101
|
Abstract
Alveolar macrophages (AMs) are lung-resident myeloid cells that sit at the interface of the airway and lung tissue. Under homeostatic conditions, their primary function is to clear debris, dead cells and excess surfactant from the airways. They also serve as innate pulmonary sentinels for respiratory pathogens and environmental airborne particles and as regulators of pulmonary inflammation. However, they have not typically been viewed as primary therapeutic targets for respiratory diseases. Here, we discuss the role of AMs in various lung diseases, explore the potential therapeutic strategies to target these innate cells and weigh the potential risks and challenges of such therapies. Additionally, in the context of the COVID-19 pandemic, we examine the role AMs play in severe disease and the therapeutic strategies that have been harnessed to modulate their function and protect against severe lung damage. There are many novel approaches in development to target AMs, such as inhaled antibiotics, liposomal and microparticle delivery systems, and host-directed therapies, which have the potential to provide critical treatment to patients suffering from severe respiratory diseases, yet there is still much work to be done to fully understand the possible benefits and risks of such approaches.
Collapse
|
102
|
Barman TK, Metzger DW. Disease Tolerance during Viral-Bacterial Co-Infections. Viruses 2021; 13:v13122362. [PMID: 34960631 PMCID: PMC8706933 DOI: 10.3390/v13122362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/29/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
Disease tolerance has emerged as an alternative way, in addition to host resistance, to survive viral-bacterial co-infections. Disease tolerance plays an important role not in reducing pathogen burden, but in maintaining tissue integrity and controlling organ damage. A common co-infection is the synergy observed between influenza virus and Streptococcus pneumoniae that results in superinfection and lethality. Several host cytokines and cells have shown promise in promoting tissue protection and damage control while others induce severe immunopathology leading to high levels of morbidity and mortality. The focus of this review is to describe the host cytokines and innate immune cells that mediate disease tolerance and lead to a return to host homeostasis and ultimately, survival during viral-bacterial co-infection.
Collapse
|
103
|
Sasaki E, Momose H, Furuhata K, Mizukami T, Hamaguchi I. Impact of injection buffer volume to perform bronchoalveolar lavage fluid collection for isolating alveolar macrophages to investigate fine particle-induced IL-1α secretion. J Immunotoxicol 2021; 18:163-172. [PMID: 34761701 DOI: 10.1080/1547691x.2021.1979699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The importance of alveolar macrophages has been reported in many toxicology/immunology studies. Alveolar macrophages release interleukin (IL)-1α as a damage-associated molecular pattern (DAMP) when stimulated by fine particles. However, it is unclear whether cell isolation procedures affect ex vivo particle-induced responses in primary mouse alveolar macrophages (mAM). In this study, effects of injection buffer volume used to perform bronchoalveolar lavage fluid (BALF) collection to isolate mAM for use in ex vivo particle-induced responses were assessed. Among the mAM obtained from BALF collected using a 0.55 or 0.75 ml, but not a 1.0 ml buffer injection volume, decreased cell viability and IL-1α release were observed when cells were stimulated ex vivo with silica crystal or aluminum salt. Injected buffer composition did not affect the IL-1α release. On the other hand, IL-6 secretion induced by lipopolysaccharide (LPS) did not differ among mAM obtained from BALF collected using the different volumes. Expression levels of cell surface markers like CD11c, SiglecF, and CD64 did not differ among mAM obtained from BALF collected using the different injection buffer volumes. IL-1α release (and also necroptosis) induced by ex vivoparticle stimulation was suppressed by RIPK3 inhibitor or cytochalasin D co-treatment. Decreases in RIPK3 phosphorylation were noted in mAM obtained in BALF collected using the 1.0 ml injection volume compared with mAM obtained in BALF using 0.55 or 0.75 ml buffer. These observations illustrate that larger volumes of buffer used to collect BALF from mice can affect sensitivity of the isolated mAM to ex vivo particle-induced responses by inhibiting their functions.
Collapse
Affiliation(s)
- Eita Sasaki
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Keiko Furuhata
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takuo Mizukami
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
104
|
Yau E, Chen Y, Song C, Webb J, Carillo M, Kawasawa YI, Tang Z, Takahashi Y, Umstead TM, Dovat S, Chroneos ZC. Genomic and epigenomic adaptation in SP-R210 (Myo18A) isoform-deficient macrophages. Immunobiology 2021; 226:152150. [PMID: 34735924 DOI: 10.1016/j.imbio.2021.152150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 10/20/2022]
Abstract
Macrophages play an important role in maintaining tissue homeostasis, from regulating the inflammatory response to pathogens to resolving inflammation and aiding tissue repair. The surfactant protein A (SP-A) receptor SP-R210 (MYO18A) has been shown to affect basal and inflammatory macrophage states. Specifically, disruption of the longer splice isoform SP-R210L/MYO18Aα renders macrophages hyper-inflammatory, although the mechanism by which this occurs is not well understood. We asked whether disruption of the L isoform led to the hyper-inflammatory state via alteration of global genomic responses. RNA sequencing analysis of L isoform-deficient macrophages (SP-R210L(DN)) revealed basal and influenza-induced upregulation of genes associated with inflammatory pathways, such as TLR, RIG-I, NOD, and cytoplasmic DNA signaling, whereas knockout of both SP-R210 isoforms (L and S) only resulted in increased RIG-I and NOD signaling. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis showed increased genome-wide deposition of the pioneer transcription factor PU.1 in SP-R210L(DN) cells, with increased representation around genes relevant to inflammatory pathways. Additional ChIP-seq analysis of histone H3 methylation marks showed decreases in both repressive H3K9me3 and H3K27me3 marks with a commensurate increase in transcriptionally active (H3K4me3) histone marks in the L isoform deficient macrophages. Influenza A virus (IAV) infection, known to stimulate a wide array of anti-viral responses, caused a differential redistribution of PU.1 binding between proximal promoter and distal sites and decoupling from Toll-like receptor regulated gene promoters in SP-R210L(DN) cells. These finding suggest that the inflammatory differences seen in SP-R210L-deficient macrophages are a result of transcriptional differences that are mediated by epigenetic changes brought about by differential expression of the SP-R210 isoforms. This provides an avenue to explore how the signaling pathways downstream of the receptor and the ligands can modulate the macrophage inflammatory response.
Collapse
Affiliation(s)
- Eric Yau
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA.
| | - Yan Chen
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA; Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunhua Song
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Pennsylvania State University College of Medicine, PA, USA; Department of Internal Medicine, Ohio State University College of Medicine, Columbus, OH, USA
| | - Jason Webb
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA
| | - Marykate Carillo
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Zhenyuan Tang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yoshinori Takahashi
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Todd M Umstead
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA
| | - Sinisa Dovat
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zissis C Chroneos
- Department of Pediatrics and Microbiology and Immunology, Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine, PA, USA.
| |
Collapse
|
105
|
Okamori S, Ishii M, Asakura T, Suzuki S, Namkoong H, Kagawa S, Hegab AE, Yagi K, Kamata H, Kusumoto T, Ogawa T, Takahashi H, Yoda M, Horiuchi K, Hasegawa N, Fukunaga K. ADAM10 partially protects mice against influenza pneumonia by suppressing specific myeloid cell population. Am J Physiol Lung Cell Mol Physiol 2021; 321:L872-L884. [PMID: 34523355 DOI: 10.1152/ajplung.00619.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The influenza virus infection poses a serious health threat worldwide. Myeloid cells play pivotal roles in regulating innate and adaptive immune defense. A disintegrin and metalloproteinase (ADAM) family of proteins contributes to various immune responses; however, the role of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in influenza virus infection remains largely unknown. Herein, we investigated its role, focusing on myeloid cells, during influenza virus infection in mice. ADAM10 gene (Adam10)flox/flox/Lyz2-Cre (Adam10ΔLyz2) and control Adam10flox/flox mice were intranasally infected with 200 plaque-forming units of influenza virus A/H1N1/PR8/34. Adam10ΔLyz2 mice exhibited a significantly higher mortality rate, stronger lung inflammation, and a higher virus titer in the lungs than control mice. Macrophages and inflammatory cytokines, such as TNF-α, IL-1β, and CCL2, were increased in bronchoalveolar lavage fluid from Adam10ΔLyz2 mice following infection. CD11b+Ly6G-F4/80+ myeloid cells, which had an inflammatory monocyte/macrophage-like phenotype, were significantly increased in the lungs of Adam10ΔLyz2 mice. Adoptive transfer experiments suggested that these cells likely contributed to the poorer prognosis in Adam10ΔLyz2 mice. Seven days after infection, CD11b+Ly6G-F4/80+ lung cells exhibited significantly higher arginase-1 expression levels in Adam10ΔLyz2 mice than in control mice, whereas an arginase-1 inhibitor improved the prognosis of Adam10ΔLyz2 mice. Enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF)/GM-CSF receptor signaling likely contributed to this process. Collectively, these results indicate that myeloid ADAM10 protects against influenza virus pneumonia and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Satoshi Okamori
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Society of Promotion of Science, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shoji Suzuki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Medical Education Center, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Kazuma Yagi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Kusumoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takunori Ogawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Yoda
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Orthopedic Surgery, National Defence Medical College, Saitama, Japan
| | - Naoki Hasegawa
- Center for Infectious Diseases and Infection Control, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
106
|
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
|
107
|
Martin FP, Jacqueline C, Poschmann J, Roquilly A. Alveolar Macrophages: Adaptation to Their Anatomic Niche during and after Inflammation. Cells 2021; 10:cells10102720. [PMID: 34685700 PMCID: PMC8534884 DOI: 10.3390/cells10102720] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
At the early stages of life development, alveoli are colonized by embryonic macrophages, which become resident alveolar macrophages (ResAM) and self-sustain by local division. Genetic and epigenetic signatures and, to some extent, the functions of ResAM are dictated by the lung microenvironment, which uses cytokines, ligand-receptor interactions, and stroma cells to orchestrate lung homeostasis. In resting conditions, the lung microenvironment induces in ResAM a tolerogenic programming that prevents unnecessary and potentially harmful inflammation responses to the foreign bodies, which continuously challenge the airways. Throughout life, any episode of acute inflammation, pneumonia being likely the most frequent cause, depletes the pool of ResAM, leaving space for the recruitment of inflammatory monocytes that locally develop in monocyte-derived alveolar macrophages (InfAM). During lung infection, the local microenvironment induces a temporary inflammatory signature to the recruited InfAM to handle the tissue injury and eliminate the pathogens. After a few days, the recruited InfAM, which locally self-sustain and develop as new ResAM, gain profibrotic functions required for tissue healing. After the complete resolution of the infectious episode, the functional programming of both embryonic and monocyte-derived ResAM remains altered for months and possibly for the entire life. Adult lungs thus contain a wide diversity of ResAM since every infection brings new waves of InfAM which fill the room left open by the inflammatory process. The memory of these innate cells called trained immunity constitutes an immunologic scar left by inflammation, notably pneumonia. This memory of ResAM has advantages and drawbacks. In some cases, lung-trained immunity offers better defense capacities against autoimmune disorders and the long-term risk of infection. At the opposite, it can perpetuate a harmful process and lead to a pathological state, as is the case among critically ill patients who have immune paralysis and are highly susceptible to hospital-acquired pneumonia and acute respiratory distress syndrome. The progress in understanding the kinetics of response of alveolar macrophages (AM) to lung inflammation is paving the way to new treatments of pneumonia and lung inflammatory process.
