC protein expression in the cells were detected using Western blot; immunofluorescence assay detected the expression of Caspase-1 in MH-S cells. The results revealed that both CME and COR inhibited the levels of IL-1β, IL-18, IL-6, and TNF-α in the supernatants of LTA-induced MH-S cells and the mRNA expression levels of IL-1β, IL-18, IL-6, TNF-α, TLR2 and MyD88, down-regulated the LTA-induced IL-1β, IL-18, TLR2 in MH-S cells, MyD88, NF-κB p-p65/p65, NLRP3, ASC, pro-caspase-1, and caspase-1 protein expression levels, and inhibited LTA-induced caspase-1 activation in MH-S cells. In conclusion, CME can play a therapeutic role in LTA-induced inflammation in MH-S cells via TLR2/NF-κB/NLRP3, and may serve as a potential drug for bacterial pneumonia caused by Gram-positive bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jianxi Li
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| |
Collapse
|
12
|
Kawka M, Płocińska R, Płociński P, Pawełczyk J, Słomka M, Gatkowska J, Dzitko K, Dziadek B, Dziadek J. The functional response of human monocyte-derived macrophages to serum amyloid A and Mycobacterium tuberculosis infection. Front Immunol 2023; 14:1238132. [PMID: 37781389 PMCID: PMC10540855 DOI: 10.3389/fimmu.2023.1238132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction In the course of tuberculosis (TB), the level of major acute phase protein, namely serum amyloid A (hSAA-1), increases up to a hundredfold in the pleural fluids of infected individuals. Tubercle bacilli infecting the human host can be opsonized by hSAA-1, which affects bacterial entry into human macrophages and their intracellular multiplication. Methods We applied global RNA sequencing to evaluate the functional response of human monocyte-derived macrophages (MDMs), isolated from healthy blood donors, under elevated hSAA-1 conditions and during infection with nonopsonized and hSAA-1-opsonized Mycobacterium tuberculosis (Mtb). In the same infection model, we also examined the functional response of mycobacteria to the intracellular environment of macrophages in the presence and absence of hSAA-1. The RNASeq analysis was validated using qPCR. The functional response of MDMs to hSAA-1 and/or tubercle bacilli was also evaluated for selected cytokines at the protein level by applying the Milliplex system. Findings Transcriptomes of MDMs cultured in the presence of hSAA-1 or infected with Mtb showed a high degree of similarity for both upregulated and downregulated genes involved mainly in processes related to cell division and immune response, respectively. Among the most induced genes, across both hSAA-1 and Mtb infection conditions, CXCL8, CCL15, CCL5, IL-1β, and receptors for IL-7 and IL-2 were identified. We also observed the same pattern of upregulated pro-inflammatory cytokines (TNFα, IL-6, IL-12, IL-18, IL-23, and IL-1) and downregulated anti-inflammatory cytokines (IL-10, TGFβ, and antimicrobial peptide cathelicidin) in the hSAA-1 treated-MDMs or the phagocytes infected with tubercle bacilli. At this early stage of infection, Mtb genes affected by the inside microenvironment of MDMs are strictly involved in iron scavenging, adaptation to hypoxia, low pH, and increasing levels of CO2. The genes for the synthesis and transport of virulence lipids, but not cholesterol/fatty acid degradation, were also upregulated. Conclusion Elevated serum hSAA-1 levels in tuberculosis enhance the response of host phagocytes to infection, including macrophages that have not yet been in contact with mycobacteria. SAA induces antigen processing and presentation processes by professional phagocytes reversing the inhibition caused by Mtb infection.
Collapse
Affiliation(s)
- Malwina Kawka
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Renata Płocińska
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | | | - Jakub Pawełczyk
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Marcin Słomka
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Justyna Gatkowska
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Katarzyna Dzitko
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Bożena Dziadek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jarosław Dziadek
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
13
|
Park EJ, Yang MJ, Kang MS, Jo YM, Yoon C, Kim HB, Kim DW, Lee GH, Kwon IH, Park HJ, Kim JB. Subway station dust-induced pulmonary inflammation may be due to the dysfunction of alveolar macrophages: Possible contribution of bound elements. Toxicology 2023; 496:153618. [PMID: 37611816 DOI: 10.1016/j.tox.2023.153618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/09/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023]
Abstract
With its increasing value as a means of public transportation, the health effects of the air in subway stations have attracted public concern. In the current study, we investigated the pulmonary toxicity of dust collected from an air purifier installed on the platform of the busiest subway station in Seoul. We found that the dust contained various elements which are attributable to the facilities and equipment used to operate the subway system. Particularly, iron (Fe), chromium (Cr), zirconium (Zr), barium (Ba), and molybdenum (Mo) levels were more notable in comparison with those in dust collected from the ventilation chamber of a subway station. To explore the health effects of inhaled dust, we first instilled via the trachea in ICR mice for 13 weeks. The total number of pulmonary macrophages increased significantly with the dose, accompanying hematological changes. Dust-laden alveolar macrophages and inflammatory cells accumulated in the perivascular regions in the lungs of the treated mice, and pulmonary levels of CXCL-1, TNF-α, and TGF-β increased clearly compared with the control. The CCR5 and CD54 level expressed on BAL cell membranes was also enhanced following exposure to dust, whereas the CXCR2 level tended to decrease in the same samples. In addition, we treated the dust to alveolar macrophages (known as dust cells), lysosomal and mitochondrial function decreased, accompanied by cell death, and NO production was rapidly elevated with concentration. Moreover, the expression of autophagy- (p62) and anti-oxidant (SOD-2)-related proteins increased, and the expression of inflammation-related genes was dramatically up-regulated in the dust-treated cells. Therefore, we suggest that dysfunction of alveolar macrophages may importantly contribute to dust-induced inflammatory responses and that the exposure concentrations of Cr, Fe, Mo, Zr, and Ba should be considered carefully when assessing the health risks associated with subway dust. We also hypothesize that the bound elements may contribute to dust-induced macrophage dysfunction by inhibiting viability.
Collapse
Affiliation(s)
- Eun-Jung Park
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea.
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Min-Sung Kang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea; Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Young-Min Jo
- Department of Environmental Science and Engineering, Global Campus, Kyung Hee University, 17104, Republic of Korea
| | - Cheolho Yoon
- Ochang Center, Korea Basic Science Institute, 28119, Republic of Korea
| | - Hyun-Bin Kim
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Dong-Wan Kim
- School of Civil, Environmental and Architectural Engineering, Korea University, 02841, Republic of Korea
| | - Gwang-Hee Lee
- School of Civil, Environmental and Architectural Engineering, Korea University, 02841, Republic of Korea
| | - Ik-Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 34113, Republic of Korea
| | - Hee-Jin Park
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, Kyung-Hee University Hospital, Kyung Hee University, 02447, Republic of Korea.
| |
Collapse
|
14
|
Ng LG, Liu Z, Kwok I, Ginhoux F. Origin and Heterogeneity of Tissue Myeloid Cells: A Focus on GMP-Derived Monocytes and Neutrophils. Annu Rev Immunol 2023; 41:375-404. [PMID: 37126421 DOI: 10.1146/annurev-immunol-081022-113627] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Myeloid cells are a significant proportion of leukocytes within tissues, comprising granulocytes, monocytes, dendritic cells, and macrophages. With the identification of various myeloid cells that perform separate but complementary functions during homeostasis and disease, our understanding of tissue myeloid cells has evolved significantly. Exciting findings from transcriptomics profiling and fate-mapping mouse models have facilitated the identification of their developmental origins, maturation, and tissue-specific specializations. This review highlights the current understanding of tissue myeloid cells and the contributing factors of functional heterogeneity to better comprehend the complex and dynamic immune interactions within the healthy or inflamed tissue. Specifically, we discuss the new understanding of the contributions of granulocyte-monocyte progenitor-derived phagocytes to tissue myeloid cell heterogeneity as well as the impact of niche-specific factors on monocyte and neutrophil phenotype and function. Lastly, we explore the developing paradigm of myeloid cell heterogeneity during inflammation and disease.
