51
|
Abstract
Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
52
|
Ferrara F, Rial A, Suárez N, Chabalgoity JA. Polyvalent Bacterial Lysate Protects Against Pneumonia Independently of Neutrophils, IL-17A or Caspase-1 Activation. Front Immunol 2021; 12:562244. [PMID: 33981296 PMCID: PMC8108696 DOI: 10.3389/fimmu.2021.562244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 03/18/2021] [Indexed: 11/28/2022] Open
Abstract
Polyvalent bacterial lysates have been in use for decades for prevention and treatment of respiratory infections with reported clinical benefits. However, besides claims of broad immune activation, the mode of action is still a matter of debate. The lysates, formulated with the main bacterial species involved in respiratory infections, are commonly prepared by chemical or mechanical disruption of bacterial cells, what is believed influences the biological activity of the product. Here, we prepared two polyvalent lysates with the same composition but different method of bacterial cell disruption and evaluated their biological activity in a comparative fashion. We found that both bacterial lysates induce NF-kB activation in a MyD88 dependent manner, suggesting they work as TLR agonists. Further, we found that a single intranasal dose of any of the two lysates, is sufficient to protect against pneumococcal pneumonia, suggesting that they exert similar biological activity. We have previously shown that protection against pneumococcal pneumonia can also be induced by prior S. pneumoniae sub lethal infection or therapeutic treatment with a TLR5 agonist. Protection in those cases depends on neutrophil recruitment to the lungs, and can be associated with increased local expression of IL-17A. Here, we show that bacterial lysates exert protection against pneumococcal pneumonia independently of neutrophils, IL-17A or Caspase-1/11 activation, suggesting the existence of redundant mechanisms of protection. Trypsin-treated lysates afford protection to the same extent, suggesting that just small peptides suffice to exert the protective effect or that the molecules responsible for the protective effect are not proteins. Understanding the mechanism of action of bacterial lysates and deciphering the active components shall allow redesigning them with more precisely defined formulations and expanding their range of action.
Collapse
Affiliation(s)
- Florencia Ferrara
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - Analía Rial
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - Norma Suárez
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - José Alejandro Chabalgoity
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| |
Collapse
|
53
|
Zajac M, Dreano E, Edwards A, Planelles G, Sermet-Gaudelus I. Airway Surface Liquid pH Regulation in Airway Epithelium Current Understandings and Gaps in Knowledge. Int J Mol Sci 2021; 22:3384. [PMID: 33806154 PMCID: PMC8037888 DOI: 10.3390/ijms22073384] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Knowledge on the mechanisms of acid and base secretion in airways has progressed recently. The aim of this review is to summarize the known mechanisms of airway surface liquid (ASL) pH regulation and their implication in lung diseases. Normal ASL is slightly acidic relative to the interstitium, and defects in ASL pH regulation are associated with various respiratory diseases, such as cystic fibrosis. Basolateral bicarbonate (HCO3-) entry occurs via the electrogenic, coupled transport of sodium (Na+) and HCO3-, and, together with carbonic anhydrase enzymatic activity, provides HCO3- for apical secretion. The latter mainly involves CFTR, the apical chloride/bicarbonate exchanger pendrin and paracellular transport. Proton (H+) secretion into ASL is crucial to maintain its relative acidity compared to the blood. This is enabled by H+ apical secretion, mainly involving H+/K+ ATPase and vacuolar H+-ATPase that carry H+ against the electrochemical potential gradient. Paracellular HCO3- transport, the direction of which depends on the ASL pH value, acts as an ASL protective buffering mechanism. How the transepithelial transport of H+ and HCO3- is coordinated to tightly regulate ASL pH remains poorly understood, and should be the focus of new studies.
Collapse
Affiliation(s)
- Miroslaw Zajac
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Elise Dreano
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA;
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Laboratoire de Physiologie rénale et Tubulopathies, CNRS ERL 8228, 75006 Paris, France
| | - Isabelle Sermet-Gaudelus
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Centre de Référence Maladies Rares, Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, 75015 Paris, France
- Clinical Trial Network, European Cystic Fibrosis Society, BT2 Belfast, Ireland
- European Respiratory Network Lung, 75006 Paris, France
| |
Collapse
|
54
|
Domon H, Terao Y. The Role of Neutrophils and Neutrophil Elastase in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2021; 11:615959. [PMID: 33796475 PMCID: PMC8008068 DOI: 10.3389/fcimb.2021.615959] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae, also known as pneumococcus, is a Gram-positive diplococcus and a major human pathogen. This bacterium is a leading cause of bacterial pneumonia, otitis media, meningitis, and septicemia, and is a major cause of morbidity and mortality worldwide. To date, studies on S. pneumoniae have mainly focused on the role of its virulence factors including toxins, cell surface proteins, and capsules. However, accumulating evidence indicates that in addition to these studies, knowledge of host factors and host-pathogen interactions is essential for understanding the pathogenesis of pneumococcal diseases. Recent studies have demonstrated that neutrophil accumulation, which is generally considered to play a critical role in host defense during bacterial infections, can significantly contribute to lung injury and immune subversion, leading to pneumococcal invasion of the bloodstream. Here, we review bacterial and host factors, focusing on the role of neutrophils and their elastase, which contribute to the progression of pneumococcal pneumonia.
Collapse
Affiliation(s)
- Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
55
|
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are caused by an exaggerated inflammatory response arising from a wide variety of pulmonary and systemic insults. Lung tissue is composed of a variety of cell populations, including parenchymal and immune cells. Emerging evidence has revealed that multiple cell populations in the lung work in concert to regulate lung inflammation in response to both direct and indirect stimulations. To date, the question of how different types of pulmonary cells communicate with each other and subsequently regulate or modulate inflammatory cascades remains to be fully addressed. In this review, we provide an overview of current advancements in understanding the role of cell-cell interaction in the development of ALI and depict molecular mechanisms by which cell-cell interactions regulate lung inflammation, focusing on inter-cellular activities and signaling pathways that point to possible therapeutic opportunities for ALI/ARDS.
