101
|
Pulmonary immune cell transcriptome changes in double-hit model of BPD induced by chorioamnionitis and postnatal hyperoxia. Pediatr Res 2021; 90:565-575. [PMID: 33446917 PMCID: PMC7808307 DOI: 10.1038/s41390-020-01319-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/23/2020] [Accepted: 11/09/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Preterm infants with bronchopulmonary dysplasia (BPD) have lifelong increased risk of respiratory morbidities associated with environmental pathogen exposure and underlying mechanisms are poorly understood. The resident immune cells of the lung play vital roles in host defense. However, the effect of perinatal events associated with BPD on pulmonary-specific immune cells is not well understood. METHODS We used a double-hit model of BPD induced by prenatal chorioamnionitis followed by postnatal hyperoxia, and performed a global transcriptome analysis of all resident pulmonary immune cells. RESULTS We show significant up-regulation of genes involved in chemokine-mediated signaling and immune cell chemotaxis, and down-regulation of genes involved in multiple T lymphocyte functions. Multiple genes involved in T cell receptor signaling are downregulated and Cd8a gene expression remains downregulated at 2 months of age in spite of recovery in normoxia for 6 weeks. Furthermore, the proportion of CD8a+CD3+ pulmonary immune cells is decreased. CONCLUSIONS Our study has highlighted that perinatal lung inflammation in a double-hit model of BPD results in short- and long-term dysregulation of genes associated with the pulmonary T cell receptor signaling pathway, which may contribute to increased environmental pathogen-associated respiratory morbidities seen in children and adults with BPD. IMPACT In a translationally relevant double-hit model of BPD induced by chorioamnionitis and postnatal hyperoxia, we identified pulmonary immune cell-specific transcriptomic changes and showed that T cell receptor signaling genes are downregulated in short term and long term. This is the first comprehensive report delineating transcriptomic changes in resident immune cells of the lung in a translationally relevant double-hit model of BPD. Our study identifies novel resident pulmonary immune cell-specific targets for potential therapeutic modulation to improve short- and long-term respiratory health of preterm infants with BPD.
Collapse
|
102
|
Crosstalk Between Lung and Extrapulmonary Organs in Infection and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:333-350. [PMID: 33788201 DOI: 10.1007/978-3-030-63046-1_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute and chronic lung inflammation is a risk factor for various diseases involving lungs and extrapulmonary organs. Intercellular and interorgan networks, including crosstalk between lung and brain, intestine, heart, liver, and kidney, coordinate host immunity against infection, protect tissue, and maintain homeostasis. However, this interaction may be counterproductive and cause acute or chronic comorbidities due to dysregulated inflammation in the lung. In this chapter, we review the relationship of the lung with other key organs during normal cell processes and disease development. We focus on how pneumonia may lead to a systemic pathophysiological response to acute lung injury and chronic lung disease through organ interactions, which can facilitate the development of undesirable and even deleterious extrapulmonary sequelae.
Collapse
|
103
|
Evren E, Ringqvist E, Tripathi KP, Sleiers N, Rives IC, Alisjahbana A, Gao Y, Sarhan D, Halle T, Sorini C, Lepzien R, Marquardt N, Michaëlsson J, Smed-Sörensen A, Botling J, Karlsson MCI, Villablanca EJ, Willinger T. Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. Immunity 2020; 54:259-275.e7. [PMID: 33382972 DOI: 10.1016/j.immuni.2020.12.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/15/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.
Collapse
Affiliation(s)
- Elza Evren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Kumar Parijat Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Natalie Sleiers
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Inés Có Rives
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Arlisa Alisjahbana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Tor Halle
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden.
| |
Collapse
|
104
|
Chapman TJ, Morris MC, Xu L, Pichichero ME. Nasopharyngeal colonization with pathobionts is associated with susceptibility to respiratory illnesses in young children. PLoS One 2020; 15:e0243942. [PMID: 33306743 PMCID: PMC7732056 DOI: 10.1371/journal.pone.0243942] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Some children are more susceptible to viral and bacterial respiratory infections in the first few years of life than others. However, the factors contributing to this susceptibility are incompletely understood. In a retrospective analysis of clinical samples collected from a prospectively-enrolled cohort of 358 children we sought associations between physician-attended illness visits and bacterial colonization in the first five years of life. A subset of children was identified by unsupervised clustering analysis as infection and allergy prone (IAP). Several respiratory infection- and allergy-mediated illnesses co-occurred at higher rates in IAP children, while the rates of other illnesses were not significantly different between the groups. Analyses of nasopharyngeal (NP) pathobionts and microbiota commensals showed that early age of first colonization with pathobionts Streptococcus pneumonia, non-typeable Haemophilus influenzae, and Moraxella catarrhalis was associated with IAP children, and particularly Moraxella abundance was negatively associated with NP microbiome diversity. We conclude that mucosal pathobiont exposures in early life can influence susceptibility to respiratory illnesses in children.
Collapse
Affiliation(s)
- Timothy J. Chapman
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, United States of America
| | - Matthew C. Morris
- Center for Clinical Systems Biology, Rochester General Hospital Medical Office Building, Rochester, NY, United States of America
| | - Lei Xu
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, United States of America
| | - Michael E. Pichichero
- Center for Infectious Diseases and Immunology, Rochester General Hospital Research Institute, Rochester, NY, United States of America
| |
Collapse
|
105
|
Wong-Rolle A, Wei HK, Zhao C, Jin C. Unexpected guests in the tumor microenvironment: microbiome in cancer. Protein Cell 2020; 12:426-435. [PMID: 33296049 PMCID: PMC8106554 DOI: 10.1007/s13238-020-00813-8] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/01/2020] [Indexed: 12/19/2022] Open
Abstract
Although intestinal microbiome have been established as an important biomarker and regulator of cancer development and therapeutic response, less is known about the role of microbiome at other body sites in cancer. Emerging evidence has revealed that the local microbiota make up an important part of the tumor microenvironment across many types of cancer, especially in cancers arising from mucosal sites, including the lung, skin and gastrointestinal tract. The populations of bacteria that reside specifically within tumors have been found to be tumor-type specific, and mechanistic studies have demonstrated that tumor-associated microbiota may directly regulate cancer initiation, progression and responses to chemo- or immuno-therapies. This review aims to provide a comprehensive review of the important literature on the microbiota in the cancerous tissue, and their function and mechanism of action in cancer development and treatment.
Collapse
Affiliation(s)
- Abigail Wong-Rolle
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, 4 Memorial Drive, Bethesda, MD, 20892, USA
| | - Haohan Karen Wei
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Chen Zhao
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, 4 Memorial Drive, Bethesda, MD, 20892, USA.
| | - Chengcheng Jin
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
106
|
Wu Q, Jorde I, Kershaw O, Jeron A, Bruder D, Schreiber J, Stegemann-Koniszewski S. Resolved Influenza A Virus Infection Has Extended Effects on Lung Homeostasis and Attenuates Allergic Airway Inflammation in a Mouse Model. Microorganisms 2020; 8:microorganisms8121878. [PMID: 33260910 PMCID: PMC7761027 DOI: 10.3390/microorganisms8121878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
Allergic airway inflammation (AAI) involves T helper cell type 2 (Th2) and pro-inflammatory responses to aeroallergens and many predisposing factors remain elusive. Influenza A virus (IAV) is a major human pathogen that causes acute respiratory infections and induces specific immune responses essential for viral clearance and resolution of the infection. Beyond acute infection, IAV has been shown to persistently affect lung homeostasis and respiratory immunity. Here we asked how resolved IAV infection affects subsequently induced AAI. Mice infected with a sublethal dose of IAV were sensitized and challenged in an ovalbumin mediated mouse model for AAI after resolution of the acute viral infection. Histological changes, respiratory leukocytes, cytokines and airway hyperreactivity were analyzed in resolved IAV infection alone and in AAI with and without previous IAV infection. More than five weeks after infection, we detected persistent pneumonia with increased activated CD4+ and CD8+ lymphocytes as well as dendritic cells and MHCII expressing macrophages in the lung. Resolved IAV infection significantly affected subsequently induced AAI on different levels including morphological changes, respiratory leukocytes and lymphocytes as well as the pro-inflammatory cytokine responses, which was clearly diminished. We conclude that IAV has exceptional persisting effects on respiratory immunity with substantial consequences for subsequently induced AAI.
Collapse
Affiliation(s)
- Qingyu Wu
- Experimental Pneumology, Department of Pneumology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (Q.W.); (I.J.); (J.S.)
| | - Ilka Jorde
- Experimental Pneumology, Department of Pneumology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (Q.W.); (I.J.); (J.S.)
| | - Olivia Kershaw
- Institute of Veterinary Pathology, Department of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (A.J.); (D.B.)
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (A.J.); (D.B.)
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (Q.W.); (I.J.); (J.S.)
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (Q.W.); (I.J.); (J.S.)
- Correspondence:
| |
Collapse
|
107
|
Orešič M, McGlinchey A, Wheelock CE, Hyötyläinen T. Metabolic Signatures of the Exposome-Quantifying the Impact of Exposure to Environmental Chemicals on Human Health. Metabolites 2020; 10:metabo10110454. [PMID: 33182712 PMCID: PMC7698239 DOI: 10.3390/metabo10110454] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Human health and well-being are intricately linked to environmental quality. Environmental exposures can have lifelong consequences. In particular, exposures during the vulnerable fetal or early development period can affect structure, physiology and metabolism, causing potential adverse, often permanent, health effects at any point in life. External exposures, such as the “chemical exposome” (exposures to environmental chemicals), affect the host’s metabolism and immune system, which, in turn, mediate the risk of various diseases. Linking such exposures to adverse outcomes, via intermediate phenotypes such as the metabolome, is one of the central themes of exposome research. Much progress has been made in this line of research, including addressing some key challenges such as analytical coverage of the exposome and metabolome, as well as the integration of heterogeneous, multi-omics data. There is strong evidence that chemical exposures have a marked impact on the metabolome, associating with specific disease risks. Herein, we review recent progress in the field of exposome research as related to human health as well as selected metabolic and autoimmune diseases, with specific emphasis on the impacts of chemical exposures on the host metabolome.
Collapse
Affiliation(s)
- Matej Orešič
- School of Medical Sciences, Örebro University, SE-701 82 Örebro, Sweden; (M.O.); (A.M.)