Collapse
Affiliation(s)
- Florian Pierre Martin
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
| | - Cédric Jacqueline
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
| | - Jeremie Poschmann
- Centre de Recherche en Transplantation et Immunologie, University of Nantes, UMR 1064, ITUN, Inserm, F-44000 Nantes, France;
| | - Antoine Roquilly
- EA3826 Host Pathogen Interactions, Inflammation and Mucosal Immunity, Department of Anesthesiology and Intensive Medicine, Hôtel Dieu, CHU Nantes, University of Nantes, F-44000 Nantes, France; (F.P.M.); (C.J.)
- Correspondence: ; Tel.: +33-253482230
| |
Collapse
|
108
|
Hong C, Lu H, Jin R, Huang X, Chen M, Dai X, Gong F, Dong H, Wang H, Gao XM. Cytokine Cocktail Promotes Alveolar Macrophage Reconstitution and Functional Maturation in a Murine Model of Haploidentical Bone Marrow Transplantation. Front Immunol 2021; 12:719727. [PMID: 34621268 PMCID: PMC8490745 DOI: 10.3389/fimmu.2021.719727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/02/2021] [Indexed: 11/23/2022] Open
Abstract
Infectious pneumonia is one of the most common complications after bone marrow transplantation (BMT), which is considered to be associated with poor reconstitution and functional maturation of alveolar macrophages (AMs) post-transplantation. Here, we present evidence showing that lack of IL-13-secreting group 2 innate lymphoid cells (ILC2s) in the lungs may underlay poor AM reconstitution in a mouse model of haploidentical BMT (haplo-BMT). Recombinant murine IL-13 was able to potentiate monocyte-derived AM differentiation in vitro. When intranasally administered, a cocktail of granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-13, and CCL2 not only promoted donor monocyte-derived AM reconstitution in haplo-BMT-recipient mice but also enhanced the innate immunity of the recipient animals against pulmonary bacterial infection. These results provide a useful clue for a clinical strategy to prevent pulmonary bacterial infection at the early stage of recipients post-BMT.
Collapse
Affiliation(s)
- Chao Hong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongyun Lu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Rong Jin
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaohong Huang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ming Chen
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaoqiu Dai
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Fangyuan Gong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongliang Dong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongmin Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
109
|
Gschwend J, Sherman SP, Ridder F, Feng X, Liang HE, Locksley RM, Becher B, Schneider C. Alveolar macrophages rely on GM-CSF from alveolar epithelial type 2 cells before and after birth. J Exp Med 2021; 218:e20210745. [PMID: 34431978 PMCID: PMC8404471 DOI: 10.1084/jem.20210745] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/27/2021] [Accepted: 08/04/2021] [Indexed: 12/31/2022] Open
Abstract
Programs defining tissue-resident macrophage identity depend on local environmental cues. For alveolar macrophages (AMs), these signals are provided by immune and nonimmune cells and include GM-CSF (CSF2). However, evidence to functionally link components of this intercellular cross talk remains scarce. We thus developed new transgenic mice to profile pulmonary GM-CSF expression, which we detected in both immune cells, including group 2 innate lymphoid cells and γδ T cells, as well as AT2s. AMs were unaffected by constitutive deletion of hematopoietic Csf2 and basophil depletion. Instead, AT2 lineage-specific constitutive and inducible Csf2 deletion revealed the nonredundant function of AT2-derived GM-CSF in instructing AM fate, establishing the postnatal AM compartment, and maintaining AMs in adult lungs. This AT2-AM relationship begins during embryogenesis, where nascent AT2s timely induce GM-CSF expression to support the proliferation and differentiation of fetal monocytes contemporaneously seeding the tissue, and persists into adulthood, when epithelial GM-CSF remains restricted to AT2s.
Collapse
Affiliation(s)
- Julia Gschwend
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Frederike Ridder
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Xiaogang Feng
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Hong-Erh Liang
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Richard M. Locksley
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California San Francisco, San Francisco, CA
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
110
|
Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021; 13:v13091725. [PMID: 34578306 PMCID: PMC8472850 DOI: 10.3390/v13091725] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory tract infections constitute a significant public health problem, with a therapeutic arsenal that remains relatively limited and that is threatened by the emergence of antiviral and/or antibiotic resistance. Viral–bacterial co-infections are very often associated with the severity of these respiratory infections and have been explored mainly in the context of bacterial superinfections following primary influenza infection. This review summarizes our current knowledge of the mechanisms underlying these co-infections between respiratory viruses (influenza viruses, RSV, and SARS-CoV-2) and bacteria, at both the physiological and immunological levels. This review also explores the importance of the microbiome and the pathological context in the evolution of these respiratory tract co-infections and presents the different in vitro and in vivo experimental models available. A better understanding of the complex functional interactions between viruses/bacteria and host cells will allow the development of new, specific, and more effective diagnostic and therapeutic approaches.
Collapse
|
111
|
Chhetri G. Emerging roles of IL-34 in neurodegenerative and neurological infectious disease. Int J Neurosci 2021; 133:660-671. [PMID: 34347576 DOI: 10.1080/00207454.2021.1963962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neurological infections are often devastating in their clinical presentation. Although significant advances have made in neuroimaging techniques and molecular tools for diagnosis, as well as in anti-infective therapy, these diseases always difficult to diagnose and treat. Neuroparasitic infections and virus infections lead to neurological infections. In the nervous system, various cytokines and chemokines act as neuroinflammatory agents, neuromodulators, regulate neurodevelopment, and synaptic transmission. Among the most important cytokines, interleukins (ILs) are a large group of immunomodulatory proteins that elicit a wide variety of responses in cells and tissues. These ILs are involved in pro and anti-inflammatory effects, systemic inflammation, immune system modulation and play crucial roles in fighting cancer, infectious disease, and neurological disorders. Interleukin-34 (IL-34) identified by screening a comprehensive human protein library containing ∼3400 secreted and extracellular domain proteins in a human monocyte viability assay. Recent evidence has disclosed the crucial roles of IL-34 in the proliferation and differentiation of mononuclear phagocyte lineage cells, osteoclastogenesis, and inflammation. Additionally, IL-34 plays an important role in development, homeostasis, and disease. Dysregulation in IL-34 function can lead to various inflammatory and infectious diseases (e.g. Inflammatory bowel disease, liver fibrosis, Systemic Lupus erythematosus, rheumatoid arthritis), neurological disorders (e.g. Alzheimer disease) and neurological infectious disease (e.g. West Nile virus disease). In this review, we explore the biological role of IL-34 in addition to various impairments caused by dysregulation in IL-34 and discuss their potential links that may lead to important therapeutic and/or preventive strategies for these disorders.
Collapse
Affiliation(s)
- Gaurav Chhetri
- School of Pharmacy, Shanghai Jiao Tong University, Minhang, Shanghai, P.R. China
| |
Collapse
|
112
|
Development of mode of action networks related to the potential role of PPARγ in respiratory diseases. Pharmacol Res 2021; 172:105821. [PMID: 34403731 DOI: 10.1016/j.phrs.2021.105821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022]
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) is a key transcription factor, operating at the intercept of metabolic control and immunomodulation. It is ubiquitously expressed in multiple tissues and organs, including lungs. There is a growing body of information supporting the role of PPARγ signalling in respiratory diseases. The aim of the present study was to develop mode of action (MoA) networks reflecting the relationships between PPARγ signalling and the progression/alleviation of a spectrum of lung pathologies. Data mining was performed using the resources of the NIH PubMed and PubChem information systems. By linking available data on pathological/therapeutic effects of PPARγ modulation, knowledge-based MoA networking at different levels of biological organization (molecular, cellular, tissue, organ, and system) was performed. Multiple MoA networks were developed to relate PPARγ modulation to the progress or the alleviation of pulmonary disorders, triggered by diverse pathogenic, genetic, chemical, or mechanical factors. Pharmacological targeting of PPARγ signalling was discussed with regard to ligand- and cell type-specific effects in the context of distinct disease inductor- and disease stage-dependent patterns. The proposed MoA networking analysis allows for a better understanding of the potential role of PPARγ modulation in lung pathologies. It presents a mechanistically justified basis for further computational, experimental, and clinical monitoring studies on the dynamic control of PPARγ signalling in respiratory diseases.