Collapse
Affiliation(s)
- Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institut Gustave Roussy, INSERM U1015, Villejuif, France
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| |
Collapse
|
15
|
Ma X, Liu Z, Yu Y, Jiang Y, Wang C, Zuo Z, Ling S, He M, Cao S, Wen Y, Zhao Q, Wu R, Huang X, Zhong Z, Peng G, Gu Y. Microsporum gypseum Isolated from Ailuropoda melanoleuca Provokes Inflammation and Triggers Th17 Adaptive Immunity Response. Int J Mol Sci 2022; 23:ijms231912037. [PMID: 36233337 PMCID: PMC9570494 DOI: 10.3390/ijms231912037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 12/02/2022] Open
Abstract
Microsporum gypseum causes dermatomycoses in giant pandas (Ailuropoda melanoleuca). This study aimed to investigate the immune response of M. gypseum following deep infection. The degree of damage to the heart, liver, spleen, lungs, and kidneys was evaluated using tissue fungal load, organ index, and histopathological methods. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) detected the mRNA expression of receptors and cytokines in the lung, and immunofluorescence staining and flow cytometry, were used to assess immune cells in the lung. The results indicated that conidia mainly colonized the lungs and caused serious injury with M. gypseum infection. Furthermore, dectin-1, TLR-2, and TLR-4 played a role in recognizing M. gypseum cells. Numerous inflammatory cells, mainly macrophages, dendritic cells, polymorphonuclear neutrophils, and inflammatory cytokines (TGF-β, TNF-α, IL-1β, IL-6, IL-10, IL-12, and IL-23), were activated in the early stages of infection. With the high expression of IL-22, IL-17A, and IL-17F, the Th17 pathway exerted an adaptive immune response to M. gypseum infection. These results can potentially aid in the diagnosis and treatment of diseases caused by M. gypseum in giant pandas.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhen Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yaozhang Jiang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shanshan Ling
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Ming He
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- China Conservation and Research Center for the Giant Panda, Chengdu 611800, China
| | - Sanjie Cao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaobo Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: ; Tel.: +86-18190681226
| |
Collapse
|
16
|
Zaki MSA, El-Kott AF, AlGwaiz HIM, Shehata SF, Eldeen MA, Andarawi M, Eid RA, Abd-Ella EM. The possible effects of α-tocopherol against amiodarone-treated lungs in rats: vimentin detection, lipid peroxidation assay, and histological and ultrastructural evaluations. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:58231-58239. [PMID: 35366726 DOI: 10.1007/s11356-022-19883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/20/2022] [Indexed: 06/14/2023]
Abstract
The purpose of this study was to learn more about the pathogenesis of amiodarone (AD) on alveoli and also the possible preventive effect of α-tocopherol (α-T) against these hazards. Rats were divided into 4 groups, one of which acted as a control, the second received α-T, the third AD, and the fourth AD and α-T for 2 weeks. Light microscopy (LM), immunohistochemistry, transmission electron microscopy (TEM), and malondialdehyde (MDA) activity were analyzed in sections of lung tissue. Alveoli of lung tissue AD examined with LM showed dilatation of alveolar spaces, aggregation of red blood cells, and narrowing of alveolar septa. When stained with vimentin (VIM), alveoli showed a positive reaction in the majority and a moderate reaction in others. In the pneumocytes of the type II, some cytoplasmic vesicles had been deflated, whereas others contained lamellar bodies, a damaged nucleus, and vesicles in their heterochromatin. In the interstitial space, collagen fibers with aggregation of red blood cells and a disrupted blood-air barrier were detected. In rat lung alveoli treated with AD and α-T, the alveolar septum thickened and the alveolar spaces expanded as estimated. The alveoli of this group had more or less intact type I and II pneumocytes and a better appearance of the blood-air barrier. In the cells of the alveolar lining, the VIM staining leads to a diffuse positive response. Finally, lung parenchyma also improved, suggesting that α-T may help minimize the effects of AD.
Collapse
Affiliation(s)
- Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia.
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig, 31527, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Hussah I M AlGwaiz
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11474, Saudi Arabia
| | - Shehata F Shehata
- Department of Family and Community Medicine, King Khalid University, Abha, Saudi Arabia
- Biostatistics Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Muhammad Alaa Eldeen
- Biology Department, Physiology Section, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Mohamed Andarawi
- Department of Pathology, College of Medicine, King Khalid University, P.O. 641, Abha, 62529, Saudi Arabia
| | - Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, P.O. 641, Abha, 62529, Saudi Arabia.
| | - Eman M Abd-Ella
- Department of Zoology, College of Science, Fayoum University, Fayoum, Egypt
- Department of Biology, College of Science and Art, Al-Baha University, Al-Mandaq, Saudi Arabia
| |
Collapse
|
17
|
Furlong-Silva J, Cook PC. Fungal-mediated lung allergic airway disease: The critical role of macrophages and dendritic cells. PLoS Pathog 2022; 18:e1010608. [PMID: 35834490 PMCID: PMC9282651 DOI: 10.1371/journal.ppat.1010608] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Fungi are abundant in the environment, causing our lungs to be constantly exposed to a diverse range of species. While the majority of these are cleared effectively in healthy individuals, constant exposure to spores (especially Aspergillus spp.) can lead to the development of allergic inflammation that underpins and worsen diseases such as asthma. Despite this, the precise mechanisms that underpin the development of fungal allergic disease are poorly understood. Innate immune cells, such as macrophages (MΦs) and dendritic cells (DCs), have been shown to be critical for mediating allergic inflammation to a range of different allergens. This review will focus on the crucial role of MΦ and DCs in mediating antifungal immunity, evaluating how these immune cells mediate allergic inflammation within the context of the lung environment. Ultimately, we aim to highlight important future research questions that will lead to novel therapeutic strategies for fungal allergic diseases.
Collapse
Affiliation(s)
- Julio Furlong-Silva
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Peter Charles Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
Bouch S, Litvack ML, Litman K, Luo L, Post A, Williston E, Park AJ, Roach EJ, Berezuk AM, Khursigara CM, Post M. Therapeutic stem cell-derived alveolar-like macrophages display bactericidal effects and resolve Pseudomonas aeruginosa-induced lung injury. J Cell Mol Med 2022; 26:3046-3059. [PMID: 35441437 PMCID: PMC9097833 DOI: 10.1111/jcmm.17324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 12/24/2021] [Accepted: 01/28/2022] [Indexed: 01/19/2023] Open
Abstract
Bacterial lung infections lead to greater than 4 million deaths per year with antibiotic treatments driving an increase in antibiotic resistance and a need to establish new therapeutic approaches. Recently, we have generated mouse and rat stem cell‐derived alveolar‐like macrophages (ALMs), which like primary alveolar macrophages (1'AMs), phagocytose bacteria and promote airway repair. Our aim was to further characterize ALMs and determine their bactericidal capabilities. The characterization of ALMs showed that they share known 1'AM cell surface markers, but unlike 1'AMs are highly proliferative in vitro. ALMs effectively phagocytose and kill laboratory strains of P. aeruginosa (P.A.), E. coli (E.C.) and S. aureus, and clinical strains of P.A. In vivo, ALMs remain viable, adapt additional features of native 1'AMs, but proliferation is reduced. Mouse ALMs phagocytose P.A. and E.C. and rat ALMs phagocytose and kill P.A. within the lung 24 h post‐instillation. In a pre‐clinical model of P.A.‐induced lung injury, rat ALM administration mitigated weight loss and resolved lung injury observed seven days post‐instillation. Collectively, ALMs attenuate pulmonary bacterial infections and promote airway repair. ALMs could be utilized as an alternative or adjuvant therapy where current treatments are ineffective against antibiotic‐resistant bacteria or to enhance routine antibiotic delivery.