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Aidan McGlinchey
- School of Medical Sciences, Örebro University, SE-701 82 Örebro, Sweden; (M.O.); (A.M.)
| | - Craig E. Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77 Stockholm, Sweden;
| | - Tuulia Hyötyläinen
- MTM Research Centre, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
- Correspondence:
| |
Collapse
|
108
|
Kolbe U, Yi B, Poth T, Saunders A, Boutin S, Dalpke AH. Early Cytokine Induction Upon Pseudomonas aeruginosa Infection in Murine Precision Cut Lung Slices Depends on Sensing of Bacterial Viability. Front Immunol 2020; 11:598636. [PMID: 33250899 PMCID: PMC7673395 DOI: 10.3389/fimmu.2020.598636] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/07/2020] [Indexed: 01/06/2023] Open
Abstract
Breathing allows a multitude of airborne microbes and microbial compounds to access the lung. Constant exposure of the pulmonary microenvironment to immunogenic particles illustrates the need for proper control mechanisms ensuring the differentiation between threatening and harmless encounters. Discrimination between live and dead bacteria has been suggested to be such a mechanism. In this study, we performed infection studies of murine precision cut lung slices (PCLS) with live or heat-killed P. aeruginosa, in order to investigate the role of viability for induction of an innate immune response. We demonstrate that PCLS induce a robust transcriptomic rewiring upon infection with live but not heat-killed P. aeruginosa. Using mutants of the P. aeruginosa clinical isolate CHA, we show that the viability status of P. aeruginosa is assessed in PCLS by TLR5-independent sensing of flagellin and recognition of the type three secretion system. We further demonstrate that enhanced cytokine expression towards live P. aeruginosa is mediated by uptake of viable but not heat-killed bacteria. Finally, by using a combined approach of receptor blockage and genetically modified PCLS we report a redundant involvement of MARCO and CD200R1 in the uptake of live P. aeruginosa in PCLS. Altogether, our results show that PCLS adapt the extent of cytokine expression to the viability status of P. aeruginosa by specifically internalizing live bacteria.
Collapse
Affiliation(s)
- Ulrike Kolbe
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Buqing Yi
- Institute of Medical Microbiology and Hygiene, Technische Universität Dresden, Dresden, Germany
| | - Tanja Poth
- CMCP-Center for Model System and Comparative Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amy Saunders
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Alexander H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany.,Institute of Medical Microbiology and Hygiene, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
109
|
Ubags NDJ, Alejandre Alcazar MA, Kallapur SG, Knapp S, Lanone S, Lloyd CM, Morty RE, Pattaroni C, Reynaert NL, Rottier RJ, Smits HH, de Steenhuijsen Piters WAA, Strickland DH, Collins JJP. Early origins of lung disease: towards an interdisciplinary approach. Eur Respir Rev 2020; 29:29/157/200191. [PMID: 33004528 DOI: 10.1183/16000617.0191-2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/18/2020] [Indexed: 12/27/2022] Open
Abstract
The prenatal and perinatal environments can have profound effects on the development of chronic inflammatory diseases. However, mechanistic insight into how the early-life microenvironment can impact upon development of the lung and immune system and consequent initiation and progression of respiratory diseases is still emerging. Recent studies investigating the developmental origins of lung diseases have started to delineate the effects of early-life changes in the lung, environmental exposures and immune maturation on the development of childhood and adult lung diseases. While the influencing factors have been described and studied in mostly animal models, it remains challenging to pinpoint exactly which factors and at which time point are detrimental in lung development leading to respiratory disease later in life. To advance our understanding of early origins of chronic lung disease and to allow for proper dissemination and application of this knowledge, we propose four major focus areas: 1) policy and education; 2) clinical assessment; 3) basic and translational research; and 4) infrastructure and tools, and discuss future directions for advancement. This review is a follow-up of the discussions at the European Respiratory Society Research Seminar "Early origins of lung disease: towards an interdisciplinary approach" (Lisbon, Portugal, November 2019).
Collapse
Affiliation(s)
- Niki D J Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland.,Authors are listed alphabetically except for N.D.J. Ubags and J.J.P. Collins
| | - Miguel A Alejandre Alcazar
- Dept of Paediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Paediatrics, Experimental Pulmonology, University of Cologne, Cologne, Germany.,Centre of Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Suhas G Kallapur
- Neonatal-Perinatal Medicine, Dept of Pediatrics, David Geffen School of Medicine, UCLA Mattel Children's Hospital, Los Angeles, CA, USA
| | - Sylvia Knapp
- Dept of Medicine I/Research Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria.,CeMM, Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sophie Lanone
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Clare M Lloyd
- Inflammation, Repair and Development, National Heart & Lung Institute, Imperial College London, London, UK
| | - Rory E Morty
- Dept of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Dept of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Centre, Member of the German Centre for Lung Research, Giessen, Germany
| | - Céline Pattaroni
- Dept of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Niki L Reynaert
- Dept of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Robbert J Rottier
- Dept of Paediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Hermelijn H Smits
- Dept of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Wouter A A de Steenhuijsen Piters
- Dept of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands.,National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Jennifer J P Collins
- Dept of Paediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands .,Authors are listed alphabetically except for N.D.J. Ubags and J.J.P. Collins
| |
Collapse
|
110
|
Elrashdy F, Redwan EM, Uversky VN. Why COVID-19 Transmission Is More Efficient and Aggressive Than Viral Transmission in Previous Coronavirus Epidemics? Biomolecules 2020; 10:E1312. [PMID: 32933047 PMCID: PMC7565143 DOI: 10.3390/biom10091312] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing a pandemic of coronavirus disease 2019 (COVID-19). The worldwide transmission of COVID-19 from human to human is spreading like wildfire, affecting almost every country in the world. In the past 100 years, the globe did not face a microbial pandemic similar in scale to COVID-19. Taken together, both previous outbreaks of other members of the coronavirus family (severe acute respiratory syndrome (SARS-CoV) and middle east respiratory syndrome (MERS-CoV)) did not produce even 1% of the global harm already inflicted by COVID-19. There are also four other CoVs capable of infecting humans (HCoVs), which circulate continuously in the human population, but their phenotypes are generally mild, and these HCoVs received relatively little attention. These dramatic differences between infection with HCoVs, SARS-CoV, MERS-CoV, and SARS-CoV-2 raise many questions, such as: Why is COVID-19 transmitted so quickly? Is it due to some specific features of the viral structure? Are there some specific human (host) factors? Are there some environmental factors? The aim of this review is to collect and concisely summarize the possible and logical answers to these questions.
Collapse
Affiliation(s)
- Fatma Elrashdy
- Department of Endemic Medicine and Hepatogastroenterology, Kasr Alainy School of Medicine, Cairo University, Cairo 11562, Egypt;
| | - Elrashdy M. Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Vladimir N. Uversky
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow, Russia
| |
Collapse
|
111
|
Sheikh Z, Bradbury P, Pozzoli M, Young PM, Ong HX, Traini D. An in vitro model for assessing drug transport in cystic fibrosis treatment: Characterisation of the CuFi-1 cell line. Eur J Pharm Biopharm 2020; 156:121-130. [PMID: 32916267 DOI: 10.1016/j.ejpb.2020.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/10/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023]
Abstract
Cystic fibrosis (CF) is a disease that most commonly affects the lungs and is characterized by mucus retention and a continuous cycle of bacterial infection and inflammation. Current CF treatment strategies are focused on targeted drug delivery to the lungs. Novel inhalable drug therapies require an in vitro CF model that appropriately mimics the in vivo CF lung environment to better understand drug delivery and transport across the CF epithelium, and predict drug therapeutic efficacy. Therefore, the aim of this research was to determine the appropriate air-liquid interface (ALI) culture method of the CuFi-1 (CF cell line) compared to the NuLi-1 (healthy cell line) cells to be used as in vitro models of CF airway epithelia. Furthermore, drug transport on both CuFi-1 and NuLi-1 was investigated to determine whether these cell lines could be used to study transport of drugs used in CF treatment using Ibuprofen (the only anti-inflammatory drug currently approved for CF) as a model drug. Differentiating characteristics specific to airway epithelia such as mucus production, inflammatory response and tight junction formation at two seeding densities (Low and High) were assessed throughout an 8-week ALI culture period. This study demonstrated that both the NuLi-1 and CuFi-1 cell lines fully differentiate in ALI culture with significant mucus secretion, IL-6 and IL-8 production, and functional tight junctions at week 8. Additionally, the High seeding density was found to alter the phenotype of the NuLi-1 cell line. For the first time, this study identifies that ibuprofen is transported via the paracellular pathway in ALI models of NuLi-1 and CuFi-1 cell lines. Overall, these findings highlight that NuLi-1 and CuFi-1 as promising in vitro ALI models to investigate the transport properties of novel inhalable drug therapies for CF treatment.
Collapse
Affiliation(s)
- Zara Sheikh
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Peta Bradbury
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michele Pozzoli
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia
| | - Paul M Young
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Hui Xin Ong
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Daniela Traini
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
112
|
Kumar V. Pulmonary Innate Immune Response Determines the Outcome of Inflammation During Pneumonia and Sepsis-Associated Acute Lung Injury. Front Immunol 2020; 11:1722. [PMID: 32849610 PMCID: PMC7417316 DOI: 10.3389/fimmu.2020.01722] [Citation(s) in RCA: 334] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
The lung is a primary organ for gas exchange in mammals that represents the largest epithelial surface in direct contact with the external environment. It also serves as a crucial immune organ, which harbors both innate and adaptive immune cells to induce a potent immune response. Due to its direct contact with the outer environment, the lung serves as a primary target organ for many airborne pathogens, toxicants (aerosols), and allergens causing pneumonia, acute respiratory distress syndrome (ARDS), and acute lung injury or inflammation (ALI). The current review describes the immunological mechanisms responsible for bacterial pneumonia and sepsis-induced ALI. It highlights the immunological differences for the severity of bacterial sepsis-induced ALI as compared to the pneumonia-associated ALI. The immune-based differences between the Gram-positive and Gram-negative bacteria-induced pneumonia show different mechanisms to induce ALI. The role of pulmonary epithelial cells (PECs), alveolar macrophages (AMs), innate lymphoid cells (ILCs), and different pattern-recognition receptors (PRRs, including Toll-like receptors (TLRs) and inflammasome proteins) in neutrophil infiltration and ALI induction have been described during pneumonia and sepsis-induced ALI. Also, the resolution of inflammation is frequently observed during ALI associated with pneumonia, whereas sepsis-associated ALI lacks it. Hence, the review mainly describes the different immune mechanisms responsible for pneumonia and sepsis-induced ALI. The differences in immune response depending on the causal pathogen (Gram-positive or Gram-negative bacteria) associated pneumonia or sepsis-induced ALI should be taken in mind specific immune-based therapeutics.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, Faculty of Medicine, School of Clinical Medicine, Mater Research, University of Queensland, Brisbane, QLD, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
113
|
Seinen J, Dieperink W, Mekonnen SA, Lisotto P, Harmsen HJM, Hiemstra B, Ott A, Schultz D, Lalk M, Oswald S, Hammerschmidt S, de Smet AMGA, van Dijl JM. Heterogeneous antimicrobial activity in broncho-alveolar aspirates from mechanically ventilated intensive care unit patients. Virulence 2020; 10:879-891. [PMID: 31662033 PMCID: PMC6844299 DOI: 10.1080/21505594.2019.1682797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Pneumonia is an infection of the lungs, where the alveoli in the affected area are filled with pus and fluid. Although ventilated patients are at risk, not all ventilated patients develop pneumonia. This suggests that the sputum environment may possess antimicrobial activities. Despite the generally acknowledged importance of antimicrobial activity in protecting the human lung against infections, this has not been systematically assessed to date. Therefore, the objective of the present study was to measure antimicrobial activity in broncho-alveolar aspirate (‘sputum”) samples from patients in an intensive care unit (ICU) and to correlate the detected antimicrobial activity with antibiotic levels, the sputum microbiome, and the respective patients’ characteristics. To this end, clinical metadata and sputum were collected from 53 mechanically ventilated ICU patients. The antimicrobial activity of sputum samples was tested against Streptococcus pneumoniae, Staphylococcus aureus and Streptococcus anginosus. Here we show that sputa collected from different patients presented a high degree of variation in antimicrobial activity, which can be partially attributed to antibiotic therapy. The sputum microbiome, although potentially capable of producing antimicrobial agents, seemed to contribute in a minor way, if any, to the antimicrobial activity of sputum. Remarkably, despite its potentially protective effect, the level of antimicrobial activity in the investigated sputa correlated inversely with patient outcome, most likely because disease severity outweighed the beneficial antimicrobial activities.