Collapse
|
113
|
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
|
114
|
Peng Y, Wang X, Wang H, Xu W, Wu K, Go X, Yin Y, Zhang X. Interleukin-4 protects mice against lethal influenza and Streptococcus pneumoniae co-infected pneumonia. Clin Exp Immunol 2021; 205:379-390. [PMID: 34061992 DOI: 10.1111/cei.13628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pneumoniae co-infection post-influenza is a major cause of mortality characterized by uncontrolled bacteria burden and excessive immune response during influenza pandemics. Interleukin (IL)-4 is a canonical type II immune cytokine known for its wide range of biological activities on different cell types. It displays protective roles in numerous infectious diseases and immune-related diseases, but its role in influenza and S. pneumoniae (influenza/S. pneumoniae) co-infected pneumonia has not been reported. In our study, we used C57BL/6 wild-type (WT) and IL-4-deficient (IL-4-/- ) mice to establish co-infection model with S. pneumoniae after influenza virus infection. Co-infected IL-4-/- mice showed increased mortality and weight loss compared with WT mice. IL-4 deficiency led to increased bacterial loads in lungs without altering influenza virus replication, suggesting a role of IL-4 in decreasing post-influenza susceptibility to S. pneumoniae co-infection. Loss of IL-4 also resulted in aggravated lung damage together with massive proinflammatory cytokine production and immune cell infiltration during co-infection. Administration of recombinant IL-4 rescued the survival and weight loss of IL-4-/- mice in lethal co-infection. Additionally, IL-4 deficiency led to more immune cell death in co-infection. Gasdermin D (GSDMD) during co-infection was induced in IL-4-/- mice that subsequently activated cell pyroptosis. Treatment of recombinant IL-4 or inhibition of GSDMD activity by disulfiram decreased immune cell death and bacterial loads in lungs of IL-4-/- co-infected mice. These results suggest that IL-4 decreases post-influenza susceptibility to S. pneumoniae co-infection via suppressing GSDMD-induced pyroptosis. Collectively, this study demonstrates the protective role of IL-4 in influenza/S. pneumoniae co-infected pneumonia.
Collapse
Affiliation(s)
- Yang Peng
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaofang Wang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China.,Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Hong Wang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Wenchun Xu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Kaifeng Wu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xuemei Go
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
115
|
Zhu B, Wu Y, Huang S, Zhang R, Son YM, Li C, Cheon IS, Gao X, Wang M, Chen Y, Zhou X, Nguyen Q, Phan AT, Behl S, Taketo MM, Mack M, Shapiro VS, Zeng H, Ebihara H, Mullon JJ, Edell ES, Reisenauer JS, Demirel N, Kern RM, Chakraborty R, Cui W, Kaplan MH, Zhou X, Goldrath AW, Sun J. Uncoupling of macrophage inflammation from self-renewal modulates host recovery from respiratory viral infection. Immunity 2021; 54:1200-1218.e9. [PMID: 33951416 PMCID: PMC8192557 DOI: 10.1016/j.immuni.2021.04.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/08/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/β-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of β-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, β-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted β-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This β-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by β-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.
Collapse
Affiliation(s)
- Bibo Zhu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yue Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Su Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Ruixuan Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Young Min Son
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Chaofan Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - In Su Cheon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Xiaochen Gao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Min Wang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yao Chen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Wauwatosa, WI 53226, USA
| | - Xian Zhou
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Division of Rheumatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Quynh Nguyen
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anthony T Phan
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Supriya Behl
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Virginia S Shapiro
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Hu Zeng
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Division of Rheumatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Hideki Ebihara
- Department of Molecular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - John J Mullon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Janani S Reisenauer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Nadir Demirel
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Ryan M Kern
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Wauwatosa, WI 53226, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ananda W Goldrath
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jie Sun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| |
Collapse
|
116
|
Dukhinova M, Kokinos E, Kuchur P, Komissarov A, Shtro A. Macrophage-derived cytokines in pneumonia: Linking cellular immunology and genetics. Cytokine Growth Factor Rev 2021; 59:46-61. [PMID: 33342718 PMCID: PMC8035975 DOI: 10.1016/j.cytogfr.2020.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
Macrophages represent the first line of anti-pathogen defense - they encounter invading pathogens to perform the phagocytic activity, to deliver the plethora of pro- and anti-inflammatory cytokines, and to shape the tissue microenvironment. Throughout pneumonia course, alveolar macrophages and infiltrated blood monocytes produce increasing cytokine amounts, which activates the antiviral/antibacterial immunity but can also provoke the risk of the so-called cytokine "storm" and normal tissue damage. Subsequently, the question of how the cytokine spectrum is shaped and balanced in the pneumonia context remains a hot topic in medical immunology, particularly in the COVID19 pandemic era. The diversity in cytokine profiles, involved in pneumonia pathogenesis, is determined by the variations in cytokine-receptor interactions, which may lead to severe cytokine storm and functional decline of particular tissues and organs, for example, cardiovascular and respiratory systems. Cytokines and their receptors form unique profiles in individual patients, depending on the (a) microenvironmental context (comorbidities and associated treatment), (b) lung monocyte heterogeneity, and (c) genetic variations. These multidisciplinary strategies can be proactively considered beforehand and during the pneumonia course and potentially allow the new age of personalized immunotherapy.
Collapse
Affiliation(s)
- Marina Dukhinova
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg, Russia.
| | - Elena Kokinos
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg, Russia
| | - Polina Kuchur
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg, Russia
| | - Alexey Komissarov
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg, Russia
| | - Anna Shtro
- International Institute "Solution Chemistry of Advanced Materials and Technology", ITMO University, St. Petersburg, Russia; Department of Chemotherapy, Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| |
Collapse
|
117
|
Li K, Cao P, McCaw JM. Modelling the Effect of MUC1 on Influenza Virus Infection Kinetics and Macrophage Dynamics. Viruses 2021; 13:v13050850. [PMID: 34066999 PMCID: PMC8150684 DOI: 10.3390/v13050850] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/20/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
MUC1 belongs to the family of cell surface (cs-) mucins. Experimental evidence indicates that its presence reduces in vivo influenza viral infection severity. However, the mechanisms by which MUC1 influences viral dynamics and the host immune response are not yet well understood, limiting our ability to predict the efficacy of potential treatments that target MUC1. To address this limitation, we use available in vivo kinetic data for both virus and macrophage populations in wildtype and MUC1 knockout mice. We apply two mathematical models of within-host influenza dynamics to this data. The models differ in how they categorise the mechanisms of viral control. Both models provide evidence that MUC1 reduces the susceptibility of epithelial cells to influenza virus and regulates macrophage recruitment. Furthermore, we predict and compare some key infection-related quantities between the two mice groups. We find that MUC1 significantly reduces the basic reproduction number of viral replication as well as the number of cumulative macrophages but has little impact on the cumulative viral load. Our analyses suggest that the viral replication rate in the early stages of infection influences the kinetics of the host immune response, with consequences for infection outcomes, such as severity. We also show that MUC1 plays a strong anti-inflammatory role in the regulation of the host immune response. This study improves our understanding of the dynamic role of MUC1 against influenza infection and may support the development of novel antiviral treatments and immunomodulators that target MUC1.
Collapse
Affiliation(s)
- Ke Li
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia; (P.C.); (J.M.M.)
- Correspondence:
| | - Pengxing Cao
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia; (P.C.); (J.M.M.)
| | - James M. McCaw
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia; (P.C.); (J.M.M.)
- Peter Doherty Institute for Infection and Immunity, The Royal Melbourne Hospital and The University of Melbourne, Parkville, VIC 3010, Australia
- Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
118
|
Nobs SP, Kopf M. Tissue-resident macrophages: guardians of organ homeostasis. Trends Immunol 2021; 42:495-507. [PMID: 33972166 DOI: 10.1016/j.it.2021.04.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Tissue-resident macrophages (MTR) have recently emerged as a key rheostat capable of regulating the balance between organ health and disease. In most organs, ontogenetically and functionally distinct macrophage subsets fulfill a plethora of functions specific to their tissue environment. In this review, we summarize recent findings regarding the ontogeny and functions of macrophage populations in different mammalian tissues, describing how these cells regulate tissue homeostasis and how they can contribute to inflammation. Furthermore, we highlight new developments concerning certain general principles of tissue macrophage biology, including the importance of metabolism for understanding macrophage activation states and the influence of intrinsic and extrinsic factors on macrophage metabolic control. We also shed light on certain open questions in the field and how answering these might pave the way for tissue-specific therapeutic approaches.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
119
|
Bertho N, Meurens F. The pig as a medical model for acquired respiratory diseases and dysfunctions: An immunological perspective. Mol Immunol 2021; 135:254-267. [PMID: 33933817 DOI: 10.1016/j.molimm.2021.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
By definition no model is perfect, and this also holds for biology and health sciences. In medicine, murine models are, and will be indispensable for long, thanks to their reasonable cost and huge choice of transgenic strains and molecular tools. On the other side, non-human primates remain the best animal models although their use is limited because of financial and obvious ethical reasons. In the field of respiratory diseases, specific clinical models such as sheep and cotton rat for bronchiolitis, or ferret and Syrian hamster for influenza and Covid-19, have been successfully developed, however, in these species, the toolbox for biological analysis remains scarce. In this view the porcine medical model is appearing as the third, intermediate, choice, between murine and primate. Herein we would like to present the pros and cons of pig as a model for acquired respiratory conditions, through an immunological point of view. Indeed, important progresses have been made in pig immunology during the last decade that allowed the precise description of immune molecules and cell phenotypes and functions. These progresses might allow the use of pig as clinical model of human respiratory diseases but also as a species of interest to perform basic research explorations.
Collapse
Affiliation(s)
| | - François Meurens
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon S7N5E3, Canada
| |
Collapse
|
120
|
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease. Respir Res 2021; 22:133. [PMID: 33926483 PMCID: PMC8082489 DOI: 10.1186/s12931-021-01722-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.