Collapse
Affiliation(s)
- Huiting Zhou
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Erica K. Fan
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
56
|
Hanchanachai N, Chumnanpuen P, E-Kobon T. Interaction study of Pasteurella multocida with culturable aerobic bacteria isolated from porcine respiratory tracts using coculture in conditioned media. BMC Microbiol 2021; 21:19. [PMID: 33422011 PMCID: PMC7796573 DOI: 10.1186/s12866-020-02071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/15/2020] [Indexed: 11/14/2022] Open
Abstract
Background The porcine respiratory tract harbours multiple microorganisms, and the interactions between these organisms could be associated with animal health status. Pasteurella multocida is a culturable facultative anaerobic bacterium isolated from healthy and diseased porcine respiratory tracts. The interaction between P. multocida and other aerobic commensal bacteria in the porcine respiratory tract is not well understood. This study aimed to determine the interactions between porcine P. multocida capsular serotype A and D strains and other culturable aerobic bacteria isolated from porcine respiratory tracts using a coculture assay in conditioned media followed by calculation of the growth rates and interaction parameters. Results One hundred and sixteen bacterial samples were isolated from five porcine respiratory tracts, and 93 isolates were identified and phylogenetically classified into fourteen genera based on 16S rRNA sequences. Thirteen isolates from Gram-negative bacterial genera and two isolates from the Gram-positive bacterial genus were selected for coculture with P. multocida. From 17 × 17 (289) interaction pairs, the majority of 220 pairs had negative interactions indicating competition for nutrients and space, while 17 pairs were identified as mild cooperative or positive interactions indicating their coexistence. All conditioned media, except those of Acinetobacter, could inhibit P. multocida growth. Conversely, the conditioned media of P. multocida also inhibited the growth of nine isolates plus themselves. Conclusion Negative interaction was the major interactions among the coculture of these 15 representative isolates and the coculture with P. multocida. The conditioned media in this study might be further analysed to identify critical molecules and examined by the in vivo experiments. The study proposed the possibility of using these molecules in conditioned media to control P. multocida growth. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-020-02071-4.
Collapse
Affiliation(s)
- Nonzee Hanchanachai
- Interdisciplinary Graduate Program in Bioscience, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.,Computational Biomodelling Laboratory for Agricultural Science and Technology, Kasetsart University, Bangkok, 10900, Thailand
| | - Pramote Chumnanpuen
- Computational Biomodelling Laboratory for Agricultural Science and Technology, Kasetsart University, Bangkok, 10900, Thailand.,Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Teerasak E-Kobon
- Computational Biomodelling Laboratory for Agricultural Science and Technology, Kasetsart University, Bangkok, 10900, Thailand. .,Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand. .,Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand.
| |
Collapse
|
57
|
Cellular and functional heterogeneity of the airway epithelium. Mucosal Immunol 2021; 14:978-990. [PMID: 33608655 PMCID: PMC7893625 DOI: 10.1038/s41385-020-00370-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/15/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
The airway epithelium protects us from environmental insults, which we encounter with every breath. Not only does it passively filter large particles, it also senses potential danger and alerts other cells, including immune and nervous cells. Together, these tissues orchestrate the most appropriate response, balancing the need to eliminate the danger with the risk of damage to the host. Each cell subset within the airway epithelium plays its part, and when impaired, may contribute to the development of respiratory disease. Here we highlight recent advances regarding the cellular and functional heterogeneity along the airway epithelium and discuss how we can use this knowledge to design more effective, targeted therapeutics.
Collapse
|
58
|
Han B, Hoang BX. Opinions on the current pandemic of COVID-19: Use functional food to boost our immune functions. J Infect Public Health 2020; 13:1811-1817. [PMID: 32948484 PMCID: PMC7831995 DOI: 10.1016/j.jiph.2020.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022] Open
Abstract
The pandemic of novel coronavirus caused COVID-19 had resulted in a high number of hospitalizations and deaths and caused a devastating toll on human and society health. The symptoms of the infected patients vary significantly, from life-threatening to mild or even asymptomatic. This clinical observation led to hypothesize on the critical role of host innate immunity in the disease development and progression. As the first defense barrier against microorganisms, the innate immune reaction determines not only the viral infection rate but also immune-mediated response. Therefore, promote healthy behaviors to enhance innate immunity with functional food and nutritional agents may be a rational strategy for minimizing damages caused by viruses to global health.
Collapse
Affiliation(s)
- Bo Han
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States
| | - Ba X Hoang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
59
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
60
|
Che KF, Tengvall S, Lindén A. Interleukin-26 in host defense and inflammatory disorders of the airways. Cytokine Growth Factor Rev 2020; 57:1-10. [PMID: 33293237 DOI: 10.1016/j.cytogfr.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 11/16/2022]
Abstract
The dimeric cytokine interleukin (IL)-26 belongs to the IL-10 family. Whereas it was originally perceived as a T-helper (Th)17 cytokine, subsequent studies have shown that IL-26 is produced by several populations of leukocytes and structural cells. This cytokine binds to a heterodimeric receptor complex including IL-10R2 and -20R1 (IL-26R) and signals through STAT 1 and 3 to induce the release of chemokines and growth factors. Remarkably, IL-26 directly kills bacteria and inhibits viral replication. The most recent studies on human airways confirm multiple cellular sources in this critical interphase of host defense and demonstrate that stimulation of toll-like receptors (TLR) trigger the release of IL-26. Once released, it exerts a dualistic effect on cytokine production and up-regulates gene expression of IL-26R. It also potentiates chemotaxis and inhibits chemokinesis for neutrophils, thereby facilitating the accumulation of innate effector cells at the site of bacterial stimulation. The high levels of IL-26 in human airways are altered in inflammatory airway disorders such as asthma and chronic obstructive pulmonary disease. Thus, IL-26 emerges as an important mediator, providing direct and indirect actions on microbes, actions that are essential for host defense and inflammation and bears potential as a biomarker of disease.