Collapse
Affiliation(s)
- Sheena Bouch
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael L Litvack
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kymberly Litman
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, The University of Toronto, Toronto, Ontario, Canada
| | - Lisha Luo
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alex Post
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emma Williston
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amber J Park
- Department of Molecular and Cellular Biology, The University of Guelph, Ontario, Canada
| | - Elyse J Roach
- Department of Molecular and Cellular Biology, The University of Guelph, Ontario, Canada
| | - Alison M Berezuk
- Department of Molecular and Cellular Biology, The University of Guelph, Ontario, Canada
| | - Cezar M Khursigara
- Department of Molecular and Cellular Biology, The University of Guelph, Ontario, Canada
| | - Martin Post
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Aerosol-Cell Exposure System Applied to Semi-Adherent Cells for Aerosolization of Lung Surfactant and Nanoparticles Followed by High Quality RNA Extraction. NANOMATERIALS 2022; 12:nano12081362. [PMID: 35458071 PMCID: PMC9028274 DOI: 10.3390/nano12081362] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023]
Abstract
Nanoparticle toxicity assessments have moved closer to physiological conditions while trying to avoid the use of animal models. An example of new in vitro exposure techniques developed is the exposure of cultured cells at the air-liquid interface (ALI), particularly in the case of respiratory airways. While the commercially available VITROCELL® Cloud System has been applied for the delivery of aerosolized substances to adherent cells under ALI conditions, it has not yet been tested on lung surfactant and semi-adherent cells such as alveolar macrophages, which are playing a pivotal role in the nanoparticle-induced immune response. OBJECTIVES In this work, we developed a comprehensive methodology for coating semi-adherent lung cells cultured at the ALI with aerosolized surfactant and subsequent dose-controlled exposure to nanoparticles (NPs). This protocol is optimized for subsequent transcriptomic studies. METHODS Semi-adherent rat alveolar macrophages NR8383 were grown at the ALI and coated with lung surfactant through nebulization using the VITROCELL® Cloud 6 System before being exposed to TiO2 NM105 NPs. After NP exposures, RNA was extracted and its quantity and quality were measured. RESULTS The VITROCELL® Cloud system allowed for uniform and ultrathin coating of cells with aerosolized surfactant mimicking physiological conditions in the lung. While nebulization of 57 μL of 30 mg/mL TiO2 and 114 μL of 15 mg/mL TiO2 nanoparticles yielded identical cell delivered dose, the reproducibility of dose as well as the quality of RNA extracted were better for 114 μL.
Collapse
|
20
|
Gorki AD, Symmank D, Zahalka S, Lakovits K, Hladik A, Langer B, Maurer B, Sexl V, Kain R, Knapp S. Murine Ex Vivo Cultured Alveolar Macrophages Provide a Novel Tool to Study Tissue-Resident Macrophage Behavior and Function. Am J Respir Cell Mol Biol 2022; 66:64-75. [PMID: 34586974 PMCID: PMC8803354 DOI: 10.1165/rcmb.2021-0190oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/28/2021] [Indexed: 11/24/2022] Open
Abstract
Tissue-resident macrophages are of vital importance as they preserve tissue homeostasis in all mammalian organs. Nevertheless, appropriate cell culture models are still limited. Here, we propose a novel culture model to study and expand murine primary alveolar macrophages (AMs), the tissue-resident macrophages of the lung, in vitro over several months. By providing a combination of granulocyte-macrophage colony-stimulating factor, TGFβ, and the PPARγ activator rosiglitazone, we maintain and expand mouse ex vivo cultured AMs (mexAMs) over several months. MexAMs maintain typical morphologic features and stably express primary AM surface markers throughout in vitro culture. They respond to microbial ligands and exhibit an AM-like transcriptional profile, including the expression of AM-specific transcription factors. Furthermore, when transferred into AM-deficient mice, mexAMs efficiently engraft in the lung and fulfill key macrophage functions, leading to a significantly reduced surfactant load in those mice. Altogether, mexAMs provide a novel, simple, and versatile tool to study AM behavior in homeostasis and disease settings.
Collapse
Affiliation(s)
- Anna-Dorothea Gorki
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| | - Dörte Symmank
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| | - Sophie Zahalka
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| | - Karin Lakovits
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| | - Anastasiya Hladik
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| | - Brigitte Langer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Research Laboratory of Infection Biology, Department of Medicine I, and
- Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; and
| |
Collapse
|
21
|
Yang Y, Wang Y. Autocrine, Paracrine, and Endocrine Signals That Can Alter Alveolar Macrophages Function. Rev Physiol Biochem Pharmacol 2022; 186:177-198. [PMID: 36472676 DOI: 10.1007/112_2022_76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar macrophages (AMs) are extremely versatile cells with complex functions involved in health or diseases such as pneumonia, asthma, and pulmonary alveolar proteinosis. In recent years, it has been widely identified that the different functions and states of macrophages are the results from the complex interplay between microenvironmental signals and macrophage lineage. Diverse and complicated signals to which AMs respond are mentioned when they are described individually or in a particular state of AMs. In this review, the microenvironmental signals are divided into autocrine, paracrine, and endocrine signals based on their secreting characteristics. This new perspective on classification provides a more comprehensive and systematic introduction to the complex signals around AMs and is helpful for understanding the roles of AMs affected by physiological environment. The existing possible treatments of AMs are also mentioned in it. The thorough understanding of AMs signals modulation may be contributed to the development of more effective therapies for AMs-related lung diseases.
Collapse
Affiliation(s)
- Yue Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
22
|
Hendrix DA, Hurowitz JA, Glotch TD, Schoonen MAA. Olivine Dissolution in Simulated Lung and Gastric Fluid as an Analog to the Behavior of Lunar Particulate Matter Inside the Human Respiratory and Gastrointestinal Systems. GEOHEALTH 2021; 5:e2021GH000491. [PMID: 34849441 PMCID: PMC8609536 DOI: 10.1029/2021gh000491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 06/13/2023]
Abstract
With the Artemis III mission scheduled to land humans on the Moon in 2025, work must be done to understand the hazards lunar dust inhalation would pose to humans. In this study, San Carlos olivine was used as an analog of lunar olivine, a common component of lunar dust. Olivine was dissolved in a flow-through apparatus in both simulated lung fluid and 0.1 M HCl (simulated gastric fluid) over a period of approximately 2 weeks at physiological temperature, 37°C. Effluent samples were collected periodically and analyzed for pH, iron, silicon, and magnesium ion concentrations. The dissolution rate data derived from our measurements allow us to estimate that an inhaled 1.0 μm diameter olivine particle would take approximately 24 years to dissolve in the human lungs and approximately 3 weeks to dissolve in gastric fluid. Results revealed that inhaled olivine particles may generate the toxic chemical, hydroxyl radical, for up to 5-6 days in lung fluid. Olivine dissolved in 0.1 M HCl for 2 weeks transformed to an amorphous silica-rich solid plus the ferric iron oxy-hydroxide ferrihydrite. Olivine dissolved in simulated lung fluid shows no detectable change in composition or crystallinity. Equilibrium thermodynamic models indicate that olivine in the human lungs can precipitate secondary minerals with fibrous crystal structures that have the potential to induce detrimental health effects similar to asbestos exposure. Our work indicates that inhaled lunar dust containing olivine can settle in the human lungs for years and could induce long-term potential health effects like that of silicosis.
Collapse
Affiliation(s)
| | | | | | - Martin A. A. Schoonen
- Environment, Biology, Nuclear Science, & NonproliferationBrookhaven National LaboratoryUptonNYUSA
| |
Collapse
|
23
|
LIGHT of pulmonary NKT cells annihilates tissue protective alveolar macrophages in augmenting severe influenza pneumonia. Sci Bull (Beijing) 2021; 66:2124-2134. [PMID: 36654270 DOI: 10.1016/j.scib.2021.01.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/20/2020] [Accepted: 01/19/2021] [Indexed: 02/03/2023]
Abstract
CD1d-restricted natural killer T (NKT) cells are innate-like T lymphocytes with protective or pathogenic roles in the development of influenza pneumonia. Here, we show that lung-infiltrated and activated NKT cells are the major cellular source of LIGHT/TNFSF14, which determines the severity of pulmonary pneumonia by highly deteriorative influenza A virus (IAV) infection. Compared to wild-type mice, LIGHT-/- mice exhibit much lower morbidity and mortality to IAV, due to alleviated lung damage and reduced apoptosis of alveolar macrophages (AMs). LIGHT preferentially promotes cell death of lymphotoxin β receptors positive (LTβR+) AMs but not herpesvirus entry mediator positive (HVEM+) AMs. Therefore, these results suggest that NKT-derived LIGHT augments cell death of the tissue protective AMs in exacerbating lung pathology and susceptibility to fatal influenza infection. Suppression of LIGHT signaling might be a viable option in the treatment of influenza-associated acute respiratory distress syndrome.