Collapse
|
121
|
Wang Q, Chen S, Li T, Yang Q, Liu J, Tao Y, Meng Y, Chen J, Feng X, Han Z, Shi M, Huang H, Han M, Jiang E. Critical Role of Lkb1 in the Maintenance of Alveolar Macrophage Self-Renewal and Immune Homeostasis. Front Immunol 2021; 12:629281. [PMID: 33968022 PMCID: PMC8100336 DOI: 10.3389/fimmu.2021.629281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/01/2021] [Indexed: 01/27/2023] Open
Abstract
Alveolar macrophages (AMs) are pivotal for maintaining lung immune homeostasis. We demonstrated that deletion of liver kinase b1 (Lkb1) in CD11c+ cells led to greatly reduced AM abundance in the lung due to the impaired self-renewal of AMs but not the impeded pre-AM differentiation. Mice with Lkb1-deficient AMs exhibited deteriorated diseases during airway Staphylococcus aureus (S. aureus) infection and allergic inflammation, with excessive accumulation of neutrophils and more severe lung pathology. Drug-mediated AM depletion experiments in wild type mice indicated a cause for AM reduction in aggravated diseases in Lkb1 conditional knockout mice. Transcriptomic sequencing also revealed that Lkb1 inhibited proinflammatory pathways, including IL-17 signaling and neutrophil migration, which might also contribute to the protective function of Lkb1 in AMs. We thus identified Lkb1 as a pivotal regulator that maintains the self-renewal and immune function of AMs.
Collapse
MESH Headings
- AMP-Activated Protein Kinases
- Animals
- Asthma/enzymology
- Asthma/genetics
- Asthma/immunology
- CD11 Antigens/genetics
- CD11 Antigens/metabolism
- Cell Self Renewal
- Disease Models, Animal
- Homeostasis
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Lung/enzymology
- Lung/immunology
- Lung/microbiology
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/microbiology
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Infiltration
- Pneumonia, Bacterial/enzymology
- Pneumonia, Bacterial/genetics
- Pneumonia, Bacterial/immunology
- Pneumonia, Bacterial/microbiology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction
- Staphylococcal Infections/enzymology
- Staphylococcal Infections/genetics
- Staphylococcal Infections/immunology
- Staphylococcal Infections/microbiology
- Transcriptome
- Mice
Collapse
Affiliation(s)
- Qianqian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Song Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tengda Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qiongmei Yang
- Department of Hematology, The First Affiliated Hospital of Kunming Medical University, Hematology Research Center of Yunnan Province, Kunming, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan Tao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuan Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jiadi Chen
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingxia Shi
- Department of Hematology, The First Affiliated Hospital of Kunming Medical University, Hematology Research Center of Yunnan Province, Kunming, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
122
|
Albers GJ, Iwasaki J, McErlean P, Ogger PP, Ghai P, Khoyratty TE, Udalova IA, Lloyd CM, Byrne AJ. IRF5 regulates airway macrophage metabolic responses. Clin Exp Immunol 2021; 204:134-143. [PMID: 33423291 PMCID: PMC7944363 DOI: 10.1111/cei.13573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Interferon regulatory factor 5 (IRF5) is a master regulator of macrophage phenotype and a key transcription factor involved in expression of proinflammatory cytokine responses to microbial and viral infection. Here, we show that IRF5 controls cellular and metabolic responses. By integrating ChIP sequencing (ChIP-Seq) and assay for transposase-accessible chromatin using sequencing (ATAC)-seq data sets, we found that IRF5 directly regulates metabolic genes such as hexokinase-2 (Hk2). The interaction of IRF5 and metabolic genes had a functional consequence, as Irf5-/- airway macrophages but not bone marrow-derived macrophages (BMDMs) were characterized by a quiescent metabolic phenotype at baseline and had reduced ability to utilize oxidative phosphorylation after Toll-like receptor (TLR)-3 activation, in comparison to controls, ex vivo. In a murine model of influenza infection, IRF5 deficiency had no effect on viral load in comparison to wild-type controls but controlled metabolic responses to viral infection, as IRF5 deficiency led to reduced expression of Sirt6 and Hk2. Together, our data indicate that IRF5 is a key component of AM metabolic responses following influenza infection and TLR-3 activation.
Collapse
Affiliation(s)
- G. J. Albers
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - J. Iwasaki
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - P. McErlean
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - P. P. Ogger
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - P. Ghai
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - T. E. Khoyratty
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - I. A. Udalova
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - C. M. Lloyd
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| | - A. J. Byrne
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
123
|
Xiao Q, He J, Lei A, Xu H, Zhang L, Zhou P, Jiang G, Zhou J. PPARγ enhances ILC2 function during allergic airway inflammation via transcription regulation of ST2. Mucosal Immunol 2021; 14:468-478. [PMID: 32811992 DOI: 10.1038/s41385-020-00339-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) represent the major player during hyperresponsive airway inflammation. Peroxisome proliferator-activated receptor-γ (PPARγ) was highly expressed on ILC2 and its potential role in asthma has been suggested. However, the detailed mechanism underlying the effects of PPARγ on ILC2-induced airway inflammation remains to be fully understood. Here we identified PPARγ as a positive regulator of lung ILC2. Expression of PPARγ on ILC2 was dramatically induced upon interleukin-33 (IL-33) challenge. Deficiency of PPARγ in hematopoietic system in mice (PPARγfl/fl Vav1Cre) significantly impaired the function of ILC2 in lung, which led to apparent alleviation of airway inflammation in response to IL-33 or Papain challenge, when compared with those in PPARγfl/fl littermates control. Mechanistic studies identified IL-33 receptor ST2 as a transcriptional target of PPARγ. Overexpression of ST2 rescued the functional defects of ILC2 lacking PPARγ. Collectively, these results demonstrated PPARγ as an important regulator of ILC2 during allergic airway inflammation, which sheds new lights on the importance of PPARγ in asthma.
Collapse
Affiliation(s)
- Qiang Xiao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China.,Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Juan He
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Aihua Lei
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Haixu Xu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China
| | - Lijuan Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China
| | - Pan Zhou
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China
| | - Guanmin Jiang
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin, China.
| |
Collapse
|
124
|
Suwankitwat N, Libby S, Liggitt HD, Avalos A, Ruddell A, Rosch JW, Park H, Iritani BM. The actin-regulatory protein Hem-1 is essential for alveolar macrophage development. J Exp Med 2021; 218:211806. [PMID: 33600594 PMCID: PMC7894047 DOI: 10.1084/jem.20200472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/16/2020] [Accepted: 01/13/2021] [Indexed: 01/27/2023] Open
Abstract
Hematopoietic protein-1 (Hem-1) is a hematopoietic cell–specific actin-regulatory protein. Loss-of-function (LOF) variants in the NCKAP1L gene encoding Hem-1 have recently been found to result in primary immunodeficiency disease (PID) in humans, characterized by recurring respiratory infections, asthma, and high mortality. However, the mechanisms of how Hem-1 variants result in PID are not known. In this study, we generated constitutive and myeloid cell–specific Nckap1l-KO mice to dissect the importance of Hem-1 in lung immunity. We found that Hem-1–deficient mice accumulated excessive surfactant and cell debris in airways (pulmonary alveolar proteinosis) due to impaired development of alveolar macrophages (AMs) and reduced expression of the AM differentiation factor Pparg. Residual Hem-1–deficient AMs shifted to a proinflammatory phenotype, and Hem-1–deficient neutrophils and monocytes failed to migrate normally. Myeloid cell–specific Hem-1–deficient mice exhibited increased morbidity following influenza A virus or Streptococcus pneumoniae challenge. These results provide potential mechanisms for how LOF variants in Hem-1 result in recurring respiratory diseases.
Collapse
Affiliation(s)
| | - Stephen Libby
- Department of Microbiology, University of Washington, Seattle, WA
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Alan Avalos
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Alanna Ruddell
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN
| | - Heon Park
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Brian M Iritani
- Department of Comparative Medicine, University of Washington, Seattle, WA
| |
Collapse
|
125
|
Bailey KL, Samuelson DR, Wyatt TA. Alcohol use disorder: A pre-existing condition for COVID-19? Alcohol 2021; 90:11-17. [PMID: 33080339 PMCID: PMC7568767 DOI: 10.1016/j.alcohol.2020.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Alcohol misuse is long established as a contributor to the pathophysiology of the lung. The intersection of multi-organ responses to alcohol-mediated tissue injury likely contributes to the modulation of lung in response to injury. Indeed, the negative impact of alcohol on susceptibility to infection and on lung barrier function is now well documented. Thus, the alcohol lung represents a very likely comorbidity for the negative consequences of both COVID-19 susceptibility and severity. In this review, we present the known alcohol misuse ramifications on the lung in the context of the current coronavirus pandemic.
Collapse
Affiliation(s)
- Kristina L Bailey
- Research Service, Department of Veterans Affairs Omaha-Western Iowa Health Care System, Omaha, NE, 68105, United States; Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Derrick R Samuelson
- Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Todd A Wyatt
- Research Service, Department of Veterans Affairs Omaha-Western Iowa Health Care System, Omaha, NE, 68105, United States; Pulmonary, Critical Care and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States; Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| |
Collapse
|
126
|
Does tissue imprinting restrict macrophage plasticity? Nat Immunol 2021; 22:118-127. [PMID: 33462453 DOI: 10.1038/s41590-020-00849-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023]
Abstract
Macrophages have long been considered as particularly plastic cells. However, recent work combining fate mapping, single-cell transcriptomics and epigenetics has undermined the macrophage plasticity dogma. Here, we discuss recent studies that have carefully dissected the response of individual macrophage subsets to pulmonary insults and call for an adjustment of the macrophage plasticity concept. We hypothesize that prolonged tissue residency shuts down much of the plasticity of macrophages and propose that the restricted plasticity of resident macrophages has been favored by evolution to safeguard tissue homeostasis. Recruited monocytes are more plastic and their differentiation into resident macrophages during inflammation can result in a dual imprinting from both the ongoing inflammation and the macrophage niche. This results in inflammation-imprinted resident macrophages, and we speculate that rewired niche circuits could maintain this inflammatory state. We believe that this revisited plasticity model offers opportunities to reset the macrophage pool after a severe inflammatory episode.