Collapse
Affiliation(s)
- Karlhans Fru Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-17177, Sweden.
| | - Sara Tengvall
- Närhälsan, Frölunda Vårdcentral, Gothenburg, SE-421 42, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-17177, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, SE-171 76, Sweden
| |
Collapse
|
61
|
Ko HK, Lin AH, Perng DW, Lee TS, Kou YR. Lung Epithelial TRPA1 Mediates Lipopolysaccharide-Induced Lung Inflammation in Bronchial Epithelial Cells and Mice. Front Physiol 2020; 11:596314. [PMID: 33281629 PMCID: PMC7705107 DOI: 10.3389/fphys.2020.596314] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/30/2020] [Indexed: 12/22/2022] Open
Abstract
Toll-like receptor (TLR) 4 was originally thought to be the sole pattern recognition receptor for lipopolysaccharide (LPS). Transient receptor potential ankyrin 1 (TRPA1), a Ca2+-permeant channel, has been suggested as a non-TLR receptor membrane-bound sensor of LPS. We recently reported that TRPA1 is expressed in lung epithelial cells (LECs) and mediates lung inflammation induced by cigarette smoke. However, the role of TRPA1 in LPS-induced lung inflammation has not been conclusively defined, and its underlying cellular mechanisms remain unclear. In this study, our in vitro results showed that LPS sequentially produced a cascade of events, including the elevation of intracellular Ca2+, the activation of NADPH oxidase, increase in intracellular reactive oxygen species (ROS), the activation of mitogen-activated protein kinase (MAPK)/nuclear factor-kB (NF-κB) signaling, and the induction of IL-8. The increase in intracellular Ca2+ was inhibited by HC030031 (a TRPA1 antagonist) but was unaffected by TAK-242 (a TLR-4 inhibitor). The activation of NADPH oxidase was prevented by its inhibitor apocynin, EGTA (an extracellular Ca2+ chelator), and HC030031. The increase in intracellular ROS was attenuated by apocynin, N-acetyl-cysteine (NAC, a ROS scavenger), EGTA, and HC030031. The activation of the MAPK/NF-κB signaling was halted by NAC, EGTA, and HC030031. IL-8 induction was suppressed by HC030031 and TRPA1 siRNA, and further reduced by the combination of HC030031 and TAK-242. Our in vivo studies showed that trpa1–/– mice exhibited a reduced level of LPS-induced lung inflammation compared with wild-type mice as evidenced by the alleviations of increases in vascular permeability, inflammatory cell infiltration, inflammatory cytokine levels, oxidative stress, and MAPK signaling activation. Thus, in LECs, LPS may activate TRPA1 resulting in an increase in Ca2+ influx. The increased intracellular Ca2+ leads to NADPH oxidase activation, which causes an increase in intracellular ROS. The intracellular ROS activates the MAPK/NF-κB signaling resulting in IL-8 induction. This mechanism may possibly be at work to induce lung inflammation in mice.
Collapse
Affiliation(s)
- Hsin-Kuo Ko
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - An-Hsuan Lin
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Diahn-Warng Perng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu Ru Kou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
62
|
van der Heide SL, Xi Y, Upham JW. Natural Killer Cells and Host Defense Against Human Rhinoviruses Is Partially Dependent on Type I IFN Signaling. Front Cell Infect Microbiol 2020; 10:510619. [PMID: 33194777 PMCID: PMC7609819 DOI: 10.3389/fcimb.2020.510619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Rhinovirus (RV), the causative agent of the common cold, causes only mild upper respiratory tract infections in healthy individuals, but can cause longer lasting and more severe pulmonary infections in people with chronic lung diseases and in the setting of immune suppression or immune deficiency. RV-infected lung structural cells release type I interferon (IFN-I), initiating the immune response, leading to protection against viruses in conjunction with migratory immune cells. However, IFN-I release is deficient in some people with asthma. Innate immune cells, such as natural killer (NK) cells, are proposed to play major roles in the control of viral infections, and may contribute to exacerbations of chronic lung diseases, such as asthma. In this study, we characterized the NK cell response to RV infection using an in vitro model of infection in healthy individuals, and determined the extent to which IFN-I signaling mediates this response. The results indicate that RV stimulation in vitro induces NK cell activation in healthy donors, leading to degranulation and the release of cytotoxic mediators and cytokines. IFN-I signaling was partly responsible for NK cell activation and functional responses to RV. Overall, our findings suggest the involvement of NK cells in the control of RV infection in healthy individuals. Further understanding of NK cell regulation may deepen our understanding of the mechanisms that contribute to susceptibility to RV infections in asthma and other chronic lung diseases.
Collapse
Affiliation(s)
- Saskia L van der Heide
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Yang Xi
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - John W Upham
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
63
|
Bissonnette EY, Lauzon-Joset JF, Debley JS, Ziegler SF. Cross-Talk Between Alveolar Macrophages and Lung Epithelial Cells is Essential to Maintain Lung Homeostasis. Front Immunol 2020; 11:583042. [PMID: 33178214 PMCID: PMC7593577 DOI: 10.3389/fimmu.2020.583042] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022] Open
Abstract
The main function of the lung is to perform gas exchange while maintaining lung homeostasis despite environmental pathogenic and non-pathogenic elements contained in inhaled air. Resident cells must keep lung homeostasis and eliminate pathogens by inducing protective immune response and silently remove innocuous particles. Which lung cell type is crucial for this function is still subject to debate, with reports favoring either alveolar macrophages (AMs) or lung epithelial cells (ECs) including airway and alveolar ECs. AMs are the main immune cells in the lung in steady-state and their function is mainly to dampen inflammatory responses. In addition, they phagocytose inhaled particles and apoptotic cells and can initiate and resolve inflammatory responses to pathogens. Although AMs release a plethora of mediators that modulate immune responses, ECs also play an essential role as they are more than just a physical barrier. They produce anti-microbial peptides and can secrete a variety of mediators that can modulate immune responses and AM functions. Furthermore, ECs can maintain AMs in a quiescent state by expressing anti-inflammatory membrane proteins such as CD200. Thus, AMs and ECs are both very important to maintain lung homeostasis and have to coordinate their action to protect the organism against infection. Thus, AMs and lung ECs communicate with each other using different mechanisms including mediators, membrane glycoproteins and their receptors, gap junction channels, and extracellular vesicles. This review will revisit characteristics and functions of AMs and lung ECs as well as different communication mechanisms these cells utilize to maintain lung immune balance and response to pathogens. A better understanding of the cross-talk between AMs and lung ECs may help develop new therapeutic strategies for lung pathogenesis.