Collapse
Affiliation(s)
- Jolien Seinen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Willem Dieperink
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Solomon A Mekonnen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University Medicine of Greifswald, Greifswald, Germany
| | - Paola Lisotto
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart Hiemstra
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alewijn Ott
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Medical Microbiology, Certe, Groningen, The Netherlands
| | - Daniel Schultz
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Michael Lalk
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Stefan Oswald
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Anne Marie G A de Smet
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
114
|
Valacchi G, Magnani N, Woodby B, Ferreira SM, Evelson P. Particulate Matter Induces Tissue OxInflammation: From Mechanism to Damage. Antioxid Redox Signal 2020; 33:308-326. [PMID: 32443938 DOI: 10.1089/ars.2019.8015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Oxidative stress and oxidative damage are central hypothetical mechanisms for the adverse effects of airborne particulate matter (PM). Activation of inflammatory cells capable of generating reactive oxygen and nitrogen species is another proposed damage pathway. Understanding the interplay between these responses can help us understand the adverse health effects attributed to breathing polluted air. Recent Advances: The consequences of PM exposure on different organs are oxidative damage, decreased function, and inflammation, which can lead to the development/exacerbation of proinflammatory disorders. Mitochondrial damage is also an important event in PM-induced cytotoxicity. Critical Issues: Reactive oxygen species (ROS) are generated during phagocytosis of the particles, leading to enhancement of oxidative stress and triggering the inflammatory response. The activation of inflammatory signaling pathways results in the release of cytokines and other mediators, which can further induce ROS production by activating endogenous enzymes, leading to a positive feedback loop, which can aggravate the effects triggered by PM exposure. Future Directions: Further research is required to elucidate the exact mechanisms by which PM exposure results in adverse health effects, in terms of the relationship between the redox responses triggered by the presence of the particles and the inflammation observed in the different organs, so the development/exacerbation of PM-associated health problems can be prevented.
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, North Carolina, USA.,Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy.,Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Brittany Woodby
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, North Carolina, USA
| | - Sandra María Ferreira
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
115
|
Paramasivan S, Bassiouni A, Shiffer A, Dillon MR, Cope EK, Cooksley C, Ramezanpour M, Moraitis S, Ali MJ, Bleier B, Callejas C, Cornet ME, Douglas RG, Dutra D, Georgalas C, Harvey RJ, Hwang PH, Luong AU, Schlosser RJ, Tantilipikorn P, Tewfik MA, Vreugde S, Wormald P, Caporaso JG, Psaltis AJ. The international sinonasal microbiome study: A multicentre, multinational characterization of sinonasal bacterial ecology. Allergy 2020; 75:2037-2049. [PMID: 32167574 DOI: 10.1111/all.14276] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/20/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
The sinonasal microbiome remains poorly defined, with our current knowledge based on a few cohort studies whose findings are inconsistent. Furthermore, the variability of the sinus microbiome across geographical divides remains unexplored. We characterize the sinonasal microbiome and its geographical variations in both health and disease using 16S rRNA gene sequencing of 410 individuals from across the world. Although the sinus microbial ecology is highly variable between individuals, we identify a core microbiome comprised of Corynebacterium, Staphylococcus, Streptococcus, Haemophilus and Moraxella species in both healthy and chronic rhinosinusitis (CRS) cohorts. Corynebacterium (mean relative abundance = 44.02%) and Staphylococcus (mean relative abundance = 27.34%) appear particularly dominant in the majority of patients sampled. Amongst patients suffering from CRS with nasal polyps, a statistically significant reduction in relative abundance of Corynebacterium (40.29% vs 50.43%; P = .02) was identified. Despite some measured differences in microbiome composition and diversity between some of the participating centres in our cohort, these differences would not alter the general pattern of core organisms described. Nevertheless, atypical or unusual organisms reported in short-read amplicon sequencing studies and that are not part of the core microbiome should be interpreted with caution. The delineation of the sinonasal microbiome and standardized methodology described within our study will enable further characterization and translational application of the sinus microbiota.
Collapse
Affiliation(s)
- Sathish Paramasivan
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Ahmed Bassiouni
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Arron Shiffer
- Pathogen and Microbiome Institute Northern Arizona University Flagstaff AZ USA
| | - Matthew R. Dillon
- Pathogen and Microbiome Institute Northern Arizona University Flagstaff AZ USA
| | - Emily K. Cope
- Pathogen and Microbiome Institute Northern Arizona University Flagstaff AZ USA
| | - Clare Cooksley
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Mahnaz Ramezanpour
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Sophia Moraitis
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | | | - Benjamin Bleier
- Department of Otolaryngology Massachusetts Eye and Ear Infirmary Harvard Medical School Boston MA USA
| | - Claudio Callejas
- Department of Otolaryngology Pontificia Universidad Catolica de Chile Santiago Chile
| | | | | | - Daniel Dutra
- Department of Otorhinolaryngology University of Sao Paulo Sao Paulo Brazil
| | - Christos Georgalas
- Department of Otorhinolaryngology Amsterdam UMC Amsterdam The Netherlands
| | - Richard J. Harvey
- Department of Otolaryngology, Rhinology and Skull base University of New South Wales Sydney NSW Australia
- Faculty of Medicine and Health sciences Macquarie University Sydney NSW Australia
| | - Peter H. Hwang
- Department of Otolaryngology ‐Head and Neck Surgery Stanford University Stanford CA USA
| | - Amber U. Luong
- Department of Otolaryngology ‐Head and Neck Surgery University of Texas Austin TX USA
| | - Rodney J. Schlosser
- Department of Otolaryngology Medical University of South Carolina Charleston SC USA
| | - Pongsakorn Tantilipikorn
- Department of Otorhinolaryngology Faculty of Medicine Siriraj Hospital Mahidol University Bangkok Thailand
| | - Marc A. Tewfik
- Department of Otolaryngology ‐ Head and Neck Surgery McGill University Montreal QC Canada
| | - Sarah Vreugde
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - Peter‐John Wormald
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| | - J. Gregory Caporaso
- Pathogen and Microbiome Institute Northern Arizona University Flagstaff AZ USA
| | - Alkis J. Psaltis
- Department of Otolaryngology, Head and Neck Surgery University of Adelaide Adelaide SA Australia
| |
Collapse
|
116
|
Khatiwada S, Subedi A. Lung microbiome and coronavirus disease 2019 (COVID-19): Possible link and implications. HUMAN MICROBIOME JOURNAL 2020; 17:100073. [PMID: 32835135 PMCID: PMC7405772 DOI: 10.1016/j.humic.2020.100073] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly emerging disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The disease begins as an infection of lungs, which is self-limiting in the majority of infections; however, some develop severe respiratory distress and organ failures. Lung microbiome, though neglected previously have received interest recently because of its association with several respiratory diseases and immunity. Lung microbiome can modify the risk and consequences of COVID-19 disease by activating an innate and adaptive immune response. In this review, we examine the current evidence on COVID-19 disease and lung microbiome, and how lung microbiome can affect SARS-CoV-2 infection and the outcomes of this disease. To date there is no direct evidence from human or animal studies on the role of lung microbiome in modifying COVID-19 disease; however, related studies support that microbiome can play an essential role in developing immunity against viral infections. Future studies need to be undertaken to find the relationship between lung microbiome and COVID-19 disease.
Collapse
Key Words
- ACE2, Angiotensin converting enzyme
- ARDS, Acute respiratory distress syndrome
- ARF, Acute respiratory failure
- COPD, Chronic obstructive pulmonary disease
- COVID-19, Coronavirus disease 2019
- CRP, C reactive protein
- Coronavirus disease (COVID-19)
- IFN, Interferon
- Immunity
- Infection
- Lung microbiome
- PAMP, Pathogen associated molecular patterns
- PRR, Pattern recognition receptor
- RSV, Respiratory syncytial virus
- SARS-CoV-2, Severe acute respiratory syndrome coronavirus 2
- Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
- TLR, Toll like receptor
- TNF, Tumour necrosis factor
- rRNA, Ribosomal RNA
Collapse
Affiliation(s)
- Saroj Khatiwada
- School of Medical Sciences, UNSW Sydney, Australia,Department of Biochemistry, Modern Technical College, Nepal,Corresponding author at: School of Medical Sciences, UNSW Sydney, Australia
| | - Astha Subedi
- Medicine ICU, Tribhuvan University Teaching Hospital, Nepal
| |
Collapse
|
117
|
Lira-Lucio JA, Falfán-Valencia R, Ramírez-Venegas A, Buendía-Roldán I, Rojas-Serrano J, Mejía M, Pérez-Rubio G. Lung Microbiome Participation in Local Immune Response Regulation in Respiratory Diseases. Microorganisms 2020; 8:E1059. [PMID: 32708647 PMCID: PMC7409050 DOI: 10.3390/microorganisms8071059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
The lung microbiome composition has critical implications in the regulation of innate and adaptive immune responses. Next-generation sequencing techniques have revolutionized the understanding of pulmonary physiology and pathology. Currently, it is clear that the lung is not a sterile place; therefore, the investigation of the participation of the pulmonary microbiome in the presentation, severity, and prognosis of multiple pathologies, such as asthma, chronic obstructive pulmonary disease, and interstitial lung diseases, contributes to a better understanding of the pathophysiology. Dysregulation of microbiota components in the microbiome-host interaction is associated with multiple lung pathologies, severity, and prognosis, making microbiome study a useful tool for the identification of potential therapeutic strategies. This review integrates the findings regarding the activation and regulation of the innate and adaptive immune response pathways according to the microbiome, including microbial patterns that could be characteristic of certain diseases. Further studies are required to verify whether the microbial profile and its metabolites can be used as biomarkers of disease progression or poor prognosis and to identify new therapeutic targets that restore lung dysbiosis safely and effectively.