Collapse
|
127
|
Nobs SP, Pohlmeier L, Li F, Kayhan M, Becher B, Kopf M. GM-CSF instigates a dendritic cell-T-cell inflammatory circuit that drives chronic asthma development. J Allergy Clin Immunol 2021; 147:2118-2133.e3. [PMID: 33440200 DOI: 10.1016/j.jaci.2020.12.638] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/06/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Steroid-resistant asthma is often characterized by high levels of neutrophils and mixed TH2/TH17 immune profiles. Indeed, neutrophils are key drivers of chronic lung inflammation in multiple respiratory diseases. Their numbers correlate strongly with disease severity, and their presence is often associated with exacerbation of chronic lung inflammation. OBJECTIVE What factors drive development of neutrophil-mediated chronic lung disease remains largely unknown, and we sought to study the role of GM-CSF as a potential regulator in chronic asthma. METHODS Different experimental animal models of chronic asthma were used in combination with alveolar macrophage-reconstitution of global GM-CSF receptor knockout mice as well as cell-type-specific knockout animals to elucidate the role of GM-CSF signaling in chronic airway inflammation. RESULTS We identify GM-CSF signaling as a critical factor regulating pulmonary accumulation of neutrophils. We show that although being not required for intrinsically regulating neutrophil migration, GM-CSF controls lung dendritic cell function, which in turn promotes T-cell-dependent recruitment of neutrophils to the airways. We demonstrate that GM-CSF regulates lung dendritic cell antigen uptake, transport, and TH2/TH17 cell priming in an intrinsic fashion, which in turn drives pulmonary granulocyte recruitment and contributes to development of airway hyperresponsiveness in chronic disease. CONCLUSIONS We identify GM-CSF as a potentially novel therapeutic target in chronic lung inflammation, describing a GM-CSF-dependent lung conventional dendritic cell-T-cell-neutrophil axis that drives chronic lung disease.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lea Pohlmeier
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Fengqi Li
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
128
|
Abstract
ABSTRACT Macrophage, as an integral component of the immune system and the first responder to local damage, is on the front line of defense against infection. Over the past century, the prevailing view of macrophage origin states that all macrophage populations resided in tissues are terminally differentiated and replenished by monocytes from bone-marrow progenitors. Nonetheless, this theory has been reformed by ground-breaking discoveries from the past decades. It is now believed that tissue-resident macrophages (TRMs) are originated from the embryonic precursors and seeded in tissue prenatally. They can replenish via self-renewal throughout the lifespan. Indeed, recent studies have demonstrated that tissue-resident macrophages should not be classified by the over-simplified macrophage polarization (M1/M2) dogma during inflammation. Moreover, multiple lines of evidence have indicated that tissue-resident macrophages play critical roles in maintaining tissue homeostasis and facilitating tissue repair through controlling infection and resolving inflammation. In this review, we summarize the properties of resident macrophages in the lung, spleen, and heart, and further highlight the impact of TRM populations on inflammation control and tissue repair. We also discuss the potential role of local proliferation in maintaining a physiologically stable TRM pool in response to acute inflammation.
Collapse
Affiliation(s)
- Xingjiang Mu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | |
Collapse
|
129
|
Ural BB, Yeung ST, Damani-Yokota P, Devlin JC, de Vries M, Vera-Licona P, Samji T, Sawai CM, Jang G, Perez OA, Pham Q, Maher L, Loke P, Dittmann M, Reizis B, Khanna KM. Identification of a nerve-associated, lung-resident interstitial macrophage subset with distinct localization and immunoregulatory properties. Sci Immunol 2020; 5:5/45/eaax8756. [PMID: 32220976 DOI: 10.1126/sciimmunol.aax8756] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/28/2019] [Accepted: 03/05/2020] [Indexed: 12/16/2022]
Abstract
Tissue-resident macrophages are a diverse population of cells that perform specialized functions including sustaining tissue homeostasis and tissue surveillance. Here, we report an interstitial subset of CD169+ lung-resident macrophages that are transcriptionally and developmentally distinct from alveolar macrophages (AMs). They are primarily localized around the airways and are found in close proximity to the sympathetic nerves in the bronchovascular bundle. These nerve- and airway-associated macrophages (NAMs) are tissue resident, yolk sac derived, self-renewing, and do not require CCR2+ monocytes for development or maintenance. Unlike AMs, the development of NAMs requires CSF1 but not GM-CSF. Bulk population and single-cell transcriptome analysis indicated that NAMs are distinct from other lung-resident macrophage subsets and highly express immunoregulatory genes under steady-state and inflammatory conditions. NAMs proliferated robustly after influenza infection and activation with the TLR3 ligand poly(I:C), and in their absence, the inflammatory response was augmented, resulting in excessive production of inflammatory cytokines and innate immune cell infiltration. Overall, our study provides insights into a distinct subset of airway-associated pulmonary macrophages that function to maintain immune and tissue homeostasis.
Collapse
Affiliation(s)
- Basak B Ural
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Stephen T Yeung
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Payal Damani-Yokota
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Joseph C Devlin
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Maren de Vries
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Paola Vera-Licona
- Center for Quantitative Medicine, Uconn Health, Farmington, CT 06030, USA.,Department of Cell Biology, Department of Cell Biology, UConn Health, Farmington, CT 06030, USA.,Department of Pediatrics, UConn Health, Farmington, CT 06030, USA.,Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Tasleem Samji
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | | | - Geunhyo Jang
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Oriana A Perez
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Quynh Pham
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Leigh Maher
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - P'ng Loke
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Boris Reizis
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA.,Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA. .,Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
130
|
Bissonnette EY, Lauzon-Joset JF, Debley JS, Ziegler SF. Cross-Talk Between Alveolar Macrophages and Lung Epithelial Cells is Essential to Maintain Lung Homeostasis. Front Immunol 2020; 11:583042. [PMID: 33178214 PMCID: PMC7593577 DOI: 10.3389/fimmu.2020.583042] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022] Open
Abstract
The main function of the lung is to perform gas exchange while maintaining lung homeostasis despite environmental pathogenic and non-pathogenic elements contained in inhaled air. Resident cells must keep lung homeostasis and eliminate pathogens by inducing protective immune response and silently remove innocuous particles. Which lung cell type is crucial for this function is still subject to debate, with reports favoring either alveolar macrophages (AMs) or lung epithelial cells (ECs) including airway and alveolar ECs. AMs are the main immune cells in the lung in steady-state and their function is mainly to dampen inflammatory responses. In addition, they phagocytose inhaled particles and apoptotic cells and can initiate and resolve inflammatory responses to pathogens. Although AMs release a plethora of mediators that modulate immune responses, ECs also play an essential role as they are more than just a physical barrier. They produce anti-microbial peptides and can secrete a variety of mediators that can modulate immune responses and AM functions. Furthermore, ECs can maintain AMs in a quiescent state by expressing anti-inflammatory membrane proteins such as CD200. Thus, AMs and ECs are both very important to maintain lung homeostasis and have to coordinate their action to protect the organism against infection. Thus, AMs and lung ECs communicate with each other using different mechanisms including mediators, membrane glycoproteins and their receptors, gap junction channels, and extracellular vesicles. This review will revisit characteristics and functions of AMs and lung ECs as well as different communication mechanisms these cells utilize to maintain lung immune balance and response to pathogens. A better understanding of the cross-talk between AMs and lung ECs may help develop new therapeutic strategies for lung pathogenesis.
Collapse
Affiliation(s)
- Elyse Y Bissonnette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Jean-François Lauzon-Joset
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Steven F Ziegler
- Department of Immunology, Benaroya Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
131
|
Xu M, Yang W, Wang X, Nayak DK. Lung Secretoglobin Scgb1a1 Influences Alveolar Macrophage-Mediated Inflammation and Immunity. Front Immunol 2020; 11:584310. [PMID: 33117399 PMCID: PMC7558713 DOI: 10.3389/fimmu.2020.584310] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/07/2020] [Indexed: 01/23/2023] Open
Abstract
Alveolar macrophage (AM) is a mononuclear phagocyte key to the defense against respiratory infections. To understand AM’s role in airway disease development, we examined the influence of Secretoglobin family 1a member 1 (SCGB1A1), a pulmonary surfactant protein, on AM development and function. In a murine model, high-throughput RNA-sequencing and gene expression analyses were performed on purified AMs isolated from mice lacking in Scgb1a1 gene and were compared with that from mice expressing the wild type Scgb1a1 at weaning (4 week), puberty (8 week), early adult (12 week), and middle age (40 week). AMs from early adult mice under Scgb1a1 sufficiency demonstrated a total of 37 up-regulated biological pathways compared to that at weaning, from which 30 were directly involved with antigen presentation, anti-viral immunity and inflammation. Importantly, these pathways under Scgb1a1 deficiency were significantly down-regulated compared to that in the age-matched Scgb1a1-sufficient counterparts. Furthermore, AMs from Scgb1a1-deficient mice showed an early activation of inflammatory pathways compared with that from Scgb1a1-sufficient mice. Our in vitro experiments with AM culture established that exogenous supplementation of SCGB1a1 protein significantly reduced AM responses to microbial stimuli where SCGB1a1 was effective in blunting the release of cytokines and chemokines (including IL-1b, IL-6, IL-8, MIP-1a, TNF-a, and MCP-1). Taken together, these findings suggest an important role for Scgb1a1 in shaping the AM-mediated inflammation and immune responses, and in mitigating cytokine surges in the lungs.