Collapse
Affiliation(s)
- Elyse Y Bissonnette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Jean-François Lauzon-Joset
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, United States
| | - Steven F Ziegler
- Department of Immunology, Benaroya Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
64
|
Pathological effects of nano-sized particles on the respiratory system. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102242. [DOI: 10.1016/j.nano.2020.102242] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/26/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
|
65
|
Evans SE, Tseng CTK, Scott BL, Höök AM, Dickey BF. Inducible Epithelial Resistance against Coronavirus Pneumonia in Mice. Am J Respir Cell Mol Biol 2020; 63:540-541. [PMID: 32706609 PMCID: PMC7528913 DOI: 10.1165/rcmb.2020-0247le] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Scott E. Evans
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - A. Magnus Höök
- Texas A&M Institute of Biosciences and Technology, Houston, Texas
| | | |
Collapse
|
66
|
Gopallawa I, Lee RJ. Targeting the phosphoinositide-3-kinase/protein kinase B pathway in airway innate immunity. World J Biol Chem 2020; 11:30-51. [PMID: 33024516 PMCID: PMC7520643 DOI: 10.4331/wjbc.v11.i2.30] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The airway innate immune system maintains the first line of defense against respiratory infections. The airway epithelium and associated immune cells protect the respiratory system from inhaled foreign organisms. These cells sense pathogens via activation of receptors like toll-like receptors and taste family 2 receptors (T2Rs) and respond by producing antimicrobials, inflammatory cytokines, and chemokines. Coordinated regulation of fluid secretion and ciliary beating facilitates clearance of pathogens via mucociliary transport. Airway cells also secrete antimicrobial peptides and radicals to directly kill microorganisms and inactivate viruses. The phosphoinositide-3-kinase/protein kinase B (Akt) kinase pathway regulates multiple cellular targets that modulate cell survival and proliferation. Akt also regulates proteins involved in innate immune pathways. Akt phosphorylates endothelial nitric oxide synthase (eNOS) enzymes expressed in airway epithelial cells. Activation of eNOS can have anti-inflammatory, anti-bacterial, and anti-viral roles. Moreover, Akt can increase the activity of the transcription factor nuclear factor erythroid 2 related factor-2 that protects cells from oxidative stress and may limit inflammation. In this review, we summarize the recent findings of non-cancerous functions of Akt signaling in airway innate host defense mechanisms, including an overview of several known downstream targets of Akt involved in innate immunity.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Robert J Lee
- Department of Otorhinolaryngology and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
67
|
Paracellular Pathway-Mediated Mycoplasma hyopneumoniae Migration across Porcine Airway Epithelial Barrier under Air-Liquid Interface Conditions. Infect Immun 2020; 88:IAI.00470-20. [PMID: 32747599 DOI: 10.1128/iai.00470-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 02/05/2023] Open
Abstract
Mycoplasma hyopneumoniae is an important respiratory pathogen of pigs that causes persistent and secondary infections. However, the mechanisms by which this occurs are unclear. In this study, we established air-liquid interface culture systems for pig bronchial epithelial cells (ALI-PBECs) that were comparable to the conditions in the native bronchus in vivo We used this ALI-PBECs model to study the infection and migration characteristics of M. hyopneumoniae in vitro Based on the results, we confirmed that M. hyopneumoniae was able to adhere to ALI-PBECs and disrupt mucociliary function. Importantly, M. hyopneumoniae could migrate to the basolateral chamber through the paracellular route but not the transcellular pathway, and this was achieved by reversibly disrupting tight junctions (TJs) and increasing the permeability and damaging the integrity of the epithelial barrier. We examined the migration ability of M. hyopneumoniae using an ALI-PBECs model for the first time. The disruption of the epithelial barrier allowed M. hyopneumoniae to migrate to the basolateral chamber through the paracellular route, which may be related to immune evasion, extrapulmonary dissemination, and persistent infection of M. hyopneumoniae.
Collapse
|
68
|
O'Boyle N, Berry CC, Davies RL. Differentiated ovine tracheal epithelial cells support the colonisation of pathogenic and non-pathogenic strains of Mannheimia haemolytica. Sci Rep 2020; 10:14971. [PMID: 32917945 PMCID: PMC7486916 DOI: 10.1038/s41598-020-71604-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/07/2020] [Indexed: 11/15/2022] Open
Abstract
Mannheimia haemolytica is the primary bacterial species associated with respiratory disease of ruminants. A lack of cost-effective, reproducible models for the study of M. haemolytica pathogenesis has hampered efforts to better understand the molecular interactions governing disease progression. We employed a highly optimised ovine tracheal epithelial cell model to assess the colonisation of various pathogenic and non-pathogenic M. haemolytica isolates of bovine and ovine origin. Comparison of single representative pathogenic and non-pathogenic ovine isolates over ten time-points by enumeration of tissue-associated bacteria, histology, immunofluorescence microscopy and scanning electron microscopy revealed temporal differences in adhesion, proliferation, bacterial cell physiology and host cell responses. Comparison of eight isolates of bovine and ovine origin at three key time-points (2 h, 48 h and 72 h), revealed that colonisation was not strictly pathogen or serotype specific, with isolates of serotype A1, A2, A6 and A12 being capable of colonising the cell layer regardless of host species or disease status of the host. A trend towards increased proliferative capacity by pathogenic ovine isolates was observed. These results indicate that the host-specific nature of M. haemolytica infection may result at least partially from the colonisation-related processes of adhesion, invasion and proliferation at the epithelial interface.