Collapse
Affiliation(s)
- Juan Alberto Lira-Lucio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| | - Alejandra Ramírez-Venegas
- Tobacco Smoking and COPD Research Department, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Jorge Rojas-Serrano
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-S.); (M.M.)
| | - Mayra Mejía
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-S.); (M.M.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| |
Collapse
|
118
|
Wirsching E, Fauler M, Fois G, Frick M. P2 Purinergic Signaling in the Distal Lung in Health and Disease. Int J Mol Sci 2020; 21:E4973. [PMID: 32674494 PMCID: PMC7404078 DOI: 10.3390/ijms21144973] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
The distal lung provides an intricate structure for gas exchange in mammalian lungs. Efficient gas exchange depends on the functional integrity of lung alveoli. The cells in the alveolar tissue serve various functions to maintain alveolar structure, integrity and homeostasis. Alveolar epithelial cells secrete pulmonary surfactant, regulate the alveolar surface liquid (ASL) volume and, together with resident and infiltrating immune cells, provide a powerful host-defense system against a multitude of particles, microbes and toxicants. It is well established that all of these cells express purinergic P2 receptors and that purinergic signaling plays important roles in maintaining alveolar homeostasis. Therefore, it is not surprising that purinergic signaling also contributes to development and progression of severe pathological conditions like pulmonary inflammation, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pulmonary fibrosis. Within this review we focus on the role of P2 purinergic signaling in the distal lung in health and disease. We recapitulate the expression of P2 receptors within the cells in the alveoli, the possible sources of ATP (adenosine triphosphate) within alveoli and the contribution of purinergic signaling to regulation of surfactant secretion, ASL volume and composition, as well as immune homeostasis. Finally, we summarize current knowledge of the role for P2 signaling in infectious pneumonia, ALI/ARDS and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
| | | | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (E.W.); (M.F.); (G.F.)
| |
Collapse
|
119
|
Jang YO, Lee SH, Choi JJ, Kim DH, Choi JM, Kang MJ, Oh YM, Park YJ, Shin Y, Lee SW. Fecal microbial transplantation and a high fiber diet attenuates emphysema development by suppressing inflammation and apoptosis. Exp Mol Med 2020; 52:1128-1139. [PMID: 32681029 PMCID: PMC8080776 DOI: 10.1038/s12276-020-0469-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/24/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Recent work has suggested a microbial dysbiosis association between the lung and gut in respiratory diseases. Here, we demonstrated that gut microbiome modulation attenuated emphysema development. To modulate the gut microbiome, fecal microbiota transplantation (FMT) and diet modification were adopted in mice exposed to smoking and poly I:C for the emphysema model. We analyzed the severity of emphysema by the mean linear intercept (MLI) and apoptosis by the fluorescent TUNEL assay. Microbiome analysis was also performed in feces and fecal extracellular vesicles (EVs). The MLI was significantly increased with smoking exposure. FMT or a high-fiber diet (HFD) attenuated the increase. Weight loss, combined with smoking exposure, was not noted in mice with FMT. HFD significantly decreased macrophages and lymphocytes in bronchoalveolar lavage fluid. Furthermore, IL-6 and IFN-γ were decreased in the bronchoalveolar lavage fluid and serum. The TUNEL score was significantly lower in mice with FMT or HFD, suggesting decreased cell apoptosis. In the microbiome analysis, Bacteroidaceae and Lachnospiraceae, which are alleged to metabolize fiber into short-chain fatty acids (SCFAs), increased at the family level with FMT and HFD. FMT and HFD attenuated emphysema development via local and systemic inhibition of inflammation and changes in gut microbiota composition, which could provide a new paradigm in COPD treatment.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Se Hee Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Department of Pulmonology, Allergy and Critical Care Medicine, CHA Bundang Medical Center, CHA University, Seongnam-si, 13496, Republic of Korea
| | - Jong Jin Choi
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Do-Hyun Kim
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, 06520-8057, Connecticut, USA
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Young-Jun Park
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Yong Shin
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
120
|
Monaghan KL, Farris BY, Zheng W, Wan ECK. Characterization of Immune Cells and Proinflammatory Mediators in the Pulmonary Environment. J Vis Exp 2020. [PMID: 32658184 DOI: 10.3791/61359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immune cell expansion, activation, and trafficking to the lungs, which are controlled by the expression of multiple cytokines and chemokines, may be altered by severe brain injury. This is evidenced by the fact that pneumonia is a major cause of mortality in patients who have suffered from ischemic stroke. The goal of this protocol is to describe the use of multicolor flow cytometric analysis to identify 13 types of immune cells in the lungs of mice, including alveolar macrophages, interstitial macrophages, CD103+ or CD11b+ dendritic cells (DCs), plasmacytoid DCs, eosinophils, monocytes/monocyte-derived cells, neutrophils, lymphoid-derived T and B cells, NK cells, and NKT cells, following ischemic stroke induction by transient middle cerebral artery occlusion. Moreover, we describe the preparation of lung homogenates using a bead homogenization method, to determine the expression levels of 13 different cytokines or chemokines simultaneously by multiplex bead arrays coupled with flow cytometric analysis. This protocol can also be used to investigate the pulmonary immune response in other disease settings, such as infectious lung disease or allergic disease.
Collapse
Affiliation(s)
- Kelly L Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Breanne Y Farris
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Wen Zheng
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Edwin C K Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University; Department of Neuroscience, West Virginia University; Rockefeller Neuroscience Institute, West Virginia University;
| |
Collapse
|
121
|
Yen BL, Yen ML, Wang LT, Liu KJ, Sytwu HK. Current status of mesenchymal stem cell therapy for immune/inflammatory lung disorders: Gleaning insights for possible use in COVID-19. Stem Cells Transl Med 2020; 9:1163-1173. [PMID: 32526079 PMCID: PMC7300965 DOI: 10.1002/sctm.20-0186] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 01/08/2023] Open
Abstract
The broad immunomodulatory properties of human mesenchymal stem cells (MSCs) have allowed for wide application in regenerative medicine as well as immune/inflammatory diseases, including unmatched allogeneic use. The novel coronavirus disease COVID‐19 has unleashed a pandemic in record time accompanied by an alarming mortality rate mainly due to pulmonary injury and acute respiratory distress syndrome. Because there are no effective preventive or curative therapies currently, MSC therapy (MSCT) has emerged as a possible candidate despite the lack of preclinical data of MSCs for COVID‐19. Interestingly, MSCT preclinical data specifically on immune/inflammatory disorders of the lungs were among the earliest to be reported in 2003, with the first clinical use of MSCT for graft‐vs‐host disease reported in 2004. Since these first reports, preclinical data showing beneficial effects of MSC immunomodulation have accumulated substantially, and as a consequence, over a third of MSCT clinical trials now target immune/inflammatory diseases. There is much preclinical evidence for MSCT in noninfectious—including chronic obstructive pulmonary disease, asthma, and idiopathic pulmonary fibrosis—as well as infectious bacterial immune/inflammatory lung disorders, with data generally demonstrating therapeutic effects; however, for infectious viral pulmonary conditions, the preclinical evidence is more scarce with some inconsistent outcomes. In this article, we review the mechanistic evidence for clinical use of MSCs in pulmonary immune/inflammatory disorders, and survey the ongoing clinical trials—including for COVID‐19—of MSCT for these diseases, with some perspectives and comment on MSCT for COVID‐19.
Collapse
Affiliation(s)
- B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan
| | - Li-Tzu Wang
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Tainan, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases & Vaccinology, NHRI, Zhunan, Taiwan.,Department & Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
122
|
Domingo-Gonzalez R, Zanini F, Che X, Liu M, Jones RC, Swift MA, Quake SR, Cornfield DN, Alvira CM. Diverse homeostatic and immunomodulatory roles of immune cells in the developing mouse lung at single cell resolution. eLife 2020; 9:e56890. [PMID: 32484158 PMCID: PMC7358008 DOI: 10.7554/elife.56890] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.
Collapse
Affiliation(s)
- Racquel Domingo-Gonzalez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Fabio Zanini
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South WalesSydneyAustralia
| | - Xibing Che
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Michael A Swift
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - David N Cornfield
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
123
|
Early dynamics of innate immunity during pulmonary tuberculosis. Immunol Lett 2020; 221:56-60. [DOI: 10.1016/j.imlet.2020.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 01/22/2023]
|
124
|
Kelly AM, McLoughlin RM. Target the Host, Kill the Bug; Targeting Host Respiratory Immunosuppressive Responses as a Novel Strategy to Improve Bacterial Clearance During Lung Infection. Front Immunol 2020; 11:767. [PMID: 32425944 PMCID: PMC7203494 DOI: 10.3389/fimmu.2020.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
The lung is under constant pressure to protect the body from invading bacteria. An effective inflammatory immune response must be tightly orchestrated to ensure complete clearance of any invading bacteria, while simultaneously ensuring that inflammation is kept under strict control to preserve lung viability. Chronic bacterial lung infections are seen as a major threat to human life with the treatment of these infections becoming more arduous as the prevalence of antibiotic resistance becomes increasingly commonplace. In order to survive within the lung bacteria target the host immune system to prevent eradication. Many bacteria directly target inflammatory cells and cytokines to impair inflammatory responses. However, bacteria also have the capacity to take advantage of and strongly promote anti-inflammatory immune responses in the host lung to inhibit local pro-inflammatory responses that are critical to bacterial elimination. Host cells such as T regulatory cells and myeloid-derived suppressor cells are often enhanced in number and activity during chronic pulmonary infection. By increasing suppressive cell populations and cytokines, bacteria promote a permissive environment suitable for their prolonged survival. This review will explore the anti-inflammatory aspects of the lung immune system that are targeted by bacteria and how bacterial-induced immunosuppression could be inhibited through the use of host-directed therapies to improve treatment options for chronic lung infections.
Collapse
Affiliation(s)
- Alanna M Kelly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
125
|
Helfrich S, Duerr CU. Regulating the development of pulmonary Group 2 innate lymphoid cells. Biol Chem 2020; 400:1497-1507. [PMID: 31256061 DOI: 10.1515/hsz-2019-0175] [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: 02/28/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) are members of the family of innate lymphoid cells and are innately committed to type 2 immune responses. In the lungs, ILC2s are the predominant population of innate lymphoid cells (ILCs) and their development is orchestrated by several different transcription factors ensuring lineage commitment by intrinsic regulation. ILC2s are present in the lungs from the foetal period onwards and are thus exposed to extrinsic regulation due to the airways' continuous morphological changes upon birth. In this review, we will briefly summarise the dependence of ILC2s on transcription factors and discuss recently described characteristics and function of early life ILC2s in the lungs.