Collapse
|
24
|
Knoll R, Schultze JL, Schulte-Schrepping J. Monocytes and Macrophages in COVID-19. Front Immunol 2021; 12:720109. [PMID: 34367190 PMCID: PMC8335157 DOI: 10.3389/fimmu.2021.720109] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a contagious viral disease caused by SARS-CoV-2 that led to an ongoing pandemic with massive global health and socioeconomic consequences. The disease is characterized primarily, but not exclusively, by respiratory clinical manifestations ranging from mild common cold symptoms, including cough and fever, to severe respiratory distress and multi-organ failure. Macrophages, a heterogeneous group of yolk-sac derived, tissue-resident mononuclear phagocytes of complex ontogeny present in all mammalian organs, play critical roles in developmental, homeostatic and host defense processes with tissue-dependent plasticity. In case of infection, they are responsible for early pathogen recognition, initiation and resolution of inflammation, as well as repair of tissue damage. Monocytes, bone-marrow derived blood-resident phagocytes, are recruited under pathological conditions such as viral infections to the affected tissue to defend the organism against invading pathogens and to aid in efficient resolution of inflammation. Given their pivotal function in host defense and the potential danger posed by their dysregulated hyperinflammation, understanding monocyte and macrophage phenotypes in COVID-19 is key for tackling the disease's pathological mechanisms. Here, we outline current knowledge on monocytes and macrophages in homeostasis and viral infections and summarize concepts and key findings on their role in COVID-19. While monocytes in the blood of patients with moderate COVID-19 present with an inflammatory, interferon-stimulated gene (ISG)-driven phenotype, cellular dysfunction epitomized by loss of HLA-DR expression and induction of S100 alarmin expression is their dominant feature in severe disease. Pulmonary macrophages in COVID-19 derived from infiltrating inflammatory monocytes are in a hyperactivated state resulting in a detrimental loop of pro-inflammatory cytokine release and recruitment of cytotoxic effector cells thereby exacerbating tissue damage at the site of infection.
Collapse
Affiliation(s)
- Rainer Knoll
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L. Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Jonas Schulte-Schrepping
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
25
|
Quan YH, Oh CH, Jung D, Lim JY, Choi BH, Rho J, Choi Y, Han KN, Kim BM, Kim C, Park JH, Kim HK. Evaluation of Intraoperative Near-Infrared Fluorescence Visualization of the Lung Tumor Margin With Indocyanine Green Inhalation. JAMA Surg 2021; 155:732-740. [PMID: 32579150 DOI: 10.1001/jamasurg.2020.1314] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Importance Identification of the tumor margin during surgery is important for precise minimal resection of lung tumors. Intravenous injection of indocyanine green (ICG) has several limitations when used for intraoperative visualization of lung cancer. Objectives To describe a technique for intraoperative visualization of lung tumor margin using ICG inhalation and evaluate the clinical applicability of the technique in mouse and rabbit lung tumor models as well as lung specimens of patients with lung tumors. Design, Setting, and Participants In lung tumor models of both mice and rabbits, the distribution of inhaled ICG in the lung tumor margin was investigated in vivo and ex vivo using a near-infrared imaging system. Lung tumor margin detection via inhalation of ICG was evaluated by comparing the results obtained with those of the intravenous injection method (n = 32, each time point for 4 mice). Based on preclinical data, use of ICG inhalation to help detect the tumor margin in patients with lung cancer was also evaluated (n = 6). This diagnostic study was conducted from May 31, 2017, to March 30, 2019. Main Outcomes and Measures The use of tumor margin detection by inhaled ICG was evaluated by comparing the inhaled formulation with intravenous administration of ICG. Results From 10 minutes after inhalation of ICG to 24 hours, the distribution of ICG in the lungs was significantly higher than that in other organs (signal to noise ratio in the lungs: 39 486.4; interquartile range [IQR], 36 983.74-43 592.5). Ex vivo and histologic analysis showed that, in both lung tumor models, inhaled ICG was observed throughout the healthy lung tissue but was rarely found in tumor tissue. The difference in the fluorescent signal between healthy and tumor lung tissues was associated with the mechanical airway obstruction caused by the tumor and with alveolar macrophage uptake of the inhaled ICG in healthy tissues. Inhalation at a 20-fold lower dose of ICG had a 2-fold higher efficiency for tumor margin detection than did the intravenous injection (2.9; IQR, 2.7-3.2; P < .001). Conclusions and Relevance The results of this study suggest that lung-specific inhalation delivery of ICG is feasible and may be useful for the intraoperative visualization of lung tumor margin in clinical practice.
Collapse
Affiliation(s)
- Yu Hua Quan
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Chan Hee Oh
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Daeho Jung
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ji-Young Lim
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jiyun Rho
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yeonho Choi
- Department of Bio-Convergence, Korea University, Seoul, Republic of Korea
| | - Kook Nam Han
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Beop-Min Kim
- Department of Bio-Convergence, Korea University, Seoul, Republic of Korea
| | - Chungyeul Kim
- Department of Pathology, Korea University Guro Hospital, College of Medicine, Republic of Korea University, Seoul, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Is There An Explanation for How An Irritant Causes A Nonallergic Asthmatic Disorder Such as Reactive Airways Dysfunction Syndrome (RADS)? J Occup Environ Med 2021; 62:e139-e141. [PMID: 31934909 DOI: 10.1097/jom.0000000000001814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
L Dragan A, E Voth D. Take my breath away: studying pathogen invasion of the human lung using primary tissue models. Pathog Dis 2021; 79:6177680. [PMID: 33734371 DOI: 10.1093/femspd/ftab016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
The human pulmonary environment is complex, containing a matrix of cells, including fibroblasts, epithelial cells, interstitial macrophages, alveolar macrophages and neutrophils. When confronted with foreign material or invading pathogens, these cells mount a robust response. Nevertheless, many bacterial pathogens with an intracellular lifecycle stage exploit this environment for replication and survival. These include, but are not limited to, Coxiella burnetii, Legionella pneumophila, Yersinia pestis, Mycobacterium tuberculosis and Staphylococcus aureus. Currently, few human disease-relevant model systems exist for studying host-pathogen interactions during these bacterial infections in the lung. Here, we present two novel infection platforms, human alveolar macrophages (hAMs) and human precision-cut lung slices (hPCLS), along with an up-to-date synopsis of research using said models. Additionally, alternative uses for these systems in the absence of pathogen involvement are presented, such as tissue banking and further characterization of the human lung environment. Overall, hAMs and hPCLS allow novel human disease-relevant investigations that other models, such as cell lines and animal models, cannot completely provide.
Collapse
Affiliation(s)
- Amanda L Dragan
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
28
|
Ma X, Hu J, Yu Y, Wang C, Gu Y, Cao S, Huang X, Wen Y, Zhao Q, Wu R, Zuo Z, Deng J, Ren Z, Yu S, Shen L, Zhong Z, Peng G. Assessment of the pulmonary adaptive immune response to Cladosporium cladosporioides infection using an experimental mouse model. Sci Rep 2021; 11:909. [PMID: 33441700 PMCID: PMC7806624 DOI: 10.1038/s41598-020-79642-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/10/2020] [Indexed: 01/29/2023] Open
Abstract
Cladosporium cladosporioides causes asthma and superficial and deep infections, mostly in immunodeficient individuals and animals. This study aimed to investigate whether C. cladosporioides spores can enter the lungs through pulmonary circulation and influence pulmonary immune response. We intravenously injected mice with C. cladosporioides spore suspension and conducted several assays on the lungs. Pulmonary hemorrhage symptoms and congestion were most severe on days 1, 2, and 3 post-inoculation (PI). Extensive inflammatory cell infiltration occurred throughout the period of infection. More spores and hyphae colonizing the lungs were detected on days 1, 2, and 3 PI, and fewer spores and hyphae were observed within 21 d of infection. Numerous macrophages, dendritic cells, and neutrophils were observed on day 5 PI, along with upregulation of CD54, an intercellular adhesion molecule. Th1 and Th2 cells increased after infection; specifically, Th2 cells increased considerably on day 5 PI. These results suggest that days 2 and 5 PI represent the inflammatory peak in the lungs and that the Th2 and Th1 signaling pathways are potentially involved in pulmonary immune responses. In conclusion, the further adaptive immune responses played important roles in establishing effective pulmonary immunity against C. cladosporioides systemic infections based on innate immune responses.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chengdong Wang
- China Conservation and Research Center for the Giant Panda, Chengdu, 611800, Sichuan, China.