Collapse
Affiliation(s)
- Nicky O'Boyle
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Catherine C Berry
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Robert L Davies
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
69
|
Dumm RE, Wellford SA, Moseman EA, Heaton NS. Heterogeneity of Antiviral Responses in the Upper Respiratory Tract Mediates Differential Non-lytic Clearance of Influenza Viruses. Cell Rep 2020; 32:108103. [PMID: 32877682 PMCID: PMC7462569 DOI: 10.1016/j.celrep.2020.108103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/01/2020] [Accepted: 08/11/2020] [Indexed: 01/06/2023] Open
Abstract
Influenza viruses initiate infection in the upper respiratory tract (URT), but early viral tropism and the importance of cell-type-specific antiviral responses in this tissue remain incompletely understood. By infecting transgenic lox-stop-lox reporter mice with a Cre-recombinase-expressing influenza B virus, we identify olfactory sensory neurons (OSNs) as a major viral cell target in the URT. These cells become infected, then eliminate the virus and survive in the host post-resolution of infection. OSN responses to infection are characterized by a strong induction of interferon-stimulated genes and more rapid clearance of viral protein relative to other cells in the epithelium. We speculate that this cell-type-specific response likely serves to protect the central nervous system from infection. More broadly, these results highlight the importance of evaluating antiviral responses across different cell types, even those within the same tissue, to more fully understand the mechanisms of viral disease.
Collapse
Affiliation(s)
- Rebekah E Dumm
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sebastian A Wellford
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - E Ashley Moseman
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Nicholas S Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
70
|
Cardinale F, Lombardi E, Rossi O, Bagnasco D, Bellocchi A, Menzella F. Epithelial dysfunction, respiratory infections and asthma: the importance of immunomodulation. A focus on OM-85. Expert Rev Respir Med 2020; 14:1019-1026. [PMID: 32635771 DOI: 10.1080/17476348.2020.1793673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Damage to the respiratory epithelium, is often a multifactorial phenomenon. The risk for developing a damage in respiratory epithelium and recurrent respiratory infections may vary among individuals. Preventive measures are based on strengthening the immune function, thus increasing the natural response to pathogens. Immunomodulatory agents are: i. synthetic molecules; ii. Probiotics, prebiotics, symbiotics; iii. Lysates, bacterial extracts immunomodulators: OM-85, RU 41740, D53; iv. Trace elements, vitamins. OM-85 is used for the prevention of recurrent respiratory tract infections and/or exacerbations both in adults and children, showing a good efficacy and safety profile. Its active principle, an extract of bacterial lysates isolated from 21 known respiratory pathogenic strains, shows protection against airway infections of bacterial and viral origin. AREAS COVERED This non-systematic review focuses on bacterial lysates and in particular on OM-85 and its effects on respiratory epithelium function and activity in asthma respiratory infections. Studies were selected by PubMed search of "bacterial lysate" or "OM-85" and "respiratory epithelium" or "respiratory infections", from 1993 to 2019. EXPERT OPINION Results highlight the ability of OM-85 to trigger immunomodulatory and protective immune responses against different pathogens in vivo, including influenza and respiratory syncytial virus as well bacterial superinfection following influenza.
Collapse
Affiliation(s)
- Fabio Cardinale
- University of Bari, Azienda Ospedaliero-Universitaria 'Policlinico-Giovanni XXIII', UOC di Pediatria e Pronto Soccorso , Bari, Italy
| | - Enrico Lombardi
- 'Meyer' Pediatric University Hospital, Pediatric Pumonary Unit , Firenze, Italy
| | - Oliviero Rossi
- Azienda Ospedaliero-Universitaria Careggi, UOC di Immunoallergologia , Firenze, Italy
| | - Diego Bagnasco
- Casa di Cura Villa Montallegro, Unità di Malattie Dell'apparato Respiratorio , Genova, Italy
| | - Aldo Bellocchi
- ASL ROMA 4/Dist.4, Pediatrician- Family Doctor , Roma, Italy
| | - Francesco Menzella
- Azienda USL di Reggio Emilia-IRCSS, Department of Medical Specialties, Santa Maria Nuova Hospital, Pneumology Unit , Reggio Emilia, Italy
| |
Collapse
|
71
|
Guo M, Li F, Ji J, Liu Y, Liu F, Zhao Y, Li J, Han S, Wang Q, Ding G. Inhibition of miR-93 promotes interferon effector signaling to suppress influenza A infection by upregulating JAK1. Int Immunopharmacol 2020; 86:106754. [PMID: 32652502 DOI: 10.1016/j.intimp.2020.106754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 01/07/2023]
Abstract
Type I interferons play a critical role in host defense against influenza virus infection. Interferon cascade induces the expression of interferon-stimulated genes then subsequently promotes antiviral immune responses. The microRNAs are important regulators of innate immunity, but microRNAs-mediated regulation of interferon cascade during influenza infection remains to be fully identified. Here we found influenza A virus (IAV) infection significantly inhibited miR-93 expression in alveolar epithelial type II cells through RIG-I/JNK pathway. IAV-induced downregulation of miR-93 was found to upregulate JAK1, the target of miR-93, and then feedback promote antiviral innate response by facilitating IFN effector signaling. Importantly, in vivo administration of miR-93 antagomiR markedly suppressed IAV infection, protecting mice form IAVs -associated death. Hence, the inducible downregulation of miR-93 feedback suppress IAV infection by strengthening IFN-JAK-STAT pathway via JAK1 upregulation, and in vivo inhibition of miR-93 bears considerable therapeutic potential for suppressing IAV infection.