Collapse
Affiliation(s)
- Min Xu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Xuanchuan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Deepak Kumar Nayak
- Interdisciplinary Oncology, University of Arizona College of Medicine, Phoenix, AZ, United States
| |
Collapse
|
132
|
Hwang HS, Chang M, Kim YA. Influenza-Host Interplay and Strategies for Universal Vaccine Development. Vaccines (Basel) 2020; 8:vaccines8030548. [PMID: 32962304 PMCID: PMC7564814 DOI: 10.3390/vaccines8030548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022] Open
Abstract
Influenza is an annual epidemic and an occasional pandemic caused by pathogens that are responsible for infectious respiratory disease. Humans are highly susceptible to the infection mediated by influenza A viruses (IAV). The entry of the virus is mediated by the influenza virus hemagglutinin (HA) glycoprotein that binds to the cellular sialic acid receptors and facilitates the fusion of the viral membrane with the endosomal membrane. During IAV infection, virus-derived pathogen-associated molecular patterns (PAMPs) are recognized by host intracellular specific sensors including toll-like receptors (TLRs), C-type lectin receptors, retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) either on the cell surface or intracellularly in endosomes. Herein, we comprehensively review the current knowledge available on the entry of the influenza virus into host cells and the molecular details of the influenza virus–host interface. We also highlight certain strategies for the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Hye Suk Hwang
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
| | - Mincheol Chang
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju 61186, Korea
- School of Polymer Science and Engineering, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (M.C.); (Y.A.K.); Tel.: +82-62-530-1771 (M.C.); +82-62-530-1871 (Y.A.K.)
| | - Yoong Ahm Kim
- Alan G. MacDiarmid Energy Research Institute, Chonnam National University, Gwangju 61186, Korea;
- Department of Polymer Engineering, Graduate School, Chonnam National University, Gwangju 61186, Korea
- School of Polymer Science and Engineering, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (M.C.); (Y.A.K.); Tel.: +82-62-530-1771 (M.C.); +82-62-530-1871 (Y.A.K.)
| |
Collapse
|
133
|
What Makes the Lung Unique – Tissue-Specific Immunity in the Respiratory Tract. EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The immune system constitutes a critical mechanism of the human body to preserve health and mitigate disease. In the lung, immunity is seen as a critical driver in many respiratory diseases, in particular in those characterised by aberrant inflammation, such as chronic obstructive pulmonary disease, fibrosis, and asthma. In this review, the specialised set of immune cells and lung tissue-specific regulators, including key cytokines such as granulocyte-macrophage colony-stimulating factor and transforming growth factor β, that control immune responses in the respiratory tract will be discussed. Furthermore, the current understanding of the impact of key environmental components such as the role of oxygen and lung microbiota on lung immunity will be highlighted. The goal is to identify the unique aspects of lung immune biology to facilitate insights into the aetiology of common lung inflammatory diseases and to provide the basis for a deeper mechanistic understanding of the underlying immune processes. Finally, key future avenues of research such as using more comprehensive quantitative approaches for elucidating molecular disease mechanisms as well as the potential to exploit tissue-specific regulators of immunity for therapy of lung inflammatory disorders will be discussed.
Collapse
|
134
|
Verma AK, Bansal S, Bauer C, Muralidharan A, Sun K. Influenza Infection Induces Alveolar Macrophage Dysfunction and Thereby Enables Noninvasive Streptococcus pneumoniae to Cause Deadly Pneumonia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1601-1607. [PMID: 32796026 PMCID: PMC7484308 DOI: 10.4049/jimmunol.2000094] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023]
Abstract
Secondary Streptococcus pneumoniae infection is a significant cause of morbidity and mortality during influenza epidemics and pandemics. Multiple pathogenic mechanisms, such as lung epithelial damage and dysregulation of neutrophils and alveolar macrophages (AMs), have been suggested to contribute to the severity of disease. However, the fundamental reasons for influenza-induced susceptibility to secondary bacterial pneumonia remain unclear. In this study, we revisited these controversies over key pathogenic mechanisms in a lethal model of secondary bacterial pneumonia with an S. pneumoniae strain that is innocuous to mice in the absence of influenza infection. Using a series of in vivo models, we demonstrate that rather than a systemic suppression of immune responses or neutrophil function, influenza infection activates IFN-γR signaling and abrogates AM-dependent bacteria clearance and thereby causes extreme susceptibility to pneumococcal infection. Importantly, using mice carrying conditional knockout of Ifngr1 gene in different myeloid cell subsets, we demonstrate that influenza-induced IFN-γR signaling in AMs impairs their antibacterial function, thereby enabling otherwise noninvasive S. pneumoniae to cause deadly pneumonia.
Collapse
Affiliation(s)
- Atul K Verma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Shruti Bansal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Christopher Bauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Abenaya Muralidharan
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Keer Sun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
135
|
Neupane AS, Willson M, Chojnacki AK, Vargas E Silva Castanheira F, Morehouse C, Carestia A, Keller AE, Peiseler M, DiGiandomenico A, Kelly MM, Amrein M, Jenne C, Thanabalasuriar A, Kubes P. Patrolling Alveolar Macrophages Conceal Bacteria from the Immune System to Maintain Homeostasis. Cell 2020; 183:110-125.e11. [PMID: 32888431 DOI: 10.1016/j.cell.2020.08.020] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 07/14/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
During respiration, humans breathe in more than 10,000 liters of non-sterile air daily, allowing some pathogens access to alveoli. Interestingly, alveoli outnumber alveolar macrophages (AMs), which favors alveoli devoid of AMs. If AMs, like most tissue macrophages, are sessile, then this numerical advantage would be exploited by pathogens unless neutrophils from the blood stream intervened. However, this would translate to omnipresent persistent inflammation. Developing in vivo real-time intravital imaging of alveoli revealed AMs crawling in and between alveoli using the pores of Kohn. Importantly, these macrophages sensed, chemotaxed, and, with high efficiency, phagocytosed inhaled bacterial pathogens such as P. aeruginosa and S. aureus, cloaking the bacteria from neutrophils. Impairing AM chemotaxis toward bacteria induced superfluous neutrophil recruitment, leading to inappropriate inflammation and injury. In a disease context, influenza A virus infection impaired AM crawling via the type II interferon signaling pathway, and this greatly increased secondary bacterial co-infection.
Collapse
Affiliation(s)
- Arpan Sharma Neupane
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michelle Willson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Fernanda Vargas E Silva Castanheira
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Christopher Morehouse
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Agostina Carestia
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ashley Elaine Keller
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Moritz Peiseler
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Antonio DiGiandomenico
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Margaret Mary Kelly
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Matthias Amrein
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Craig Jenne
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ajitha Thanabalasuriar
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal Canada H3G1Y6; Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA.
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
136
|
Chen S, Yan D, Qiu A. The role of macrophages in pulmonary hypertension: Pathogenesis and targeting. Int Immunopharmacol 2020; 88:106934. [PMID: 32889242 DOI: 10.1016/j.intimp.2020.106934] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/22/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a pathophysiological disorder that can complicate most cardiovascular and respiratory diseases and may involve multiple clinical conditions, but its pathogenesis is poorly understood. Despite recent developments in the management of PH, there is an urgent need for new ways to effectively treat PH and reduce the risk of further complications. Recent studies have shown that dysregulated immunity underlies the development of PH. Myeloid cells, including monocytes and macrophages, participate in immune homeostasis and the adaptive immune response, but the function and production of these cells in PH is not well understood. A prominent pathological feature of pH is the accumulation of macrophages near the arterioles of the lung, indicating that pulmonary inflammation mediated by lung perivascular macrophages is a key driver of pulmonary remodelling, which leads to increased right ventricular systolic pressure. An improved understanding of the roles macrophages play in immune responses associated with PH may lead to new therapeutic targets. In this review, we highlight the relationship between macrophages and PH, the molecular mechanisms involved, and the recent advances in targeting these processes to treat PH.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Dongmei Yan
- Department of Clinical Laboratory, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Aimin Qiu
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China.
| |
Collapse
|
137
|
Percopo CM, Ma M, Mai E, Redes JL, Kraemer LS, Minai M, Moore IN, Druey KM, Rosenberg HF. Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection. Viruses 2020; 12:v12090946. [PMID: 32867061 PMCID: PMC7552021 DOI: 10.3390/v12090946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic inhalation of fungi and fungal components has been linked to the development of respiratory disorders, although their role with respect to the pathogenesis of acute respiratory virus infection remains unclear. Here, we evaluate inflammatory pathology induced by repetitive administration of a filtrate of the ubiquitous fungus, Alternaria alternata, and its impact on susceptibility to infection with influenza A. We showed previously that A. alternata at the nasal mucosae resulted in increased susceptibility to an otherwise sublethal inoculum of influenza A in wild-type mice. Here we demonstrate that A. alternata-induced potentiation of influenza A infection was not dependent on fungal serine protease or ribonuclease activity. Repetitive challenge with A. alternata prior to virus infection resulted proinflammatory cytokines, neutrophil recruitment, and loss of alveolar macrophages to a degree that substantially exceeded that observed in response to influenza A infection alone. Concomitant administration of immunomodulatory Lactobacillus plantarum, a strategy shown previously to limit virus-induced inflammation in the airways, blocked the exaggerated lethal response. These observations promote an improved understanding of severe influenza infection with potential clinical relevance for individuals subjected to continuous exposure to molds and fungi.
Collapse
Affiliation(s)
- Caroline M. Percopo
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (C.M.P.); (M.M.); (E.M.); (L.S.K.)
| | - Michelle Ma
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (C.M.P.); (M.M.); (E.M.); (L.S.K.)
| | - Eric Mai
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (C.M.P.); (M.M.); (E.M.); (L.S.K.)
| | - Jamie L. Redes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (J.L.R.); (K.M.D.)
| | - Laura S. Kraemer
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (C.M.P.); (M.M.); (E.M.); (L.S.K.)
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.M.); (I.N.M.)
| | - Ian N. Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.M.); (I.N.M.)
| | - Kirk M. Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (J.L.R.); (K.M.D.)
| | - Helene F. Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (C.M.P.); (M.M.); (E.M.); (L.S.K.)