Collapse
Affiliation(s)
- Sofia Helfrich
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, D-12203 Berlin, Germany
| | - Claudia U Duerr
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, D-12203 Berlin, Germany
| |
Collapse
|
126
|
Precision medicine in the clinical management of respiratory tract infections including multidrug-resistant tuberculosis: learning from innovations in immuno-oncology. Curr Opin Pulm Med 2020; 25:233-241. [PMID: 30883448 DOI: 10.1097/mcp.0000000000000575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In the light of poor management outcomes of antibiotic-resistant respiratory tract infection (RTI)-associated sepsis syndrome and multidrug-resistant tuberculosis (MDR-TB), new management interventions based on host-directed therapies (HDTs) are warranted to improve morbidity, mortality and long-term functional outcomes. We review developments in potential HDTs based on precision cancer therapy concepts applicable to RTIs including MDR-TB. RECENT FINDINGS Immune reactivity, tissue destruction and repair processes identified during studies of cancer immunotherapy share common pathogenetic mechanisms with RTI-associated sepsis syndrome and MDR-TB. T-cell receptors (TCRs) and chimeric antigen receptors targeting pathogen-specific or host-derived mutated molecules (major histocompatibility class-dependent/ major histocompatibility class-independent) can be engineered for recognition by TCR γδ and natural killer (NK) cells. T-cell subsets and, more recently, NK cells are shown to be host-protective. These cells can also be activated by immune checkpoint inhibitor (ICI) or derived from allogeneic sources and serve as potential for improving clinical outcomes in RTIs and MDR-TB. SUMMARY Recent developments of immunotherapy in cancer reveal common pathways in immune reactivity, tissue destruction and repair. RTIs-related sepsis syndrome exhibits mixed immune reactions, making cytokine or ICI therapy guided by robust biomarker analyses, viable treatment options.
Collapse
|
127
|
Bolte C, Kalin TV, Kalinichenko VV. Molecular, cellular, and bioengineering approaches to stimulate lung regeneration after injury. Semin Cell Dev Biol 2020; 100:101-108. [PMID: 31669132 DOI: 10.1016/j.semcdb.2019.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 01/03/2023]
Abstract
The lung is susceptible to damage from a variety of sources throughout development and in adulthood. As a result, the lung has great capacities for repair and regeneration, directed by precisely controlled sequences of molecular and signaling pathways. Impairments or alterations in these signaling events can have deleterious effects on lung structure and function, ultimately leading to chronic lung disorders. When lung injury is too severe for the normal pathways to repair, or if those pathways do not function properly, lung regenerative medicine is needed to restore adequate structure and function. Great progress has been made in recent years in the number of regenerative techniques and their efficacy. This review will address recent progress in lung regenerative medicine focusing on pharmacotherapy including the expanding role of nanotechnology, stem cell-based therapies, and bioengineering techniques. The use of these techniques individually and collectively has the potential to significantly improve morbidity and mortality associated with congenital and acquired lung disorders.
Collapse
Affiliation(s)
- Craig Bolte
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, United States.
| | - Tanya V Kalin
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, United States
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, United States.
| |
Collapse
|
128
|
Interaction of Long Noncoding RNAs and Notch Signaling: Implications for Tissue Homeostasis Loss. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:107-129. [PMID: 32072502 DOI: 10.1007/978-3-030-36422-9_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Notch signaling is a crucial pathway involved in cellular development, progression, and differentiation. Deregulation of Notch signaling pathway commonly impacts tissue homeostasis, being highly associated with proliferative disorders. The long noncoding RNAs (lncRNAs), which are transcripts with more than 200 nucleotides that do not code for proteins, were already described as Notch signaling pathway-interacting molecules. Many of them act as important transcriptional and posttranscriptional regulators, affecting gene expression and targeting other regulatory molecules, such as miRNAs. Due to their strong impact on function and gene expression of Notch-related molecules, lncRNAs influence susceptibility to cancer and other diseases, and can be regarded as potential biomarkers and therapeutic targets. Along this chapter, we summarize the cross talk between the Notch signaling pathway and their most important modulating lncRNAs, as well as the pathological consequences of these interactions, in different tissues.
Collapse
|
129
|
Matt SM, Gaskill PJ. Where Is Dopamine and how do Immune Cells See it?: Dopamine-Mediated Immune Cell Function in Health and Disease. J Neuroimmune Pharmacol 2020; 15:114-164. [PMID: 31077015 PMCID: PMC6842680 DOI: 10.1007/s11481-019-09851-4] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023]
Abstract
Dopamine is well recognized as a neurotransmitter in the brain, and regulates critical functions in a variety of peripheral systems. Growing research has also shown that dopamine acts as an important regulator of immune function. Many immune cells express dopamine receptors and other dopamine related proteins, enabling them to actively respond to dopamine and suggesting that dopaminergic immunoregulation is an important part of proper immune function. A detailed understanding of the physiological concentrations of dopamine in specific regions of the human body, particularly in peripheral systems, is critical to the development of hypotheses and experiments examining the effects of physiologically relevant dopamine concentrations on immune cells. Unfortunately, the dopamine concentrations to which these immune cells would be exposed in different anatomical regions are not clear. To address this issue, this comprehensive review details the current information regarding concentrations of dopamine found in both the central nervous system and in many regions of the periphery. In addition, we discuss the immune cells present in each region, and how these could interact with dopamine in each compartment described. Finally, the review briefly addresses how changes in these dopamine concentrations could influence immune cell dysfunction in several disease states including Parkinson's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, as well as the collection of pathologies, cognitive and motor symptoms associated with HIV infection in the central nervous system, known as NeuroHIV. These data will improve our understanding of the interactions between the dopaminergic and immune systems during both homeostatic function and in disease, clarify the effects of existing dopaminergic drugs and promote the creation of new therapeutic strategies based on manipulating immune function through dopaminergic signaling. Graphical Abstract.
Collapse
Affiliation(s)
- S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
130
|
Deinhardt-Emmer S, Rennert K, Schicke E, Cseresnyés Z, Windolph M, Nietzsche S, Heller R, Siwczak F, Haupt KF, Carlstedt S, Schacke M, Figge MT, Ehrhardt C, Löffler B, Mosig AS. Co-infection with Staphylococcus aureus after primary influenza virus infection leads to damage of the endothelium in a human alveolus-on-a-chip model. Biofabrication 2020; 12:025012. [PMID: 31994489 DOI: 10.1088/1758-5090/ab7073] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pneumonia is one of the most common infectious diseases worldwide. The influenza virus can cause severe epidemics, which results in significant morbidity and mortality. Beyond the virulence of the virus itself, epidemiological data suggest that bacterial co-infections are the major cause of increased mortality. In this context, Staphylococcus aureus represents a frequent causative bacterial pathogen. Currently available models have several limitations in the analysis of the pathogenesis of infections, e.g. some bacterial toxins strongly act in a species-specific manner. Human 2D mono-cell culture models often fail to maintain the differentiation of alveolus-specific functions. A detailed investigation of the underlying pathogenesis mechanisms requires a physiological interaction of alveolus-specific cell types. The aim of the present work was to establish a human in vitro alveolus model system composed of vascular and epithelial cell structures with cocultured macrophages resembling the human alveolus architecture and functions. We demonstrate that high barrier integrity maintained for up to 14 d in our model containing functional tissue-resident macrophages. We show that flow conditions and the presence of macrophages increased the barrier function. The infection of epithelial cells induced a high inflammatory response that spread to the endothelium. Although the integrity of the epithelium was not compromised by a single infection or co-infection, we demonstrated significant endothelial cell damage associated with loss of barrier function. We established a novel immune-responsive model that reflects the complex crosstalk between pathogens and host. The in vitro model allows for the monitoring of spatiotemporal spreading of the pathogens and the characterization of morphological and functional alterations attributed to infection. The alveolus-on-a-chip represents a promising platform for mechanistic studies of host-pathogen interactions and the identification of molecular and cellular targets of novel treatment strategies in pneumonia.
Collapse
Affiliation(s)
- Stefanie Deinhardt-Emmer
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, D-07747 Jena, Germany. Center for Sepsis Control and Care, Jena University Hospital, D-07747 Jena, Germany. Section of Experimental Virology, Institute of Medical Microbiology, Jena University Hospital, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Oherle K, Acker E, Bonfield M, Wang T, Gray J, Lang I, Bridges J, Lewkowich I, Xu Y, Ahlfeld S, Zacharias W, Alenghat T, Deshmukh H. Insulin-like Growth Factor 1 Supports a Pulmonary Niche that Promotes Type 3 Innate Lymphoid Cell Development in Newborn Lungs. Immunity 2020; 52:275-294.e9. [PMID: 32075728 PMCID: PMC7382307 DOI: 10.1016/j.immuni.2020.01.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 05/16/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
Type 3 innate lymphoid cells (ILC3s) are critical for lung defense against bacterial pneumonia in the neonatal period, but the signals that guide pulmonary ILC3 development remain unclear. Here, we demonstrated that pulmonary ILC3s descended from ILC precursors that populated a niche defined by fibroblasts in the developing lung. Alveolar fibroblasts produced insulin-like growth factor 1 (IGF1), which instructed expansion and maturation of pulmonary ILC precursors. Conditional ablation of IGF1 in alveolar fibroblasts or deletion of the IGF-1 receptor from ILC precursors interrupted ILC3 biogenesis and rendered newborn mice susceptible to pneumonia. Premature infants with bronchopulmonary dysplasia, characterized by interrupted postnatal alveolar development and increased morbidity to respiratory infections, had reduced IGF1 concentrations and pulmonary ILC3 numbers. These findings indicate that the newborn period is a critical window in pulmonary immunity development, and disrupted lung development in prematurely born infants may have enduring effects on host resistance to respiratory infections.
Collapse
Affiliation(s)
- Katherine Oherle
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA
| | - Elizabeth Acker
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA
| | - Madeline Bonfield
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Timothy Wang
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jerilyn Gray
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA
| | - Ian Lang
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA
| | - James Bridges
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Ian Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yan Xu
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shawn Ahlfeld
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - William Zacharias
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Hitesh Deshmukh
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
132
|
Nelson BN, Hawkins AN, Wozniak KL. Pulmonary Macrophage and Dendritic Cell Responses to Cryptococcus neoformans. Front Cell Infect Microbiol 2020; 10:37. [PMID: 32117810 PMCID: PMC7026008 DOI: 10.3389/fcimb.2020.00037] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
The fungal pathogen Cryptococcus neoformans can cause life-threatening infections in immune compromised individuals. This pathogen is typically acquired via inhalation, and enters the respiratory tract. Innate immune cells such as macrophages and dendritic cells (DCs) are the first host cells that encounter C. neoformans, and the interactions between Cryptococcus and innate immune cells play a critical role in the progression of disease. Cryptococcus possesses several virulence factors and evasion strategies to prevent its killing and destruction by pulmonary phagocytes, but these phagocytic cells can also contribute to anti-cryptococcal responses. This review will focus on the interactions between Cryptococcus and primary macrophages and dendritic cells (DCs), dealing specifically with the cryptococcal/pulmonary cell interface.