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Sanjie Cao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaobo Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yiping Wen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qin Zhao
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rui Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhihua Ren
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shumin Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liuhong Shen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| |
Collapse
|
29
|
Macrophage metabolic reprogramming during chronic lung disease. Mucosal Immunol 2021; 14:282-295. [PMID: 33184475 PMCID: PMC7658438 DOI: 10.1038/s41385-020-00356-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/24/2020] [Indexed: 02/04/2023]
Abstract
Airway macrophages (AMs) play key roles in the maintenance of lung immune tolerance. Tissue tailored, highly specialised and strategically positioned, AMs are critical sentinels of lung homoeostasis. In the last decade, there has been a revolution in our understanding of how metabolism underlies key macrophage functions. While these initial observations were made during steady state or using in vitro polarised macrophages, recent studies have indicated that during many chronic lung diseases (CLDs), AMs adapt their metabolic profile to fit their local niche. By generating reactive oxygen species (ROS) for pathogen defence, utilising aerobic glycolysis to rapidly generate cytokines, and employing mitochondrial respiration to fuel inflammatory responses, AMs utilise metabolic reprogramming for host defence, although these changes may also support chronic pathology. This review focuses on how metabolic alterations underlie AM phenotype and function during CLDs. Particular emphasis is given to how our new understanding of AM metabolic plasticity may be exploited to develop AM-focused therapies.
Collapse
|
30
|
Yukina GY, Polovnikov IV, Sukhorukova EG, Zhuravskii SG, Galagudza MM. Morphological Analysis of the Respiratory Tract of Rats after Parenteral Administration of Silicon Dioxide Nanoparticles. Bull Exp Biol Med 2020; 170:93-97. [PMID: 33231801 DOI: 10.1007/s10517-020-05011-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Indexed: 11/28/2022]
Abstract
Morphological analysis of the respiratory tract of Wistar rats was performed after a single parenteral administration of 12-nm silicon dioxide nanoparticles (1 ml, 2 mg/ml, intravenously) was performed. On day 21 and in 2, 4, and 6 months after the administration of nanoparticles, the development of macrophage infiltration in the interstitium of the respiratory tract was demonstrated by histological and immunohistochemical methods. The pool of alveolar macrophages increased in 4 months after administration (p=0.004) and returned to the control values in 6 months. The number of mast cells did not significantly change at all stages of the experiment. Connective tissue remodeling in the interstitium of the respiratory tract was not observed throughout the observation period.
Collapse
Affiliation(s)
- G Yu Yukina
- I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
| | - I V Polovnikov
- I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
| | - E G Sukhorukova
- I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.
| | - S G Zhuravskii
- I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.,V. A. Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - M M Galagudza
- I. P. Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.,V. A. Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| |
Collapse
|
31
|
Different Sensitivity of Macrophages to Phospholipidosis Induction by Amphiphilic Cationic Drugs. Int J Mol Sci 2020; 21:ijms21218391. [PMID: 33182310 PMCID: PMC7664898 DOI: 10.3390/ijms21218391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
Phospholipidosis (PLD), the intracellular accumulation of phospholipids, is an adaptive response to toxic stimuli and serves as an important parameter in the biological assessment of compounds. Cationic amphiphilic drugs are the main inducers of PLD and may impair the function of alveolar macrophages. In vivo and in vitro models are used for PLD screening but the choice of the cellular model may be important because PLD develops in a cell- and species-specific manner. In this study, a panel of different staining (LysoSensor, Acridine Orange, Nile Red, HCS LipidTOX, LysoID) was evaluated in murine (DMBM-2, J774, RAW264.7) and human (THP-1, monocyte-derived macrophages from peripheral blood) cells to identify the most sensitive and easy to analyze staining method and to detect species-specific differences in the reaction pattern. Amiodarone and chloroquine served as inducers of PLD. High content screening was used to compare number, area, and intensity of the staining. Due to the fast staining protocol and the sensitivity of the detection, LysoID proved to be the most suitable dye of the testing. The lower induction of PLD by chloroquine reported in vivo was also seen in this study. THP-1 macrophages, followed by DMBM-2 cells, produced the most similar reaction pattern to human monocyte-derived macrophages.
Collapse
|
32
|
Silla L. Double-bright (CD56bright/CD16bright) natural killer cell adoptive immunotherapy for SARS-CoV-2. Br J Haematol 2020; 190:e322-e323. [PMID: 32645204 PMCID: PMC7404630 DOI: 10.1111/bjh.17010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Lúcia Silla
- Hematologia e Hemoterapia, Hospital de Clínicas, Porto Alegre, Brazil
| |
Collapse
|
33
|
Chang D, Feng J, Liu H, Liu W, Sharma L, Dela Cruz CS. Differential effects of the Akt pathway on the internalization of Klebsiella by lung epithelium and macrophages. Innate Immun 2020; 26:618-626. [PMID: 32762278 PMCID: PMC7556185 DOI: 10.1177/1753425920942582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Host response to lung infection includes coordinated efforts of multiple cell types, including the lung epithelium and macrophages. Importantly, both the lung epithelium and macrophages can internalize and clear invading pathogens. However, the mechanisms and their ability to internalize or phagocytose differ. Akt is a key cellular pathway that controls cell proliferation and survival, in addition to its role in host defense. The role of the Akt pathway was assessed using pharmacological Akt modulators in lung epithelial (A549) and macrophage (RAW 264.7) cell lines during Klebsiella bacterial infection. Our data show that the inhibition of the Akt pathway using specific Akt inhibitor MK2206 increased the phagocytic ability of lung epithelial cells but not of macrophages. In contrast, the activation of Akt using specific activator SC-79 decreased the phagocytic ability of epithelial cells, while it increased the phagocytic ability of macrophages. The altered phagocytic ability in both cell types using Akt modulators was not due to changes in bacterial adhesion to the host cell. The clinical usefulness of these Akt modulators may vary based on the type of infection and on the relative contribution of epithelial cells and macrophages in clearing the particular bacterial infection. The Akt pathway has differential roles in the internalization of Klebsiella bacteria by respiratory epithelial cells and immune cells.
Collapse
Affiliation(s)
- De Chang
- Third Medical Center of Chinese PLA General Hospital, PR China.,Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| | - Jingjing Feng
- Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| | - Hongbo Liu
- Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| | - Wei Liu
- Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| | - Lokesh Sharma
- Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Internal Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, USA
| |
Collapse
|
34
|
Pai S, Muruganandah V, Kupz A. What lies beneath the airway mucosal barrier? Throwing the spotlight on antigen-presenting cell function in the lower respiratory tract. Clin Transl Immunology 2020; 9:e1158. [PMID: 32714552 PMCID: PMC7376394 DOI: 10.1002/cti2.1158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of respiratory infectious and inflammatory diseases remains a major public health concern. Prevention and management strategies have not kept pace with the increasing incidence of these diseases. The airway mucosa is the most common portal of entry for infectious and inflammatory agents. Therefore, significant benefits would be derived from a detailed understanding of how immune responses regulate the filigree of the airways. Here, the role of different antigen‐presenting cells (APC) in the lower airways and the mechanisms used by pathogens to modulate APC function during infectious disease is reviewed. Features of APC that are unique to the airways and the influence they have on uptake and presentation of antigen to T cells directly in the airways are discussed. Current information on the crucial role that airway APC play in regulating respiratory infection is summarised. We examine the clinical implications of APC dysregulation in the airways on asthma and tuberculosis, two chronic diseases that are the major cause of illness and death in the developed and developing world. A brief overview of emerging therapies that specifically target APC function in the airways is provided.