- Correspondence: ; Tel.: +1-301-761-6682
| |
Collapse
|
138
|
Schneider DJ, Smith KA, Latuszek CE, Wilke CA, Lyons DM, Penke LR, Speth JM, Marthi M, Swanson JA, Moore BB, Lauring AS, Peters‐Golden M. Alveolar macrophage-derived extracellular vesicles inhibit endosomal fusion of influenza virus. EMBO J 2020; 39:e105057. [PMID: 32643835 PMCID: PMC7429743 DOI: 10.15252/embj.2020105057] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 01/09/2023] Open
Abstract
Alveolar macrophages (AMs) and epithelial cells (ECs) are the lone resident lung cells positioned to respond to pathogens at early stages of infection. Extracellular vesicles (EVs) are important vectors of paracrine signaling implicated in a range of (patho)physiologic contexts. Here we demonstrate that AMs, but not ECs, constitutively secrete paracrine activity localized to EVs which inhibits influenza infection of ECs in vitro and in vivo. AMs exposed to cigarette smoke extract lost the inhibitory activity of their secreted EVs. Influenza strains varied in their susceptibility to inhibition by AM-EVs. Only those exhibiting early endosomal escape and high pH of fusion were inhibited via a reduction in endosomal pH. By contrast, strains exhibiting later endosomal escape and lower fusion pH proved resistant to inhibition. These results extend our understanding of how resident AMs participate in host defense and have broader implications in the defense and treatment of pathogens internalized within endosomes.
Collapse
Affiliation(s)
- Daniel J Schneider
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Katherine A Smith
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Catrina E Latuszek
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Carol A Wilke
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Danny M Lyons
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Infectious DiseaseDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Loka R Penke
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Matangi Marthi
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Joel A Swanson
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Bethany B Moore
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Graduate Program in ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Adam S Lauring
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Infectious DiseaseDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Graduate Program in ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Marc Peters‐Golden
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Graduate Program in ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
139
|
Sica A, Colombo MP, Trama A, Horn L, Garassino MC, Torri V. Immunometabolic Status of COVID-19 Cancer Patients. Physiol Rev 2020; 100:1839-1850. [PMID: 32721181 PMCID: PMC7839651 DOI: 10.1152/physrev.00018.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cancer patients appear to be more likely to be diagnosed with coronavirus disease 2019 (COVID-19). This is supported by the understanding of immunometabolic pathways that intersect patients with infection and cancer. However, data derived by case series and retrospective studies do not offer a coherent interpretation, since data from China suggest an increased risk of COVID-19, while data from the United States and Italy show a prevalence of COVID-19 in cancer patients comparable with the general population. Noteworthy, cancer and COVID-19 exploit distinct patterns of macrophage activation that promote disease progression in the most severe forms. In particular, the alternative activation of M2-polarized macrophages plays a crucial role in cancer progression. In contrast, the macrophage-activation syndrome appears as the source of M1-related cytokine storm in severe COVID-19 disease, thus indicating macrophages as a source of distinct inflammatory states in the two diseases, nonetheless as a common therapeutic target. New evidence indicates that NAMPT/NAD metabolism can direct both innate immune cell effector functions and the homeostatic robustness, in both cancer and infection. Moreover, a bidirectional relationship exists between the metabolism of NAD and the protective role that angiotensin converting enzyme 2, the COVID-19 receptor, can play against hyperinflammation. Within this immunometabolic framework, the review considers possible interference mechanisms that viral infections and tumors elicit on therapies and provides an overview for the management of patients with cancer affected by COVID-19, particularly for the balance of risk and benefit when planning normally routine cancer treatments and follow-up appointments.
Collapse
Affiliation(s)
- A Sica
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| | - M P Colombo
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| | - A Trama
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| | - L Horn
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| | - M C Garassino
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| | - V Torri
- Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro," Novara, Italy; Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Evaluative Epidemiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee; Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy; and Clinical Research Lab, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri," Milan, Italy
| |
Collapse
|
140
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Nair A, Dib S, Yemelyanov A, Gates KL, Budinger GRS, Beitel GJ, Sporn PHS. Hypercapnia Suppresses Macrophage Antiviral Activity and Increases Mortality of Influenza A Infection via Akt1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:489-501. [PMID: 32540997 PMCID: PMC7343622 DOI: 10.4049/jimmunol.2000085] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
Hypercapnia (HC), elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that HC inhibits multiple macrophage and neutrophil antimicrobial functions and increases the mortality of bacterial pneumonia in mice. In this study, we show that normoxic HC increases viral replication, lung injury, and mortality in mice infected with influenza A virus (IAV). Elevated CO2 increased IAV replication and inhibited antiviral gene and protein expression in macrophages in vivo and in vitro. HC potentiated IAV-induced activation of Akt, whereas specific pharmacologic inhibition or short hairpin RNA knockdown of Akt1 in alveolar macrophages blocked HC's effects on IAV growth and the macrophage antiviral response. Our findings suggest that targeting Akt1 or the downstream pathways through which elevated CO2 signals could enhance macrophage antiviral host defense and improve clinical outcomes in hypercapnic patients with advanced lung disease.
Collapse
Affiliation(s)
- S Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611;
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Francisco J Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Aisha Nair
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Sandra Dib
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Alex Yemelyanov
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Khalilah L Gates
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612; and
| | - Greg J Beitel
- Department of Molecular Biosciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208
| | - Peter H S Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612; and
| |
Collapse
|
141
|
Bellomo A, Mondor I, Spinelli L, Lagueyrie M, Stewart BJ, Brouilly N, Malissen B, Clatworthy MR, Bajénoff M. Reticular Fibroblasts Expressing the Transcription Factor WT1 Define a Stromal Niche that Maintains and Replenishes Splenic Red Pulp Macrophages. Immunity 2020; 53:127-142.e7. [PMID: 32562599 DOI: 10.1016/j.immuni.2020.06.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/20/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Located within red pulp cords, splenic red pulp macrophages (RPMs) are constantly exposed to the blood flow, clearing senescent red blood cells (RBCs) and recycling iron from hemoglobin. Here, we studied the mechanisms underlying RPM homeostasis, focusing on the involvement of stromal cells as these cells perform anchoring and nurturing macrophage niche functions in lymph nodes and liver. Microscopy revealed that RPMs are embedded in a reticular meshwork of red pulp fibroblasts characterized by the expression of the transcription factor Wilms' Tumor 1 (WT1) and colony stimulating factor 1 (CSF1). Conditional deletion of Csf1 in WT1+ red pulp fibroblasts, but not white pulp fibroblasts, drastically altered the RPM network without altering circulating CSF1 levels. Upon RPM depletion, red pulp fibroblasts transiently produced the monocyte chemoattractants CCL2 and CCL7, thereby contributing to the replenishment of the RPM network. Thus, red pulp fibroblasts anchor and nurture RPM, a function likely conserved in humans.
Collapse
Affiliation(s)
- Alicia Bellomo
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | | | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nicolas Brouilly
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille, Marseille, France
| | | | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Marc Bajénoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
142
|
Abstract
Although, as the major organ of gas exchange, the lung is considered a nonlymphoid organ, an interconnected network of lung-resident innate cells, including epithelial cells, dendritic cells, macrophages, and natural killer cells is crucial for its protection. These cells provide defense against a daily assault by airborne bacteria, viruses, and fungi, as well as prevent the development of cancer, allergy, and the outgrowth of commensals. Our understanding of this innate immune environment has recently changed with the discovery of a family of innate lymphoid cells (ILCs): ILC1s, ILC2s, and ILC3s. All lack adaptive antigen receptors but can provide a substantial and rapid source of IFN-γ, IL-5 and IL-13, and IL-17A or IL-22, respectively. Their ability to afford immediate protection to the lung and to influence subsequent adaptive immune responses highlights the importance of understanding ILC-regulated immunity for the design of future therapeutic interventions.
Collapse
Affiliation(s)
- Jillian L Barlow
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| | - Andrew N J McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| |
Collapse
|
143
|
Abstract
It has been over 100 years since the 1918 influenza pandemic, one of the most infamous examples of viral immunopathology. Since that time, there has been an inevitable repetition of influenza pandemics every few decades and yearly influenza seasons, which have a significant impact on human health. Recently, noteworthy progress has been made in defining the cellular and molecular mechanisms underlying pathology induced by an exuberant host response to influenza virus infection. Infection with influenza viruses is associated with a wide spectrum of disease, from mild symptoms to severe complications including respiratory failure, and the severity of influenza disease is driven by a complex interplay of viral and host factors. This chapter will discuss mechanisms of infection severity using concepts of disease resistance and tolerance as a framework for understanding the balance between viral clearance and immunopathology. We review mechanistic studies in animal models of infection and correlational studies in humans that have begun to define these factors and discuss promising host therapeutic targets to improve outcomes from severe influenza disease.
Collapse
Affiliation(s)
- David F Boyd
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Taylor L Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
144
|
Baasch S, Ruzsics Z, Henneke P. Cytomegaloviruses and Macrophages-Friends and Foes From Early on? Front Immunol 2020; 11:793. [PMID: 32477336 PMCID: PMC7235172 DOI: 10.3389/fimmu.2020.00793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/07/2020] [Indexed: 01/01/2023] Open
Abstract
Starting at birth, newborn infants are exposed to numerous microorganisms. Adaptation of the innate immune system to them is a delicate process, with potentially advantageous and harmful implications for health development. Cytomegaloviruses (CMVs) are highly adapted to their specific mammalian hosts, with which they share millions of years of co-evolution. Throughout the history of mankind, human CMV has infected most infants in the first months of life without overt implications for health. Thus, CMV infections are intertwined with normal immune development. Nonetheless, CMV has retained substantial pathogenicity following infection in utero or in situations of immunosuppression, leading to pathology in virtually any organ and particularly the central nervous system (CNS). CMVs enter the host through mucosal interfaces of the gastrointestinal and respiratory tract, where macrophages (MACs) are the most abundant immune cell type. Tissue MACs and their potential progenitors, monocytes, are established target cells of CMVs. Recently, several discoveries have revolutionized our understanding on the pre- and postnatal development and site-specific adaptation of tissue MACs. In this review, we explore experimental evidences and concepts on how CMV infections may impact on MAC development and activation as part of host-virus co-adaptation.