Collapse
Affiliation(s)
- Benjamin N Nelson
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Ashlee N Hawkins
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Karen L Wozniak
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
133
|
Mariani J, Favero C, Carugno M, Pergoli L, Ferrari L, Bonzini M, Cattaneo A, Pesatori AC, Bollati V. Nasal Microbiota Modifies the Effects of Particulate Air Pollution on Plasma Extracellular Vesicles. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17020611. [PMID: 31963616 PMCID: PMC7013854 DOI: 10.3390/ijerph17020611] [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] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022]
Abstract
Air pollution exposure has been linked to modifications of both extracellular vesicle (EV) concentration and nasal microbiota structure (NMB), which might act as the respiratory health gatekeeper. This study aimed to assess whether an unbalanced NMB could modify the effect of particulate matter (PM) exposure on plasmatic EV levels. Due to two different NMB taxonomical profiles characterized by a widely different relative abundance of the Moraxella genus, the enrolled population was stratified into Mor- (balanced NMB) and Mor+ (unbalanced NMB) groups (Moraxella genus's cut-off ≤25% and >25%, respectively). EV features were assessed by nanoparticle tracking analysis (NTA) and flow-cytometry (FC). Multivariable analyses were applied on EV outcomes to evaluate a possible association between PM10 and PM2.5 and plasmatic EV levels. The Mor- group revealed positive associations between PM levels and plasmatic CD105+ EVs (GMR = 4.39 p = 0.02) as for total EV count (GMR = 1.92 p = 0.02). Conversely, the Mor+ group showed a negative association between exposure and EV outcomes (CD66+ GMR = 0.004 p = 0.01; EpCAM+ GMR = 0.005 p = 0.01). Our findings provide an insight regarding how a balanced NMB may help to counteract PM exposure effects in terms of plasmatic EV concentration. Further research is necessary to understand the relationship between the host and the NMB to disentangle the mechanism exerted by inhaled pollutants in modulating EVs and NMB.
Collapse
Affiliation(s)
- Jacopo Mariani
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
| | - Chiara Favero
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
| | - Michele Carugno
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
| | - Laura Pergoli
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
| | - Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
| | - Matteo Bonzini
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Cattaneo
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy;
| | - Angela Cecilia Pesatori
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (J.M.); (C.F.); (M.C.); (L.P.); (L.F.); (M.B.); (A.C.P.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-503-20147
| |
Collapse
|
134
|
Park EJ, Han JS, Seong E, Park EJ, Lee BS, Lee SJ, Lee K. Inhaled Kathon may induce eosinophilia-mediated disease in the lung. ENVIRONMENTAL TOXICOLOGY 2020; 35:27-36. [PMID: 31498972 DOI: 10.1002/tox.22839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
In 2011, a link between humidifier disinfectants and patients with idiopathic pulmonary fibrosis was identified in Korea, and Kathon was suggested as one of the causative agents. In this study, Kathon induced apoptotic cell death along with membrane damage at 24 h post-exposure. Additionally, on day 14 after a single instillation with Kathon, the total number of pulmonary cells and the levels of TNF-α, IL-5, IL-13, MIP-1α, and MCP-1α clearly increased in the lung of mice. The proportion of natural killer cells and eosinophils were significantly elevated in the spleen and the bloodstream, respectively, and the level of immunoglobulin (Ig) A, but not IgG, IgM, and IgE, dose-dependently increased. Therefore, we suggest that inhaled Kathon may induce eosinophilia-mediated disease in the lung by disrupting homeostasis of pulmonary surfactants. Considering that eosinophilia is closely related to cancer and fibrosis, further studies are needed to understand the relationship between them.
Collapse
Affiliation(s)
- Eun-Jung Park
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Ji-Seok Han
- Toxicologic Pathology Center, Korea Institute of Toxicology, Daejeon-si, South Korea
| | - Eunsol Seong
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Eun-Jun Park
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Byoung-Seok Lee
- Toxicologic Pathology Center, Korea Institute of Toxicology, Daejeon-si, South Korea
| | - Sang Jin Lee
- Respiratory Disease Research Center, Korea Institute of Toxicology, Jeongeup, Jellobuk-do, South Korea
| | - Kyuhong Lee
- Respiratory Disease Research Center, Korea Institute of Toxicology, Jeongeup, Jellobuk-do, South Korea
| |
Collapse
|
135
|
Abstract
The human microbiome has been identified as having a key role in health and numerous diseases. Trillions of microbial cells and viral particles comprise the microbiome, each representing modifiable working elements of an intricate bioactive ecosystem. The significance of the human microbiome as it relates to human biology has progressed through culture-dependent (for example, media-based methods) and, more recently, molecular (for example, genetic sequencing and metabolomic analysis) techniques. The latter have become increasingly popular and evolved from being used for taxonomic identification of microbiota to elucidation of functional capacity (sequencing) and metabolic activity (metabolomics). This review summarises key elements of the human microbiome and its metabolic capabilities within the context of health and disease.
Collapse
Affiliation(s)
- Wiley Barton
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| | - Orla O'Sullivan
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| | - Paul D Cotter
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| |
Collapse
|
136
|
Fastrès A, Taminiau B, Vangrinsven E, Tutunaru AC, Moyse E, Farnir F, Daube G, Clercx C. Effect of an antimicrobial drug on lung microbiota in healthy dogs. Heliyon 2019; 5:e02802. [PMID: 31844730 PMCID: PMC6895694 DOI: 10.1016/j.heliyon.2019.e02802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/20/2019] [Accepted: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Alterations of the lung microbiota (LM) are associated with clinical features in chronic lung diseases (CLDs) with growing evidence that an altered LM contributes to the pathogenesis of such disorders. The common use of antimicrobial drugs in the management of CLDs likely represents a confounding factor in the study of the LM. The aim of the present study was to assess the effect of oral administration of amoxicillin/clavulanic acid (AC) on the LM in healthy dogs (n = 6) at short (immediately after stopping AC [D10]) and medium-term (16 days after stopping AC [D26]). Metagenetic analyses were performed on the V1–V3 hypervariable region of 16S rDNA after extraction of total bacterial DNA from samples of bronchoalveolar lavage fluid (BALF). AC did not induce significant changes in BALF cellular counts or in the bacterial load or microbial richness, evenness and α-diversity, while the β-diversity was clearly modified at D10 compared with D0 (before AC administration) and D26 (P < 0.01). The relative abundance of Bacteroidetes and Proteobacteria increased at D10 (P < 0.01) in comparison with D0 and D26 (P < 0.01). The relative abundance of Firmicutes decreased from D0 to D10 (P < 0.01) and increased from D10 to D26 (P < 0.01), but was still lower than at D0 (P < 0.01). The proportion of Actinobacteria increased at D26 compared with D0 and D10 (P < 0.01). Significant differences between timepoints at the level of family, genus or species were not found. In conclusion, in healthy dogs, oral administration of AC induces significant changes in LM at the phyla level and in the β-diversity. Most changes normalize within 2 weeks after discontinuation of AC.
Collapse
Affiliation(s)
- Aline Fastrès
- Department of Clinical Sciences, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Bernard Taminiau
- Department of Food Sciences - Microbiology, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Emilie Vangrinsven
- Department of Clinical Sciences, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Alexandru-Cosmin Tutunaru
- Department of Clinical Sciences, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Evelyne Moyse
- Department of Veterinary Management of Animal Resources, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Frederic Farnir
- Department of Veterinary Management of Animal Resources, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Georges Daube
- Department of Food Sciences - Microbiology, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| | - Cécile Clercx
- Department of Clinical Sciences, FARAH, Faculty of Veterinary Medicine, University of Liège, 4000, Liège, Belgium
| |
Collapse
|
137
|
Shen F, Niu M, Zhou F, Wu Y, Zhu T. Culturability, metabolic activity and composition of ambient bacterial aerosols in a surrogate lung fluid. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 690:76-84. [PMID: 31284198 DOI: 10.1016/j.scitotenv.2019.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/11/2019] [Accepted: 07/01/2019] [Indexed: 06/09/2023]
Abstract
Interactions of particulate matter (PM) and respiratory tract play a crucial role in PM-related respiratory diseases. The majority of the work focuses on the oxidative stress induced by reactions between PM-borne redox-active components and lung lining fluid (LLF). The effects of PM-borne biological components are largely unknown. Of all PM-borne biologicals, bacteria, as living microorganisms, are closely related with inflammatory immune responses. However, its inhalation risk is usually determined without considering the respiratory physiological conditions. In this study, a surrogate lung fluid (SLF) with four typical antioxidants was applied to characterize the ambient bacteria, including concentrations of total bacteria/viable bacteria/culturable bacteria, metabolic activity, bacteria-derived endotoxin, as well as the community structure. Comparing to those determined by SLF, we find that use of PBS leads to an underestimation of the bacterial culturability and metabolic activity. No effect was seen regarding the number of total bacteria and viable bacteria (with intact membrane). Population structure change was seen for bacteria cultured from SLF-collected samples, when compared to that from PBS. Spore-forming bacteria, e.g., genus Bacillus, were found to be easily recovered with SLF. This implies that use of PBS could underestimate the bacteria inhalation risk, especially those bacterial endospores. Our work highlights the necessity to consider the respiratory airway environment when evaluating microbial inhalation risk.
Collapse
Affiliation(s)
- Fangxia Shen
- School of Space and Environment, Beihang University, Beijing 100083, China.
| | - Mutong Niu
- School of Space and Environment, Beihang University, Beijing 100083, China
| | - Feng Zhou
- School of Space and Environment, Beihang University, Beijing 100083, China
| | - Yan Wu
- School of Environmental Science and Engineering, Shandong University, Qingdao 250100, China
| | - Tianle Zhu
- School of Space and Environment, Beihang University, Beijing 100083, China
| |
Collapse
|
138
|
Surendran Nair M, Eucker T, Martinson B, Neubauer A, Victoria J, Nicholson B, Pieters M. Influence of pig gut microbiota on Mycoplasma hyopneumoniae susceptibility. Vet Res 2019; 50:86. [PMID: 31661027 PMCID: PMC6819593 DOI: 10.1186/s13567-019-0701-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/03/2019] [Indexed: 02/07/2023] Open
Abstract
This study investigated the influence of gut microbiome composition in modulating susceptibility to Mycoplasma hyopneumoniae in pigs. Thirty-two conventional M. hyopneumoniae free piglets were randomly selected from six different litters at 3 weeks of age and were experimentally inoculated with M. hyopneumoniae at 8 weeks of age. Lung lesion scores (LS) were recorded 4 weeks post-inoculation (12 weeks of age) from piglet lungs at necropsy. Fecal bacterial community composition of piglets at 3, 8 and 12 weeks of age were targeted by amplifying the V3–V4 region of the 16S rRNA gene. The LS ranged from 0.3 to 43% with an evident clustering of the scores observed in piglets within litters. There were significant differences in species richness and alpha diversity in fecal microbiomes among piglets within litters at different time points (p < 0.05). The dissimilarity matrices indicated that at 3 weeks of age, the fecal microbiota of piglets was more dissimilar compared to those from 8 to 12 weeks of age. Specific groups of bacteria in the gut that might predict the decreased severity of M. hyopneumoniae associated lesions were identified. The microbial shift at 3 weeks of age was observed to be driven by the increase in abundance of the indicator family, Ruminococcaceae in piglets with low LS (p < 0.05). The taxa, Ruminococcus_2 having the highest richness scores, correlated significantly between litters showing stronger associations with the lowest LS (r = −0.49, p = 0.005). These findings suggest that early life gut microbiota can be a potential determinant for M. hyopneumoniae susceptibility in pigs.