Collapse
Affiliation(s)
- Saparna Pai
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| | - Visai Muruganandah
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| | - Andreas Kupz
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia
| |
Collapse
|
35
|
Somerville L, Cardani A, Braciale TJ. Alveolar Macrophages in Influenza A Infection Guarding the Castle with Sleeping Dragons. Infect Dis Ther 2020; 1. [PMID: 33681871 DOI: 10.31038/idt.2020114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Lindsay Somerville
- Pulmonary and Critical Care Medicine, University of Virginia Health System, Charlottesville, Virginia, United States of America.,Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - Amber Cardani
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America.,Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas J Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America.,Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America.,Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
36
|
Brown JS. Improving Pulmonary Immunity to Bacterial Pathogens through Streptococcus pneumoniae Colonization of the Nasopharynx. Am J Respir Crit Care Med 2020; 201:268-270. [PMID: 31664865 PMCID: PMC6999096 DOI: 10.1164/rccm.201910-2047ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jeremy S Brown
- UCL RespiratoryUniversity College LondonLondon, United Kingdom
| |
Collapse
|
37
|
Mitsi E, Carniel B, Reiné J, Rylance J, Zaidi S, Soares-Schanoski A, Connor V, Collins AM, Schlitzer A, Nikolaou E, Solórzano C, Pojar S, Hill H, Hyder-Wright AD, Jambo KC, Oggioni MR, De Ste Croix M, Gordon SB, Jochems SP, Ferreira DM. Nasal Pneumococcal Density Is Associated with Microaspiration and Heightened Human Alveolar Macrophage Responsiveness to Bacterial Pathogens. Am J Respir Crit Care Med 2020; 201:335-347. [PMID: 31626559 PMCID: PMC6999099 DOI: 10.1164/rccm.201903-0607oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Rationale: Pneumococcal pneumonia remains a global health problem. Colonization of the nasopharynx with Streptococcus pneumoniae (Spn), although a prerequisite of infection, is the main source of exposure and immunological boosting in children and adults. However, our knowledge of how nasal colonization impacts on the lung cells, especially on the predominant alveolar macrophage (AM) population, is limited.Objectives: Using a controlled human infection model to achieve nasal colonization with 6B serotype, we investigated the effect of Spn colonization on lung cells.Methods: We collected BAL from healthy pneumococcal-challenged participants aged 18-49 years. Confocal microscopy and molecular and classical microbiology were used to investigate microaspiration and pneumococcal presence in the lower airways. AM opsonophagocytic capacity was assessed by functional assays in vitro, whereas flow cytometry and transcriptomic analysis were used to assess further changes on the lung cellular populations.Measurements and Main Results: AMs from Spn-colonized individuals exhibited increased opsonophagocytosis to pneumococcus (11.4% median increase) for approximately 3 months after experimental pneumococcal colonization. AMs also had increased responses against other bacterial pathogens. Pneumococcal DNA detected in the BAL samples of Spn-colonized individuals were positively correlated with nasal pneumococcal density (r = 0.71; P = 0.029). Similarly, AM-heightened opsonophagocytic capacity was correlated with nasopharyngeal pneumococcal density (r = 0.61, P = 0.025).Conclusions: Our findings demonstrate that nasal colonization with pneumococcus and microaspiration prime AMs, leading to brisker responsiveness to both pneumococcus and unrelated bacterial pathogens. The relative abundance of AMs in the alveolar spaces, alongside their potential for nonspecific protection, render them an attractive target for novel vaccines.
Collapse
Affiliation(s)
- Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Beatriz Carniel
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jesús Reiné
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jamie Rylance
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Seher Zaidi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Victoria Connor
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andrea M. Collins
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andreas Schlitzer
- The Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Elissavet Nikolaou
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sherin Pojar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Helen Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Angela D. Hyder-Wright
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kondwani C. Jambo
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Trust Clinical Research Programme, College of Medicine, Chichiri, Blantyre, Malawi
| | - Marco R. Oggioni
- Department of Genetics, University of Leicester, Leicester, United Kingdom; and
| | - Megan De Ste Croix
- Department of Genetics, University of Leicester, Leicester, United Kingdom; and
| | - Stephen B. Gordon
- Malawi Liverpool Wellcome Trust Clinical Research Programme, College of Medicine, Chichiri, Blantyre, Malawi
| | - Simon P. Jochems
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniela M. Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
38
|
Soto M, Gaffney KJ, Rodgers KE. Improving the Innate Immune Response in Diabetes by Modifying the Renin Angiotensin System. Front Immunol 2019; 10:2885. [PMID: 31921148 PMCID: PMC6914815 DOI: 10.3389/fimmu.2019.02885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022] Open
Abstract
Patients with Type 2 Diabetes Mellitus (T2DM) suffer from a higher incidence and severity of pulmonary infections. This is likely due to immune impairment and structural abnormalities caused by T2DM-induced oxidative stress (OS) and chronic inflammation. Modulation of the Renin Angiotensin System (RAS) through blockade of the actions of angiotensin II (AII), or inducing the protective pathway, has the potential to reduce these pathological pathways. The effects of Angiotensin 1–7 [A(1-7)] and NorLeu3-A(1-7) [NorLeu], ligands of the protective RAS, on the innate immune response were evaluated in the db/db mouse model of T2DM. Only NorLeu treatment reduced the structural pathologies in the lung caused by T2DM. A decreased in bactericidal activity and phagocytosis in diabetic animals was also observed; both A(1-7) and NorLeu treatment restored these functions. Myeloid progenitor CFUs were reduced and neutrophil/progenitor OS was increased in saline-treated db/db mice, and was reversed by A(1-7) and NorLeu treatment. These results demonstrate the adverse effects of diabetes on factors that contribute to pulmonary infections and the therapeutic potential of protective RAS peptides. Overall, RAS-modification may be a viable therapeutic target to treat diabetic complications that are not addressed by glucose lowering drugs.
Collapse
Affiliation(s)
- Maira Soto
- Pharmacology Department, College of Medicine, Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Kevin J Gaffney
- Pharmacology Department, College of Medicine, Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Kathleen E Rodgers
- Pharmacology Department, College of Medicine, Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
39
|
Plazyo O, Sheng JJ, Jin JP. Downregulation of calponin 2 contributes to the quiescence of lung macrophages. Am J Physiol Cell Physiol 2019; 317:C749-C761. [PMID: 31365293 PMCID: PMC6850996 DOI: 10.1152/ajpcell.00036.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022]
Abstract
Calponin 2 is an actin cytoskeleton-associated regulatory protein that inhibits the activity of myosin-ATPase and cytoskeleton dynamics. Recent studies have demonstrated that deletion of calponin 2 restricts the proinflammatory activation of macrophages in atherosclerosis and arthritis to attenuate the disease progression in mice. Here we demonstrate that the levels of calponin 2 vary among different macrophage populations, which may reflect their adaptation to specific tissue microenvironment corresponding to specific functional states. Interestingly, lung resident macrophages express significantly lower calponin 2 than peritoneal resident macrophages, which correlates with decreased substrate adhesion and reduced expression of proinflammatory cytokines and a proresolution phenotype. Deletion of calponin 2 in peritoneal macrophages also decreased substrate adhesion and downregulated the expression of proinflammatory cytokines. Providing the first line of defense against microbial invasion while receiving constant exposure to extrinsic antigens, lung macrophages need to maintain a necessary level of activity while limiting exaggerated inflammatory reaction. Therefore, their low level of calponin 2 may reflect an important physiological adaption. Downregulation of calponin 2 in macrophages may be targeted as a cytoskeleton-based novel mechanism, possibly via endoplasmic reticulum stress altering the processing and secretion of cytokines, to regulate immune response and promote quiescence for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Olesya Plazyo
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Juan-Juan Sheng
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - J-P Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
40
|
Beckmann A, Grissmer A, Meier C, Tschernig T. Intercellular communication between alveolar epithelial cells and macrophages. Ann Anat 2019; 227:151417. [PMID: 31563569 DOI: 10.1016/j.aanat.2019.151417] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The alveolus in the lung tissue is an extremely vulnerable site. Alveolar macrophages control this micro-environment both in states of health and illnesssuch as acute lung injury and infection. It has been reported in mice in vivo that intercellular communication between alveolar macrophages and alveolar epithelial cells is mediated by gap junctions. However, little is known about thismicro-environment in human cells. METHODS Since this gap junctional intercellular communication is hard to investigate in human tissues, a co-culture model of two human cell lines, one of epithelial and one of macrophage origin, was used. Immunoblot analysis, freeze fracture replica immunolabeling and electron microscopy were performed. RESULTS Connexin (Cx) 43 protein expression as well as ultrastructurally defined Cx43 gap junctions were detected in co-cultures, yielding evidence of intercellular gap junctions between human alveolar cells of two distinct entities. CONCLUSION Alveolar macrophages possibly have direct access to the alveolar epithelium via gap junctions in humans, enabling the orchestration of the microenvironment in physiology and disease states.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Alexander Grissmer
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| |
Collapse
|
41
|
Liu W, Gao C, Dai H, Zheng Y, Dong Z, Gao Y, Liu F, Zhang Z, Liu Z, Liu W, Liu B, Liu Q, Shi J. Immunological Pathogenesis of Membranous Nephropathy: Focus on PLA2R1 and Its Role. Front Immunol 2019; 10:1809. [PMID: 31447839 PMCID: PMC6691064 DOI: 10.3389/fimmu.2019.01809] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/17/2019] [Indexed: 12/30/2022] Open
Abstract
Membranous nephropathy (MN) is the major cause of nephrotic syndrome with special pathological features, caused by the formation of immune complexes in the space between podocytes and the glomerular basement membrane. In idiopathic membranous nephropathy (IMN) the immune complexes are formed by circulating antibodies binding mainly to one of two naturally-expressed podocyte antigens: the M-type receptor for secretory phospholipase A2 (PLA2R1) and the Thrombospondin type-1 domain-containing 7A (THSD7A). Formation of antibodies against PLA2R1 is much more common, accounting for 70-80% of IMN. However, the mechanism of anti-podocyte antibody production in IMN is still unclear. In this review, we emphasize that the exposure of PLA2R1 is critical for triggering the pathogenesis of PLA2R1-associated MN, and propose the potential association between inflammation, pollution and PLA2R1. Our review aims to clarify the current research of these precipitating factors in a way that may suggest future directions for discovering the pathogenesis of MN, leading to additional therapeutic targets and strategies for the prevention and early treatment of MN.