Collapse
Affiliation(s)
- Meng Guo
- National Key Laboratory of Medical Immunology &Institute of Immunology, Navy Medical University, Shanghai, China; Institute of Organ Transplantation, Changzhen Hospital, Navy Medical University, Shanghai, China
| | - Fangbing Li
- National Key Laboratory of Medical Immunology &Institute of Immunology, Navy Medical University, Shanghai, China
| | - Junsong Ji
- Institute of Organ Transplantation, Changzhen Hospital, Navy Medical University, Shanghai, China
| | - Yanfang Liu
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fang Liu
- The Medical Teaching Laboratory, the College of Basic Medical Sciences, Navy Medical University, Shanghai, China
| | - Yuanyu Zhao
- Institute of Organ Transplantation, Changzhen Hospital, Navy Medical University, Shanghai, China
| | - Junhui Li
- National Key Laboratory of Medical Immunology &Institute of Immunology, Navy Medical University, Shanghai, China
| | - Shu Han
- Institute of Organ Transplantation, Changzhen Hospital, Navy Medical University, Shanghai, China.
| | - Quanxing Wang
- National Key Laboratory of Medical Immunology &Institute of Immunology, Navy Medical University, Shanghai, China.
| | - Guoshan Ding
- Institute of Organ Transplantation, Changzhen Hospital, Navy Medical University, Shanghai, China.
| |
Collapse
|
72
|
Suárez-Cuartín G, Sibila O. Inflamación local y sistémica en bronquiectasias. Endotipos y biomarcadores. OPEN RESPIRATORY ARCHIVES 2020. [DOI: 10.1016/j.opresp.2020.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
73
|
Kim SB, Lee WY, Lee JH, Lee SJ, Lee MK, Kim SH, Uh Y, Jung SH, Shin B. A variety of bacterial aetiologies in the lower respiratory tract at patients with endobronchial tuberculosis. PLoS One 2020; 15:e0234558. [PMID: 32584852 PMCID: PMC7316277 DOI: 10.1371/journal.pone.0234558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/27/2020] [Indexed: 12/31/2022] Open
Abstract
Recently, our understanding of the elusive bacterial communities in the lower respiratory tract and their role in chronic lung disease has increased significantly. However, little is known about the respiratory microorganisms in patients with endobronchial tuberculosis (EBTB), which is a chronic inflammatory disease characterized by destruction of the tracheobronchial tree due to Mycobacterium tuberculosis (MTB) infection. We retrospectively reviewed data for histopathologically and microbiologically confirmed EBTB patients diagnosed at a tertiary referral hospital in South Korea between January 2013 and January 2019. Bacterial cultures were performed on bronchial washing from these patients at the time of EBTB diagnosis. A total of 216 patients with EBTB were included in the study. The median age was 73 years and 142 (65.7%) patients were female. Bacteria were detected in 42 (19.4%) patients. Additionally, bacterial co-infection was present in 6 (2.8%) patients. Apart from MTB, the most common microorganisms identified were Staphylococcus aureus (n = 14, 33.3%) followed by Klebsiella species (n = 12, 28.6%; 10 Klebsiella pneumoniae, 2 Klebsiella oxytoca), Streptococcus species (n = 5, 11.9%), Enterobacter species (n = 4, 9.5%), and Pseudomonas aeruginosa (n = 3, 7.1%). A variety of microorganisms were isolated from the bronchial washing indicating that changes in microorganism composition occur in the airways of patients with EBTB. Further studies are needed to investigate the clinical significance of this finding.
Collapse
Affiliation(s)
- Sae Byol Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Won-Yeon Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Ji-Ho Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Seok Jeong Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Myoung Kyu Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Sang-Ha Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Young Uh
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Soon-Hee Jung
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Beomsu Shin
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- * E-mail:
| |
Collapse
|
74
|
Host-Pathogen Responses to Pandemic Influenza H1N1pdm09 in a Human Respiratory Airway Model. Viruses 2020; 12:v12060679. [PMID: 32599823 PMCID: PMC7354428 DOI: 10.3390/v12060679] [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: 05/20/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
The respiratory Influenza A Viruses (IAVs) and emerging zoonotic viruses such as Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) pose a significant threat to human health. To accelerate our understanding of the host–pathogen response to respiratory viruses, the use of more complex in vitro systems such as normal human bronchial epithelial (NHBE) cell culture models has gained prominence as an alternative to animal models. NHBE cells were differentiated under air-liquid interface (ALI) conditions to form an in vitro pseudostratified epithelium. The responses of well-differentiated (wd) NHBE cells were examined following infection with the 2009 pandemic Influenza A/H1N1pdm09 strain or following challenge with the dsRNA mimic, poly(I:C). At 30 h postinfection with H1N1pdm09, the integrity of the airway epithelium was severely impaired and apical junction complex damage was exhibited by the disassembly of zona occludens-1 (ZO-1) from the cell cytoskeleton. wdNHBE cells produced an innate immune response to IAV-infection with increased transcription of pro- and anti-inflammatory cytokines and chemokines and the antiviral viperin but reduced expression of the mucin-encoding MUC5B, which may impair mucociliary clearance. Poly(I:C) produced similar responses to IAV, with the exception of MUC5B expression which was more than 3-fold higher than for control cells. This study demonstrates that wdNHBE cells are an appropriate ex-vivo model system to investigate the pathogenesis of respiratory viruses.
Collapse
|
75
|
Sveiven SN, Nordgren TM. Lung-resident mesenchymal stromal cells are tissue-specific regulators of lung homeostasis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L197-L210. [PMID: 32401672 DOI: 10.1152/ajplung.00049.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Until recently, data supporting the tissue-resident status of mesenchymal stromal cells (MSC) has been ambiguous since their discovery in the 1950-60s. These progenitor cells were first discovered as bone marrow-derived adult multipotent cells and believed to migrate to sites of injury, opposing the notion that they are residents of all tissue types. In recent years, however, it has been demonstrated that MSC can be found in all tissues and MSC from different tissues represent distinct populations with differential protein expression unique to each tissue type. Importantly, these cells are efficient mediators of tissue repair, regeneration, and prove to be targets for therapeutics, demonstrated by clinical trials (phase 1-4) for MSC-derived therapies for diseases like graft-versus-host-disease, multiple sclerosis, rheumatoid arthritis, and Crohn's disease. The tissue-resident status of MSC found in the lung is a key feature of their importance in the context of disease and injuries of the respiratory system, since these cells could be instrumental to providing more specific and targeted therapies. Currently, bone marrow-derived MSC have been established in the treatment of disease, including diseases of the lung. However, with lung-resident MSC representing a unique population with a different phenotypic and gene expression pattern than MSC derived from other tissues, their role in remediating lung inflammation and injury could provide enhanced efficacy over bone marrow-derived MSC methods. Through this review, lung-resident MSC will be characterized, using previously published data, by surface markers, gene expression patterns, and compared with bone-marrow MSC to highlight similarities and, importantly, differences in these cell types.