Collapse
Affiliation(s)
- Meera Surendran Nair
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Tyson Eucker
- Boehringer-Ingelheim Animal Health, Duluth, GA, USA
| | | | | | | | | | - Maria Pieters
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA.
| |
Collapse
|
139
|
Ordoñez-Rueda D, Baying B, Pavlinic D, Alessandri L, Yeboah Y, Landry JJM, Calogero R, Benes V, Paulsen M. Apoptotic Cell Exclusion and Bias-Free Single-Cell Selection Are Important Quality Control Requirements for Successful Single-Cell Sequencing Applications. Cytometry A 2019; 97:156-167. [PMID: 31603610 DOI: 10.1002/cyto.a.23898] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/27/2019] [Accepted: 09/05/2019] [Indexed: 12/23/2022]
Abstract
Single-cell sequencing experiments are a new mainstay in biology and have been advancing science especially in the biomedical field. The high pressure to integrate the technology into daily laboratory live requires solid knowledge with respect to potential limitations and precautions to be taken care of before applying it to complex research questions. In the past, we have identified two issues with quality measures neglected by the growing community involving SmartSeq and droplet or micro-well-based scRNASeq methods (1) how to ensure that only single cells are introduced without biasing on light scattering when handling complex cell mixtures and organ preparations or (2) how best to control for (pro-)apoptotic cell contaminations in single-cell sequencing approaches. Sighting of concurrent literature involving single-cell sequencing technologies revealed that these topics are generally neglected or simply approached in silico but not at the bench before generating single-cell data sets. We fear that those important quality aspects are overlooked due to reduced awareness of their importance for guaranteeing the quality of experiments. In this Cytometry rigor issue, we provide experimentally supported guidance on how to circumvent those critical shortcomings in order to promote a better use of the fantastic single-cell sequencing toolbox in biology. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Diana Ordoñez-Rueda
- Flow Cytometry Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Bianka Baying
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Dinko Pavlinic
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Luca Alessandri
- Bioinformatics and Genomics Unit, MBC Molecular Biotechnology Center, Torino, Italy
| | - Yvonne Yeboah
- Flow Cytometry Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Raffaele Calogero
- Bioinformatics and Genomics Unit, MBC Molecular Biotechnology Center, Torino, Italy
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Malte Paulsen
- Flow Cytometry Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
140
|
Shen F, Zheng Y, Niu M, Zhou F, Wu Y, Wang J, Zhu T, Wu Y, Wu Z, Hu M, Zhu T. Characteristics of biological particulate matters at urban and rural sites in the North China Plain. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 253:569-577. [PMID: 31330349 DOI: 10.1016/j.envpol.2019.07.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 06/10/2023]
Abstract
Depending on their concentrations, sizes, and types, particulate matters of biological origins (bioPM) significantly affect human health. However, for different air environments, they are not well characterized and can vary considerably. As an example, we investigated the bioPM differences at an urban (Beijing) site and a rural (Wangdu) site in the North China Plain (NCP) using an online monitoring instrument, an ultraviolet aerodynamic particle sizer (UV-APS), the limulus amebocyte lysate (LAL) assay, and a high-throughput sequencing method. Generally, lower concentrations of viable bioPM (hourly mean: 1.3 × 103 ± 1.6 × 103 m-3) and endotoxin (0.66 ± 0.16 EU/m3) in Beijing were observed compared to viable bioPM (0.79 × 105 ± 1.4 × 105 m-3) and endotoxin (15.1 ± 23.96 EU/m3) at the Wangdu site. The percentage of viable bioPM number concentration in the total PM was 3.1% in Beijing and 6.4% in Wangdu. Approximately 80% of viable bioPM was found to be in the range from 1 to 2.5 μm. Nevertheless, the size distribution patterns for viable bioPM at the Beijing and Wangdu sites differed and were affected by PM pollution, leading to distinct lung deposition profiles. Moreover, the distinct diurnal variations in viable bioPM on clean days were dimmed by the PM pollution at both sites. Distinct bacterial community structures were found in the air from the Beijing and Wangdu sites. The bacterial community in urban Beijing was dominated by genus Lactococcus (49.5%) and Pseudomonas (15.1%), while the rural Wangdu site was dominated by Enterococcus (65%) and Paenibacillus (10%). Human-derived genera, including Myroides, Streptococcus, Propionibacterium, Dietzia, Helcococcus, and Facklamia, were higher in Beijing, suggesting bacterial emission from humans in the urban air environment. Our results show that different air harbors different biological species, and people residing in different environments thus could have very different biological particle exposure.
Collapse
Affiliation(s)
- Fangxia Shen
- School of Space and Environment, Beihang University, Beijing, 102206, China.
| | - Yunhao Zheng
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Mutong Niu
- School of Space and Environment, Beihang University, Beijing, 102206, China
| | - Feng Zhou
- School of Space and Environment, Beihang University, Beijing, 102206, China
| | - Yan Wu
- School of Environmental Science and Engineering, Shandong University, Qingdao, 250100, China
| | - Junxia Wang
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Tong Zhu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Yusheng Wu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Zhijun Wu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Min Hu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Tianle Zhu
- School of Space and Environment, Beihang University, Beijing, 102206, China
| |
Collapse
|
141
|
Abstract
Although chronic obstructive pulmonary disease (COPD) is regarded as a chronic inflammatory lung disease, the disease mechanism is still not known. Intriguingly, aging lungs are quite similar to COPD-affected lungs in many ways, and COPD has been viewed as a disease of accelerated premature aging of the lungs. In this paper, based on a literature review, we would like to propose immunosenescence, age-associated decline in immunity, as a critical mechanism for the development of COPD. Immunosenescence can cause a low-grade, systemic inflammation described as inflammaging. This inflammaging may be directly involved in the COPD pathogenesis. The potential contributors to the development of inflammaging in the lungs possibly leading to COPD are discussed in the review paper. A notable fact about COPD is that only 15% to 20% of smokers develop clinically significant COPD. Given that there is a substantial inter-individual variation in inflammaging susceptibility, which is genetically determined and significantly affected by the history of the individual's exposure to pathogens, immunosenescence and inflammaging may also provide the answer for this unexpectedly low susceptibility of smokers to clinically significant COPD.
Collapse
Affiliation(s)
- Won Kyung Cho
- Department of Pulmonary and Critical Care Medicine, International Healthcare Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Division of Biology and Medical Sciences, Brown University, Providence, RI, USA
| | - Lark Kyun Kim
- Severance Biomedical Science Institute and BK21 PLUS Project for Medical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
142
|
de Souza GF, Muraro SP, Santos LD, Monteiro APT, da Silva AG, de Souza APD, Stein RT, Bozza PT, Porto BN. Macrophage migration inhibitory factor (MIF) controls cytokine release during respiratory syncytial virus infection in macrophages. Inflamm Res 2019; 68:481-491. [PMID: 30944975 DOI: 10.1007/s00011-019-01233-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/15/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE AND DESIGN Respiratory syncytial virus (RSV) is the major cause of infection in children up to 2 years old and reinfection is very common among patients. Tissue damage in the lung caused by RSV leads to an immune response and infected cells activate multiple signaling pathways and massive production of inflammatory mediators like macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine. Therefore, we sought to investigate the role of MIF during RSV infection in macrophages. METHODS We evaluated MIF expression in BALB/c mice-derived macrophages stimulated with different concentrations of RSV by Western blot and real-time PCR. Additionally, different inhibitors of signaling pathways and ROS were used to evaluate their importance for MIF expression. Furthermore, we used a specific MIF inhibitor, ISO-1, to evaluate the role of MIF in viral clearance and in RSV-induced TNF-α, MCP-1 and IL-10 release from macrophages. RESULTS We showed that RSV induces MIF expression dependently of ROS, 5-LOX, COX and PI3K activation. Moreover, viral replication is necessary for RSV-triggered MIF expression. Differently, p38 MAPK in only partially needed for RSV-induced MIF expression. In addition, MIF is important for the release of TNF-α, MCP-1 and IL-10 triggered by RSV in macrophages. CONCLUSIONS In conclusion, we demonstrate that MIF is expressed during RSV infection and controls the release of pro-inflammatory cytokines from macrophages in an in vitro model.
Collapse
Affiliation(s)
- Gabriela F de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Stéfanie P Muraro
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Leonardo D Santos
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Ana Paula T Monteiro
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Amanda G da Silva
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Ana Paula D de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Renato T Stein
- Laboratory of Pediatric Respirology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Bárbara N Porto
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
143
|
Tian X, Hellman J, Horswill AR, Crosby HA, Francis KP, Prakash A. Elevated Gut Microbiome-Derived Propionate Levels Are Associated With Reduced Sterile Lung Inflammation and Bacterial Immunity in Mice. Front Microbiol 2019; 10:159. [PMID: 30891007 PMCID: PMC6413706 DOI: 10.3389/fmicb.2019.00159] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acids (SCFA) are important dietary and microbiome metabolites that can have roles in gut immunity as well as further afield. We previously observed that gut microbiome alteration via antibiotics led to attenuated lung inflammatory responses. The rationale for this study was to identify gut microbiome factors that regulate lung immune homeostasis. We first investigated key factors within mouse colonic lumen filtrates (CLF) which could elicit direct inflammatory effects in vitro. We identified lipopolysaccharide (LPS) and SCFAs as key CLF ingredients whose levels and inflammatory capacity changed after antibiotic exposure in mice. Specifically, the SCFA propionate appeared to be a key regulator of LPS responses in vitro. Elevated propionate: acetate ratios, as seen in CLF after antibiotic exposure, strongly blunted inflammatory responses in vitro. In vivo, exposure of lungs to high dose propionate, to mimic how prior antibiotic exposure changed SCFA levels, resulted in diminished immune containment of Staphylococcus aureus pneumonia. Finally, we discovered an enrichment of propionate-producing gut bacteria in mice with reduced lung inflammation following lung ischemia reperfusion injury in vivo. Overall, our data show that propionate levels can distinctly modulate lung immune responses in vitro and in vivo and that gut microbiome increased production of propionate is associated with reduced lung inflammation.
Collapse
Affiliation(s)
- Xiaoli Tian
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Heidi A. Crosby
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | | | - Arun Prakash
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States.,San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
Approved by: Frontiers in Microbiology Editorial Office, Frontiers Media SA, Switzerland
| |
Collapse
|
144
|
Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, Ameh S, Sandel D, Liang XS, Mazzilli S, Whary MT, Meyerson M, Germain R, Blainey PC, Fox JG, Jacks T. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019; 176:998-1013.e16. [PMID: 30712876 DOI: 10.1016/j.cell.2018.12.040] [Citation(s) in RCA: 601] [Impact Index Per Article: 120.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 11/01/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Lung cancer is closely associated with chronic inflammation, but the causes of inflammation and the specific immune mediators have not been fully elucidated. The lung is a mucosal tissue colonized by a diverse bacterial community, and pulmonary infections commonly present in lung cancer patients are linked to clinical outcomes. Here, we provide evidence that local microbiota provoke inflammation associated with lung adenocarcinoma by activating lung-resident γδ T cells. Germ-free or antibiotic-treated mice were significantly protected from lung cancer development induced by Kras mutation and p53 loss. Mechanistically, commensal bacteria stimulated Myd88-dependent IL-1β and IL-23 production from myeloid cells, inducing proliferation and activation of Vγ6+Vδ1+ γδ T cells that produced IL-17 and other effector molecules to promote inflammation and tumor cell proliferation. Our findings clearly link local microbiota-immune crosstalk to lung tumor development and thereby define key cellular and molecular mediators that may serve as effective targets in lung cancer intervention.