Collapse
Affiliation(s)
- Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
145
|
Jung HE, Lee HK. Host Protective Immune Responses against Influenza A Virus Infection. Viruses 2020; 12:v12050504. [PMID: 32375274 PMCID: PMC7291249 DOI: 10.3390/v12050504] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022] Open
Abstract
Influenza viruses cause infectious respiratory disease characterized by fever, myalgia, and congestion, ranging in severity from mild to life-threating. Although enormous efforts have aimed to prevent and treat influenza infections, seasonal and pandemic influenza outbreaks remain a major public health concern. This is largely because influenza viruses rapidly undergo genetic mutations that restrict the long-lasting efficacy of vaccine-induced immune responses and therapeutic regimens. In this review, we discuss the virological features of influenza A viruses and provide an overview of current knowledge of the innate sensing of invading influenza viruses and the protective immune responses in the host.
Collapse
Affiliation(s)
- Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| |
Collapse
|
146
|
Liu H, You L, Wu J, Zhao M, Guo R, Zhang H, Su R, Mao Q, Deng D, Hao Y. Berberine suppresses influenza virus-triggered NLRP3 inflammasome activation in macrophages by inducing mitophagy and decreasing mitochondrial ROS. J Leukoc Biol 2020; 108:253-266. [PMID: 32272506 DOI: 10.1002/jlb.3ma0320-358rr] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022] Open
Abstract
Berberine (BBR) is an isoquinoline alkaloid extracted from several commonly used Chinese herbs. Our previous studies demonstrated BBR-mediated alleviation of lung injury due to inflammation and decrease in the mortality of mice with influenza viral pneumonia. The recent argument of autophagy against inflammatory responses has aroused wide concerns. This study focuses on the reactive oxygen species-Nod-like receptor protein 3 (ROS-NLRP3) pathway to investigate whether BBR inhibits NLRP3 inflammasome activation by inducing mitophagy. Our results demonstrate that BBR and mitochondrion-targeted superoxide dismutase mimetic (Mito-TEMPO; a specific mitochondrial ROS scavenger) significantly restricted NLRP3 inflammasome activation, increased mitochondrial membrane potential (MMP), and decreased mitochondrial ROS (mtROS) generation in J774A.1 macrophages infected with PR8 influenza virus. These observations suggest that the inhibitory effects of BBR on NLRP3 inflammasome activation were associated with the amelioration of mtROS generation. BBR treatment induced regular mitophagy, as evident from the increase in microtubule-associated protein 1 light chain 3 II, decrease in p62, colocalization of LC3 and mitochondria, and formation of autophagosomes. However, 3-methyladenine, an autophagy inhibitor, reversed the inhibitory effects of BBR on mitochondrial damage and NLRP3 inflammasome activation in influenza virus-infected macrophages, indicating the involvement of mitophagy in mediating the inhibitory effects of BBR on NLRP3 inflammasome activation. Furthermore, the knockdown of Bcl-2/adenovirus E18-19-kDa interacting protein 3 (BNIP3) expression attenuated the effects of BBR on mitophagy induction to some extent, suggesting that the BBR-induced mitophagy may be, at least in part, mediated in a BNIP3-dependent manner. Similar results were obtained in vivo using a mouse model of influenza viral pneumonia that was administered with BBR. Taken together, these findings suggest that restricting NLRP3 inflammasome activation by decreasing ROS generation through mitophagy induction may be crucial for the BBR-mediated alleviation of influenza virus-induced inflammatory lesions.
Collapse
Affiliation(s)
- Hui Liu
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Wu
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Mengfan Zhao
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Guo
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Haili Zhang
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Rina Su
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Qin Mao
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Di Deng
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Hao
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
147
|
Lauzon-Joset JF, Scott NM, Mincham KT, Stumbles PA, Holt PG, Strickland DH. Pregnancy Induces a Steady-State Shift in Alveolar Macrophage M1/M2 Phenotype That Is Associated With a Heightened Severity of Influenza Virus Infection: Mechanistic Insight Using Mouse Models. J Infect Dis 2020; 219:1823-1831. [PMID: 30576502 DOI: 10.1093/infdis/jiy732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Influenza virus infection during pregnancy is associated with enhanced disease severity. However, the underlying mechanisms are still not fully understood. We hypothesized that normal alveolar macrophage (AM) functions, which are central to maintaining lung immune homeostasis, are altered during pregnancy and that this dysregulation contributes to the increased inflammatory response to influenza virus infection. METHODS Time-mated BALB/c mice were infected with a low dose of H1N1 influenza A virus at gestation day 9.5. Inflammatory cells in bronchoalveolar lavage (BAL) fluid were assessed by flow cytometry. RESULTS Our findings confirm previous reports of increased severity of influenza virus infection in pregnant mice. The heightened inflammatory response detected in BAL fluid from infected pregnant mice was characterized by neutrophil-rich inflammation with concomitantly reduced numbers of AM, which were slower to return to baseline counts, compared with nonpregnant infected mice. The increased infection severity and inflammatory responses to influenza during pregnancy were associated with a pregnancy-induced shift in AM phenotype at homeostatic baseline, from the M1 (ie, classical activation) state toward the M2 (ie, alternative activation) state, as evidence by increased expression of CD301 and reduced levels of CCR7. CONCLUSION These results show that pregnancy is associated with an alternatively activated phenotype of AM before infection, which may contribute to heightened disease severity.
Collapse
Affiliation(s)
| | - Naomi M Scott
- Telethon Kids Institute, University of Western Australia, Nedlands
| | - Kyle T Mincham
- Telethon Kids Institute, University of Western Australia, Nedlands.,School of Medicine, University of Western Australia, Crawley
| | - Philip A Stumbles
- Telethon Kids Institute, University of Western Australia, Nedlands.,School of Veterinary and Life Science, Murdoch University, Perth, Australia
| | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Nedlands
| | | |
Collapse
|
148
|
Xing Z, Afkhami S, Bavananthasivam J, Fritz DK, D'Agostino MR, Vaseghi-Shanjani M, Yao Y, Jeyanathan M. Innate immune memory of tissue-resident macrophages and trained innate immunity: Re-vamping vaccine concept and strategies. J Leukoc Biol 2020; 108:825-834. [PMID: 32125045 DOI: 10.1002/jlb.4mr0220-446r] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023] Open
Abstract
In the past few years, our understanding of immunological memory has evolved remarkably due to a growing body of new knowledge in innate immune memory and immunity. Immunological memory now encompasses both innate and adaptive immune memory. The hypo-reactive and hyper-reactive types of innate immune memory lead to a suppressed and enhanced innate immune protective outcome, respectively. The latter is also named trained innate immunity (TII). The emerging information on innate immune memory has not only shed new light on the mechanisms of host defense but is also revolutionizing our long-held view of vaccination and vaccine strategies. Our current review will examine recent progress and knowledge gaps in innate immune memory with a focus on tissue-resident Mϕs, particularly lung Mϕs, and their relationship to local antimicrobial innate immunity. We will also discuss the impact of innate immune memory and TII on our understanding of vaccine concept and strategies and the significance of respiratory mucosal route of vaccination against respiratory pathogens.
Collapse
Affiliation(s)
- Zhou Xing
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jegarubee Bavananthasivam
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dominik K Fritz
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Maryam Vaseghi-Shanjani
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yushi Yao
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Current affiliation: Department of Immunology, Zhejiang University, Zhejiang, China
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
149
|
Aguilera ER, Lenz LL. Inflammation as a Modulator of Host Susceptibility to Pulmonary Influenza, Pneumococcal, and Co-Infections. Front Immunol 2020; 11:105. [PMID: 32117259 PMCID: PMC7026256 DOI: 10.3389/fimmu.2020.00105] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/15/2020] [Indexed: 12/24/2022] Open
Abstract
Bacterial and viral pathogens are predominant causes of pulmonary infections and complications. Morbidity and mortality from these infections is increased in populations that include the elderly, infants, and individuals with genetic disorders such as Down syndrome. Immune senescence, concurrent infections, and other immune alterations occur in these susceptible populations, but the underlying mechanisms that dictate increased susceptibility to lung infections are not fully defined. Here, we review unique features of the lung as a mucosal epithelial tissue and aspects of inflammatory and immune responses in model pulmonary infections and co-infections by influenza virus and Streptococcus pneumoniae. In these models, lung inflammatory responses are a double-edged sword: recruitment of immune effectors is essential to eliminate bacteria and virus-infected cells, but inflammatory cytokines drive changes in the lung conducive to increased pathogen replication. Excessive accumulation of inflammatory cells also hinders lung function, possibly causing death of the host. Some animal studies have found that targeting host modulators of lung inflammatory responses has therapeutic or prophylactic effects in these infection and co-infection models. However, conflicting results from other studies suggest microbiota, sequence of colonization, or other unappreciated aspects of lung biology also play important roles in the outcome of infections. Regardless, a predisposition to excessive or aberrant inflammatory responses occurs in susceptible human populations. Hence, in appropriate contexts, modulation of inflammatory responses may prove effective for reducing the frequency or severity of pulmonary infections. However, there remain limitations in our understanding of how this might best be achieved—particularly in diverse human populations.
Collapse
Affiliation(s)
- Elizabeth R Aguilera
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
150
|
LeMessurier KS, Tiwary M, Morin NP, Samarasinghe AE. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front Immunol 2020; 11:3. [PMID: 32117216 PMCID: PMC7011736 DOI: 10.3389/fimmu.2020.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022] Open
Abstract
The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Meenakshi Tiwary
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Nicholas P Morin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Critical Care Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| |
Collapse
|