Collapse
Affiliation(s)
- Stefanie Noel Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| |
Collapse
|
76
|
Chung YW, Cha J, Han S, Chen Y, Gucek M, Cho HJ, Nakahira K, Choi AMK, Ryu JH, Yoon JH. Apolipoprotein E and Periostin Are Potential Biomarkers of Nasal Mucosal Inflammation. A Parallel Approach of In Vitro and In Vivo Secretomes. Am J Respir Cell Mol Biol 2020; 62:23-34. [PMID: 31194918 DOI: 10.1165/rcmb.2018-0248oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
No previously suggested biomarkers of nasal mucosal inflammation have been practically applied in clinical fields, and nasal epithelium-derived secreted proteins as biomarkers have not specifically been investigated. The goal of this study was to identify secreted proteins that dynamically change during the differentiation from basal cells to fully differentiated cells and examine whether nasal epithelium-derived proteins can be used as biomarkers of nasal mucosal inflammation, such as chronic rhinosinusitis. To achieve this goal, we analyzed two secretomes using the isobaric tag for relative and absolute quantification technique. From in vitro secretomes, we identified the proteins altered in apical secretions of primary human nasal epithelial cells according to the degree of differentiation; from in vivo secretomes, we identified the increased proteins in nasal lavage fluids obtained from patients 2 weeks after endoscopic sinus surgery for chronic sinusitis. We then used a parallel approach to identify specific biomarkers of nasal mucosal inflammation; first, we selected apolipoprotein E as a nasal epithelial cell-derived biomarker through screening proteins that were upregulated in both in vitro and in vivo secretomes, and verified highly secreted apolipoprotein E in nasal lavage fluids of the patients by Western blotting. Next, we selected periostin as an inflammatory mediator-inducible biomarker from in vivo secretomes, the secretion of which was not induced under in vitro culture conditions. We demonstrated that those two nasal epithelium-derived proteins are possible biomarkers of nasal mucosal inflammation.
Collapse
Affiliation(s)
- Youn Wook Chung
- The Airway Mucus Institute.,Global Research Laboratory for Allergic Airway Disease.,Severance Biomedical Science Institute
| | - Jimin Cha
- Severance Biomedical Science Institute.,Brain Korea 21 PLUS Project for Medical Science, and
| | - Seunghan Han
- Severance Biomedical Science Institute.,Brain Korea 21 PLUS Project for Medical Science, and
| | - Yong Chen
- Proteomics Core Facility, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Marjan Gucek
- Proteomics Core Facility, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Hyung-Ju Cho
- The Airway Mucus Institute.,Global Research Laboratory for Allergic Airway Disease.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Ji-Hwan Ryu
- Severance Biomedical Science Institute.,Brain Korea 21 PLUS Project for Medical Science, and
| | - Joo-Heon Yoon
- The Airway Mucus Institute.,Global Research Laboratory for Allergic Airway Disease.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
77
|
Tu Y, Wu X, Yu F, Dang J, Wei Y, Yu H, Liao W, Zhang Y, Wang J. Tristetraprolin-RNA interaction map reveals a novel TTP-RelB regulatory network for innate immunity gene expression. Mol Immunol 2020; 121:59-71. [DOI: 10.1016/j.molimm.2020.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/03/2020] [Accepted: 02/07/2020] [Indexed: 02/03/2023]
|
78
|
Himly M, Geppert M, Hofer S, Hofstätter N, Horejs-Höck J, Duschl A. When Would Immunologists Consider a Nanomaterial to be Safe? Recommendations for Planning Studies on Nanosafety. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907483. [PMID: 32239645 DOI: 10.1002/smll.201907483] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 06/11/2023]
Abstract
The immune system is professional in recognizing and responding to non-self, including nanomaterials. Immune responses by professional and nonprofessional immune cells are thus nearly inevitable upon exposure of cells and organisms to such materials. The state of research into taking the immune system into account in nanosafety studies is reviewed and three aspects in which further improvements are desirable are identified: 1) Due to technical limitations, more stringent testing for endotoxin contamination should be made. 2) Since under overdose conditions immunity shows unphysiological responses, all doses used should be justified by being equivalent to tissue-delivered doses. 3) When markers of acute inflammation or cell stress are observed, functional assays are necessary to distinguish between homeostatic fluctuation and genuine defensive or tolerogenic responses. Since immune activation can also indicate that the immune system considers a stimulus to be harmless and induces tolerance, activation markers by themselves do not necessarily imply a danger to the body. Guidelines such as these are necessary to approach the point where specific nanomaterials are classified as safe based on reliable testing strategies.