Collapse
Affiliation(s)
- Wenbin Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Capital Medical University, Beijing, China
| | - Chang Gao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Haoran Dai
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Yang Zheng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhaocheng Dong
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yu Gao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Capital Medical University, Beijing, China
| | - Fei Liu
- Beijing University of Chinese Medicine, Beijing, China
| | - Zihan Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | | | - Weijing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jialan Shi
- Departments of Medicine, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
42
|
Veldhuizen RAW, McCaig LA, Pape C, Gill SE. The effects of aging and exercise on lung mechanics, surfactant and alveolar macrophages. Exp Lung Res 2019; 45:113-122. [PMID: 31195852 DOI: 10.1080/01902148.2019.1605633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Purpose: Advancing age leads to changes to the respiratory system associated with increased susceptibility to lung diseases, and exercise may counteract this effect. To explore the underlying processes, we investigated the effects of aging and exercise on lung mechanics, alveolar macrophage function, and surfactant pools and activity, in mice. It was hypothesized that aging would impact lung mechanics, macrophage polarization, and the status of the surfactant system, and that these changes would be mitigated by exercise. Methods: Male C57BL/6 mice were housed from 2-3 to 22 months, for the aged group, or until 4 months of age for young mice. Mice in both groups were randomized to voluntarily running exercise or to non-exercise, for a 2-month period. Mice were euthanized and lung mechanics were analyzed using a flexiVent ventilator. Subsequently, the lungs were lavaged to obtain pulmonary surfactant and alveolar macrophages. Pulmonary surfactant was analyzed for pool sizes and activity whereas alveolar macrophages were examined for response to pro and anti-inflammatory stimuli. Results: Changes in lung mechanics, such as increased compliance and decreased airway resistance, were associated with aging but were not affected by exercise. The quantity as well as the biophysical activity of the pulmonary surfactant system was unaffected by either aging or exercise. More alveolar macrophages were recovered from exercising aged mice compared to both the young and non-exercising groups. Macrophages in this aged exercise group were more responsive to an anti-inflammatory stimulus. Conclusions: Our data supports previous literature that suggest the development of emphysema-like alterations to lung mechanics with aging. This effect was independent of exercise. Our data also indicates that surfactant is unaffected by aging and exercise. Alveolar macrophage properties and numbers were affected by exercise in the aging lung and may represent the main, potentially beneficial, effect of exercise on the pulmonary system.
Collapse
Affiliation(s)
- Ruud A W Veldhuizen
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Lynda A McCaig
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Cynthia Pape
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Sean E Gill
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| |
Collapse
|
43
|
Age-related dynamics of pro-inflammatory cytokines in equine bronchoalveolar lavage (BAL) fluid and peripheral blood from horses managed on pasture. Exp Gerontol 2019; 124:110634. [PMID: 31201919 DOI: 10.1016/j.exger.2019.110634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/22/2019] [Accepted: 06/07/2019] [Indexed: 12/25/2022]
Abstract
The objectives of this study were to evaluate the natural age-related variation and compare the level of pro-inflammatory cytokines in the peripheral blood and lower airways of horses. The mRNA expression of IL-1β, IL-6, IL-8 TNF-α TLR-4 in bronchoalveolar lavage (BAL) fluid and peripheral blood mononuclear cells (PBMC) were studied by quantitative real time polymerase chain reaction (PCR) and differential cell count cytology from 44 horses of different ages. A significant age-related increase was found for the mRNA expression of IL-6, IL-8, TLR-4 and TNF-α in stimulated BAL cells and for TNF-α in stimulated PBMC. Furthermore, a significant decrease was found in the mRNA expression of IL-1β and TNF-α in stimulated BAL cells compared to stimulated PBMC. In conclusion, continued low antigen exposure of horses in the pasture environment could lead to a tight regulation of mRNA gene expression in the airway space compared to the peripheral blood and favour an age-related accumulation of mRNA genes.
Collapse
|
44
|
Wang J, Cao Y, Liu Y, Zhang X, Ji F, Li J, Zou Y. PIM1 inhibitor SMI-4a attenuated lipopolysaccharide-induced acute lung injury through suppressing macrophage inflammatory responses via modulating p65 phosphorylation. Int Immunopharmacol 2019; 73:568-574. [PMID: 31203114 DOI: 10.1016/j.intimp.2019.05.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/01/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022]
Abstract
PIM kinase is involved in the cellular processes of growth, differentiation and apoptosis. However, the role of PIM1 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains largely unknown. A trend of PIM1 in the lung tissue of LPS-induced ALI at different time points was detected. Histology, wet/dry (W/D) ratio, inflammatory cells in the bronchoalveolar lavage fluid (BALF) and survival rate analyses were performed when mice received the PIM1 inhibitor SMI-4a intratracheally 3 h before LPS administration. Cytokine production in vivo and in vitro was measured after SMI-4a pretreatment. NF-κB subunit p65 expression in nuclei and phosphorylation at Ser276 in lung tissues or cells were detected by Western blot analysis. The results showed that PIM1 mRNA and protein were upregulated in the lung tissue of LPS-induced ALI. The PIM1 inhibitor SMI-4a markedly improved the survival rate after lethal LPS administration, reduced the severity of lung edema, attenuated the histologic injuries of the lung tissue and reduced the counts of infiltrated inflammatory cells in the BALF. The PIM1 inhibitor SMI-4a suppressed the production of cytokines in LPS-treated RAW264.7 cell supernatants and BALF. Furthermore, LPS administration upregulated the levels of nuclear p65 and phosphorylated p65 (p-p65) at Ser276, whereas pretreatment with the PIM1 inhibitor SMI-4a reduced p65 upregulation in the nucleus and p-p65 at Ser276. Taken together, these data indicate that the PIM1 inhibitor SMI-4a may serve as a promising therapeutic strategy for LPS-induced ALI by suppressing macrophage production of cytokines via a reduction of p65 activities.
Collapse
Affiliation(s)
- Jinxuan Wang
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fanceng Ji
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Jinbao Li
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
45
|
Liu S, Hao C, Bao L, Zhao D, Zhang H, Hou J, Wang D, Chen H, Feng F, Yao W. Silica Particles Mediate Phenotypic and Functional Alteration of Dendritic Cells and Induce Th2 Cell Polarization. Front Immunol 2019; 10:787. [PMID: 31068929 PMCID: PMC6491578 DOI: 10.3389/fimmu.2019.00787] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
During silicosis, immune cells, including macrophages, T cells, B cells, and NK cells, participate in fibrosis development through alteration of the immune status. Dendritic cells (DCs) are professional antigen-presenting cells (APCs) with a key role in initiating immune responses and sustaining immune tolerance to maintain homeostasis. The relative contribution of DCs to silicosis progression is not well-documented. In the current study, we investigated the phenotypic and functional alterations of peripheral blood mononuclear cell (PBMC)-derived DCs of Sprague-Dawley (SD) rat during immune responses to silica exposure. We established models for direct and indirect exposure of DCs to silica by either treating DCs with silica or coculturing them with alveolar macrophages (AMs) treated with silica, respectively. The functional activity of DCs was analyzed by measuring their expression of costimulatory molecules, fluorescent microparticle uptake, cytokine production, and ability to mediate T cell polarization in vitro. In vivo, we demonstrated that silica could induce DC migration in response to silica exposure. Our results show that cytokine production by DCs was increased in response to direct silica direct exposure, while indirect silica exposure led to reduced cytokine levels. Moreover, the phagocytic capacity of DCs increased in cocultures after silica exposure. Gene and protein expression analyses showed that silica exposure altered the expression levels of Toll-like receptor pathway proteins and inflammatory factors. DC surface expression of the costimulatory molecules, CD80, CD86, and major histocompatibility complex, was inhibited by exposure to silica, which mediated a Th2-polarizing response in vitro. In rats, silica exposure induced migration of DCs from the peripheral blood into the alveoli. These results demonstrate that direct and indirect exposure to silica particles alter the phenotype and function of DCs, thereby regulating immune responses. Such changes may contribute to the development of silicosis by altering DC phenotype, function, and migration and by influencing the balance between Th1 and Th2 cells.