Collapse
Affiliation(s)
- Chengcheng Jin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Georgia K Lagoudas
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chen Zhao
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Susan Bullman
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Arjun Bhutkar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Bo Hu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Samuel Ameh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Demi Sandel
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Xu Sue Liang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sarah Mazzilli
- Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mark T Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Matthew Meyerson
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ronald Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Paul C Blainey
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - James G Fox
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
145
|
Human pluripotent stem cell-derived alveolar epithelial cells are alternatives for in vitro pulmotoxicity assessment. Sci Rep 2019; 9:505. [PMID: 30679658 PMCID: PMC6346100 DOI: 10.1038/s41598-018-37193-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/29/2018] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived alveolar epithelial cells (AECs) provide new opportunities for understanding lung development and the treatment of pulmonary diseases. However, toxicity assessments using hPSC-AECs have not been undertaken. In this study, we generated functional AECs from hPSCs and evaluated their inflammatory and apoptotic responses to cadmium (Cd) exposure (1, 5, and 10 μM) for 24 h compared with the human bronchial epithelial cell line (BEAS-2B) and primary AECs as controls. Our data showed that Cd (10 μM) treatment induced substantial inflammatory responses and apoptosis in BEAS-2B cells, but not in both hPSC-AECs and primary AECs. Interestingly, conditioned medium from AEC cultures significantly alleviated apoptotic and inflammatory responses to Cd exposure in BEAS-2B cells. Using cytokine arrays, several potential factors secreted from hPSC-AECs and primary AECs were detected and may be involved in reducing Cd-induced cytotoxicity. We also observed higher expression of surfactant proteins B and C in both hPSC-AECs and primary AECs, which may contribute to protection against Cd-induced cytotoxicity. These results suggested that hPSC-AECs phenotypically and functionally resemble primary AECs and could be more biologically relevant alternatives for evaluating the pathological contribution of confirmed or potential pulmotoxic materials included in smoking and microdust.
Collapse
|
146
|
Wang H, Zhou Q, Dai W, Feng X, Lu Z, Yang Z, Liu Y, Xie G, Yang Y, Shen K, Li Y, Li SC, Xu X, Shen Y, Li D, Zheng Y. Lung Microbiota and Pulmonary Inflammatory Cytokines Expression Vary in Children With Tracheomalacia and Adenoviral or Mycoplasma pneumoniae Pneumonia. Front Pediatr 2019; 7:265. [PMID: 31316955 PMCID: PMC6611399 DOI: 10.3389/fped.2019.00265] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
Community-acquired pneumonia (CAP) is a worldwide infectious disease caused by bacteria, viruses, or a combination of these infectious agents. Mycoplasma pneumoniae is an atypical pneumonia pathogen that causes high morbidity and mortality in children, and adenovirus can lead to severe pneumonia. However, the etiology of different types of pneumonia is still unclear. In this study, we selected a total of 52 inpatients with M. pneumoniae pneumonia (MPP) (n = 21), adenovirus pneumonia (AVP) (n = 16), or tracheomalacia (n = 15) to serve as a disease control. Bronchoalveolar lavage fluid (BALF) samples that had been obtained for clinical use were analyzed. We compared the differences in microbiota and the expression of 10 inflammatory cytokines in samples between MPP, AVP, and tracheomalacia. We found that the bacterial diversity in MPP was lower than that in AVP and tracheomalacia. Mycoplasma, Streptococcus, and Pseudomonas were predominant in samples of MPP, AVP, and tracheomalacia, respectively. The expression levels of IL-6, IL-8, and IL-10 were significantly higher in inpatients with AVP compared to children hospitalized with tracheomalacia or MPP. The lung microbiota in MPP was remarkably correlated with IL-2, IL-4, IL-5, IL-6, TNF-α, and IL-1α expressions, while this was not found in tracheomalacia and AVP. Microbiota analysis identified a high load of multi-drug resistant Acinetobacter baumannii in the lung microbiota of several inpatients, which might be associated with the long hospitalization length and intra-group differences at the individual level. This study will help to understand the microbial etiology of tracheomalacia, AVP, and MPP and to identify effective therapies for these diseases.
Collapse
Affiliation(s)
- Heping Wang
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Qian Zhou
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, China
| | - Wenkui Dai
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Xin Feng
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, China
| | - Zhiwei Lu
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Zhenyu Yang
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, China
| | - Yanhong Liu
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, China
| | - Gan Xie
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Yonghong Yang
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China.,Department of Microbial Research, WeHealthGene Institute, Shenzhen, China.,Department of Respiratory Diseases, Beijing Children's Hospital, Beijing, China
| | - Kunling Shen
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China.,Department of Respiratory Diseases, Beijing Children's Hospital, Beijing, China
| | - Yinhu Li
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Ximing Xu
- Institute of Statistics, NanKai University, Tianjin, China
| | - Yongshun Shen
- Department of Pediatrics, Shenzhen Dapeng District Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Dongfang Li
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, China.,Institute of Statistics, NanKai University, Tianjin, China
| | - Yuejie Zheng
- Department of Respiratory Diseases, Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
147
|
Ouchi R, Kawano T, Yoshida H, Ishii M, Miyasaka T, Ohkawara Y, Takayanagi M, Takahashi T, Ohno I. Maternal Separation as Early-Life Stress Causes Enhanced Allergic Airway Responses by Inhibiting Respiratory Tolerance in Mice. TOHOKU J EXP MED 2018; 246:155-165. [PMID: 30405003 DOI: 10.1620/tjem.246.155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Epidemiologic studies indicate that exposure to psychosocial stress in early childhood is a risk factor of adult-onset asthma, but the mechanisms of this relationship are poorly understood. Therefore, we examined whether early-life stress increases susceptibility to adult-onset asthma by inhibiting the development of respiratory tolerance. Neonatal BALB/c female mice were aerosolized with ovalbumin (OVA) to induce immune tolerance prior to immune sensitization with an intraperitoneal injection of OVA and the adjuvant aluminum hydroxide. Maternal separation (MS) was applied as an early-life stressor during the induction phase of immune tolerance. The mice were challenged with OVA aerosol in adulthood, and allergic airway responses were evaluated, including airway hyper-responsiveness to inhaled methacholine, inflammatory cell infiltration, bronchoalveolar lavage fluid levels of interleukin (IL)-4, IL-5, and IL-13, and serum OVA-specific IgE. We then evaluated the effects of MS on the development of regulatory T (Treg) cells in bronchial lymph nodes (BLN) and on splenocyte proliferation and cytokine expression. In mice that underwent MS and OVA tolerization, the allergic airway responses and OVA-induced proliferation and IL-4 expression of splenocytes were significantly enhanced. Furthermore, exposure to MS was associated with a lower number of Treg cells in the BLN. These findings suggest that exposure to early-life stress prevents the acquisition of respiratory tolerance to inhaled antigen due to insufficient Treg cell development, resulting in Th2-biased sensitization and asthma onset. We provide the evidence for inhibitory effects of early-life stress on immune tolerance. The present findings may help to clarify the pathogenesis of adult-onset asthma.
Collapse
Affiliation(s)
- Ryusuke Ouchi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Tasuku Kawano
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Hitomi Yoshida
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Masato Ishii
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Tomomitsu Miyasaka
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Yuichi Ohkawara
- Division of Experimental Allergy and Immunology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Motoaki Takayanagi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University
| | - Isao Ohno
- Center for Medical Education, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| |
Collapse
|
148
|
Invited Review: From nose to gut – the role of the microbiome in neurological disease. Neuropathol Appl Neurobiol 2018; 45:195-215. [DOI: 10.1111/nan.12520] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/29/2018] [Indexed: 12/14/2022]
|
149
|
Li H, Luo YF, Wang YS, Xiao YL, Cai HR, Xie CM. Pseudomonas aeruginosa induces cellular senescence in lung tissue at the early stage of two-hit septic mice. Pathog Dis 2018; 76:5289408. [PMID: 30649401 DOI: 10.1093/femspd/ftz001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023] Open
Abstract
We presume that severe secondary Pseudomonas aeruginosa (PA) infection can lead to cellular senescence in lung tissue and thus contribute to high mortality. We established a two-hit mouse model using cecal ligation and puncture (CLP) followed by sublethal PA lung infection. In lung tissue, increased infiltration of inflammatory cells, elevated lung injury and augmented cellular senescence was shown in mice with CLP followed by sublethal PA infection, and these observations reached a higher rank when higher (H) loads PA (PAO1) were administered to CLP mice (CLP + PAO1-H). Accordingly, oxidative stress-related element gp91phox and inflammation regulator NF-κB were greatly activated in CLP + PAO1-H mice compared to others. There was no obvious inflammation or cellular senescence in sham control, PAO1-infected mice. Consequently, CLP + PAO1-H mice had the highest expression levels of inflammatory cytokines IL-6, TNFα and iNOS among those groups. There was lower bacterial clearance ability in CLP + PAO1-H mice than in other mice. CLP + PAO1-H only had approximately 10% survival after 7 days of investigation and was much lower than others. In conclusion, higher mortality due to increased lung inflammation and cellular senescence are observed in mice with increased loads of PA infection secondary to CLP.
Collapse
Affiliation(s)
- Hui Li
- Department of Respiratory Medicine, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Yi-Feng Luo
- Department of Respiratory Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yong-Sheng Wang
- Department of Respiratory Medicine, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Yong-Long Xiao
- Department of Respiratory Medicine, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Hou-Rong Cai
- Department of Respiratory Medicine, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Can-Mao Xie
- Department of Respiratory Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
150
|
Antigen-specific regulatory T-cell responses against aeroantigens and their role in allergy. Mucosal Immunol 2018; 11:1537-1550. [PMID: 29858582 DOI: 10.1038/s41385-018-0038-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 02/04/2023]
Abstract
The mucosal immune system of the respiratory tract is specialized to continuously monitor the external environment and to protect against invading pathogens, while maintaining tolerance to innocuous inhaled particles. Allergies result from a loss of tolerance against harmless antigens characterized by formation of allergen-specific Th2 cells and IgE. Tolerance is often described as a balance between harmful Th2 cells and various types of protective "regulatory" T cells. However, the identity of the protective T cells in healthy vs. allergic individuals or following successful allergen-specific therapy is controversially discussed. Recent technological progress enabling the identification of antigen-specific effector and regulatory T cells has significantly contributed to our understanding of tolerance. Here we discuss the experimental evidence for the various tolerance mechanisms described. We try to integrate the partially contradictory data into a new model proposing different mechanism of tolerance depending on the quality and quantity of the antigens as well as the way of antigen exposure. Understanding the basis of tolerance is essential for the rational design of novel and more efficient immunotherapies.
Collapse
|