Collapse
Affiliation(s)
- Suna Liu
- Department of Henan Newborn Screening Center, Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Changfu Hao
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Lei Bao
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Dehua Zhao
- Department of Henan Newborn Screening Center, Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyi Zhang
- Hospital Infection Management, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Jianyong Hou
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Di Wang
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Huiting Chen
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Feifei Feng
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wu Yao
- Department of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Clinical Application of Stem/Stromal Cells in COPD. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121219 DOI: 10.1007/978-3-030-29403-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive life-threatening disease that is significantly increasing in prevalence and is predicted to become the third leading cause of death worldwide by 2030. At present, there are no true curative treatments that can stop the progression of the disease, and new therapeutic strategies are desperately needed. Advances in cell-based therapies provide a platform for the development of new therapeutic approaches in severe lung diseases such as COPD. At present, a lot of focus is on mesenchymal stem (stromal) cell (MSC)-based therapies, mainly due to their immunomodulatory properties. Despite increasing number of preclinical studies demonstrating that systemic MSC administration can prevent or treat experimental COPD and emphysema, clinical studies have not been able to reproduce the preclinical results and to date no efficacy or significantly improved lung function or quality of life has been observed in COPD patients. Importantly, the completed appropriately conducted clinical trials uniformly demonstrate that MSC treatment in COPD patients is well tolerated and no toxicities have been observed. All clinical trials performed so far, have been phase I/II studies, underpowered for the detection of potential efficacy. There are several challenges ahead for this field such as standardized isolation and culture procedures to obtain a cell product with high quality and reproducibility, administration strategies, improvement of methods to measure outcomes, and development of potency assays. Moreover, COPD is a complex pathology with a diverse spectrum of clinical phenotypes, and therefore it is essential to develop methods to select the subpopulation of patients that is most likely to potentially respond to MSC administration. In this chapter, we will discuss the current state of the art of MSC-based cell therapy for COPD and the hurdles that need to be overcome.
Collapse
|
48
|
Chen IC, Lin YT, Huang JS, Wu BN, Hsu JH, Tan MS, Dai ZK. Decreased Ambient Oxygen Tension Alters the Expression of Endothelin-1, iNOS and cGMP in Rat Alveolar Macrophages. Int J Med Sci 2019; 16:443-449. [PMID: 30911278 PMCID: PMC6428981 DOI: 10.7150/ijms.28353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/28/2018] [Indexed: 01/20/2023] Open
Abstract
Background: Hypoxia plays an important role in the vascular tone of pulmonary circulation via the vasculature and parenchymal tissue. Endothelin-1 (ET-1), a potent vasoconstrictive peptide, plays a role in inflammation in mononuclear cells. Nitric oxide synthase (NOS), which generates nitric oxide (NO)/cyclic 3', 5'-monophosphate (cGMP), is coexpressed with ET-1 in many cell types. The aim of this study was to assess whether hypoxia induces the production of ET-1 and associated expression of NOS, NO/cGMP and chemokines in rat alveolar macrophages (AMs). Methods: NR8383 cells were cultured under hypoxic (1% oxygen) conditions for 0, 2, 4, 8 and 12 hours. Levels of ET-1, inducible NOS (iNOS), phosphorylated iNOS (p-iNOS), nitrite/nitrate (NOx), cGMP and monocyte chemoattractant protein-1 (MCP-1) were measured. Results: ET-1, p-iNOS, NOx, and cGMP increased significantly in AMs after 4 hours of hypoxia (p < 0.05). ET-1 and MCP-1 mRNA increased after 8 hours (p < 0.05). The protein expression of ET-1, MCP-1, and p-iNOS increased in a time-dependent manner, while iNOS expression decreased with time. Conclusions: The changes in ET-1, p-iNOS, and the NO/cGMP pathway in AMs may help elucidate the mechanisms in the hypoxic lung. Understanding changes in the endothelin axis in hypoxic AMs is a crucial first step to unravel its role in pulmonary circulation.
Collapse
Affiliation(s)
- I-Chen Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Tsai Lin
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jhy-Shrian Huang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mian-Shin Tan
- Department of Biomedical Science and Environmental Biology, College of Life Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zen-Kong Dai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
49
|
Hyphae fragments from A. fumigatus sensitize lung cells to silica particles (Min-U-Sil): Increased release of IL-1β. Toxicol In Vitro 2018; 55:1-10. [PMID: 30414920 DOI: 10.1016/j.tiv.2018.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 12/16/2022]
Abstract
Exposure to particulate matter (PM), such as mineral particles and biological particles/components may be linked to aggravation of respiratory diseases, including asthma. Here we report that exposure to Aspergillus fumigatus hyphae fragments (AFH) and lipopolysaccharide (LPS) induced both mRNA synthesis and release of pro-inflammatory interleukin-1 beta (IL-1β) in both human THP-1 monocytes (THP-1 Mo) and phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 monocytes (THP-1 macrophages; THP-1 Ma); while Min-U-Sil alone enhanced the release of IL-1β only in THP-1 Ma. Co-exposure to LPS or AFH with Min-U-Sil caused a synergistic release of IL-1β when compared to single exposures. In contrast, Min-U-Sil did not markedly change LPS- and AFH-induced release of tumor necrosis factor alpha (TNF-α). The combined exposures did not increase the LPS- and AFH-induced expression of IL-1β mRNA. Notably, the AFH- and LPS-induced IL-1β responses with and without co-exposure to Min-U-Sil in THP-1 Mo were found to be caspase-dependent as shown by inhibition with zYVAD-fmk. Furthermore, co-exposure with AFH and Min-U-Sil resulted in similar synergistic releases of IL-1β in primary human airway macrophages (AM; sputum), peripheral blood monocyte-derived macrophages (MDM) and in the human bronchial epithelial cell line (BEAS-2B). In conclusion, AFH induce both the synthesis and release of IL-1β. However, Min-U-Sil further enhanced the cleavage of the induced pro-IL-1β.
Collapse
|
50
|
Zhang S, Dai H, Zhu L, Lin F, Hu Z, Jing R, Zhang W, Zhao C, Hong X, Zhong JH, Pan L. Microvesicles packaging IL-1β and TNF-α enhance lung inflammatory response to mechanical ventilation in part by induction of cofilin signaling. Int Immunopharmacol 2018; 63:74-83. [PMID: 30075431 DOI: 10.1016/j.intimp.2018.07.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/13/2018] [Accepted: 07/26/2018] [Indexed: 12/18/2022]
Abstract
Microvesicles shed from pulmonary cells are capable of transferring inflammatory cargo to recipient cells nearby or in distant to enhance inflammation. Some authors believe that cofilin controls actin dynamics and regulates vesicle mobilization. We therefore investigated the potential role and mechanism of microvesicles in ventilator-induced lung injury (VILI). Fifty male C57BL/6 mice were orotracheally intubated and either allowed to breathe spontaneously or they were mechanically ventilated with different tidal volumes (Vt) and ventilation times. Lung tissue injury was assessed in terms of lung histopathologic examination, wet/dry weight ratios, and levels of total proteins and of cytokines. Microvesicle characteristics, sizes, contents and levels as well as cofilin were also measured. We found that lung inflammation increased significantly after ventilation with high Vt for 4 h; these conditions led to secretion of larger and more microvesicles into the alveoli than animals with/without ventilation at low Vt. Intratracheal instillation of microvesicles obtained from animals ventilated with low or high Vt triggered significant lung inflammation in naive mice, and these high-Vt microvesicles not only carried more IL-1β and TNF-α but also induced more severe lung inflammation compared to low-Vt microvesicles; And high-Vt microvesicles at 2 h carried more molecular cargo than that at 1 h or 4 h, which may involve the shift and amplification of inflammation. Furthermore, blocking the phosphorylation of cofilin can not only inhibit microvesicle formation in the lung, but also reduce lung injury. Collectively, our data suggest that microvesicles packaging IL-1β and TNF-α enhance lung inflammation in VILI.
Collapse
Affiliation(s)
- Suisui Zhang
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Huijun Dai
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lingyu Zhu
- Department of Breast Surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Fei Lin
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhaokun Hu
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Ren Jing
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Weikang Zhang
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chen Zhao
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xueqi Hong
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Hong Zhong
- Department of Hepatobiliary Surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Linghui Pan
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|