Collapse
Affiliation(s)
- Martin Himly
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Mark Geppert
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Sabine Hofer
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Norbert Hofstätter
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Jutta Horejs-Höck
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Albert Duschl
- Department for Biosciences & Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| |
Collapse
|
79
|
Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM, Wegmann M. More Than Just a Barrier: The Immune Functions of the Airway Epithelium in Asthma Pathogenesis. Front Immunol 2020; 11:761. [PMID: 32411147 PMCID: PMC7198799 DOI: 10.3389/fimmu.2020.00761] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
Allergic bronchial asthma is a chronic disease of the airways that is characterized by symptoms like respiratory distress, chest tightness, wheezing, productive cough, and acute episodes of broncho-obstruction. This symptom-complex arises on the basis of chronic allergic inflammation of the airway wall. Consequently, the airway epithelium is central to the pathogenesis of this disease, because its multiple abilities directly have an impact on the inflammatory response and thus the formation of the disease. In turn, its structure and functions are markedly impaired by the inflammation. Hence, the airway epithelium represents a sealed, self-cleaning barrier, that prohibits penetration of inhaled allergens, pathogens, and other noxious agents into the body. This barrier is covered with mucus that further contains antimicrobial peptides and antibodies that are either produced or specifically transported by the airway epithelium in order to trap these particles and to remove them from the body by a process called mucociliary clearance. Once this first line of defense of the lung is overcome, airway epithelial cells are the first cells to get in contact with pathogens, to be damaged or infected. Therefore, these cells release a plethora of chemokines and cytokines that not only induce an acute inflammatory reaction but also have an impact on the alignment of the following immune reaction. In case of asthma, all these functions are impaired by the already existing allergic immune response that per se weakens the barrier integrity and self-cleaning abilities of the airway epithelium making it more vulnerable to penetration of allergens as well as of infection by bacteria and viruses. Recent studies indicate that the history of allergy- and pathogen-derived insults can leave some kind of memory in these cells that can be described as imprinting or trained immunity. Thus, the airway epithelium is in the center of processes that lead to formation, progression and acute exacerbation of asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, Borstel, Germany.,Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
| | - Lars P Lunding
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| | - Johanna C Ehlers
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Experimental Pneumology, Research Center Borstel, Borstel, Germany
| | - Markus Weckmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Department of Pediatric Pulmonology and Allergology, University Children's Hospital, Lübeck, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany.,Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Michael Wegmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| |
Collapse
|
80
|
van Riet S, van Schadewijk A, de Vos S, Vandeghinste N, Rottier RJ, Stolk J, Hiemstra PS, Khedoe P. Modulation of Airway Epithelial Innate Immunity and Wound Repair by M(GM-CSF) and M(M-CSF) Macrophages. J Innate Immun 2020; 12:410-421. [PMID: 32289801 DOI: 10.1159/000506833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/26/2020] [Indexed: 12/25/2022] Open
Abstract
Airway epithelial cells and macrophages participate in inflammatory responses to external noxious stimuli, which can cause epithelial injury. Upon injury, epithelial cells and macrophages act in concert to ensure rapid restoration of epithelial integrity. The nature of the interactions between these cell types during epithelial repair is incompletely understood. We used an in vitro human coculture model of primary bronchial epithelial cells cultured at the air-liquid interface (ALI-PBEC) and polarized primary monocyte-derived macrophages. Using this coculture, we studied the contribution of macrophages to epithelial innate immunity, wound healing capacity, and epithelial exposure to whole cigarette smoke (WCS). Coculture of ALI-PBEC with lipopolysaccharide (LPS)-activated M(GM-CSF) macrophages increased the expression of DEFB4A, CXCL8, and IL6 at 24 h in the ALI-PBEC, whereas LPS-activated M(M-CSF) macrophages only increased epithelial IL6 expression. Furthermore, wound repair was accelerated by coculture with both activated M(GM-CSF) and M(M-CSF) macrophages, also following WCS exposure. Coculture of ALI-PBEC and M(GM-CSF) macrophages resulted in increased CAMP expression in M(GM-CSF) macrophages, which was absent in M(M-CSF) macrophages. CAMP encodes LL-37, an antimicrobial peptide with immune-modulating and repair-enhancing activities. In conclusion, dynamic crosstalk between ALI-PBEC and macrophages enhances epithelial innate immunity and wound repair, even upon concomitant cigarette smoke exposure.
Collapse
Affiliation(s)
- Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands,
| | | | | | | | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Padmini Khedoe
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
81
|
IRF7 Is Required for the Second Phase Interferon Induction during Influenza Virus Infection in Human Lung Epithelia. Viruses 2020; 12:v12040377. [PMID: 32235406 PMCID: PMC7232147 DOI: 10.3390/v12040377] [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: 01/10/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 11/17/2022] Open
Abstract
Influenza A virus (IAV) infection is a major cause of morbidity and mortality. Retinoic acid-inducible protein I (RIG-I) plays an important role in the recognition of IAV in most cell types, and leads to the activation of interferon (IFN). We investigated mechanisms of RIG-I and IFN induction by IAV in the BCi-NS1.1 immortalized human airway basal cell line and in the A549 human alveolar epithelial cell line. We found that the basal expression levels of RIG-I and regulatory transcription factor (IRF) 7 were very low in BCi-NS1.1 cells. IAV infection induced robust RIG-I and IRF7, not IRF3, expression. siRNA against IRF7 and mitochondrial antiviral-signaling protein (MAVS), but not IRF3, significantly inhibited RIG-I mRNA expression and IFN induction by IAV infection. Most importantly, even without virus infection, IFN-β alone induced RIG-I, and siRNA against IRF7 did not inhibit RIG-I induction by IFN-β. Similar results were found in the alveolar basal epithelial A549 cell line. RIG-I and IRF7 expression in humans is highly inducible and greatly amplified by IFN produced from virus infected cells. IFN induction can be separated into two phases, that initially induced by the virus with basal RIG-I (the first phase), and that induced by the subsequent virus with amplified RIG-I from the first phase IFN (the second phase). The de novo synthesis of IRF7 is required for the second phase IFN induction during influenza virus infection in human lung bronchial and alveolar epithelial cells.
Collapse
|
82
|
den Hartog G, Schijf MA, Berbers GAM, van der Klis FRM, Buisman AM. Bordetella pertussis induces IFN-γ production by NK cells resulting in chemo-attraction by respiratory epithelial cells. J Infect Dis 2020; 225:1248-1260. [PMID: 32219323 PMCID: PMC8974844 DOI: 10.1093/infdis/jiaa140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/24/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Whooping cough is caused by infection of the airways with Bordetella pertussis (Bp). As IFN-γ is essential for protective immunity against Bp we investigated how IFN-γ is induced by Bp or the virulence antigens FHA, Prn or PT, and how IFN-γ contributes to local immune responses in humans. METHODS PBMCs from healthy donors and/or respiratory epithelial cells were stimulated with soluble antigens or inactivated intact Bp and the presence or absence of blocking antibodies or chemokines. Supernatants and cells were analyzed for IFN-γ and chemokine production and lymphocyte migration tested using epithelial supernatants. RESULTS The soluble antigens failed to induce IFN-γ production, whereas inactivated Bp induced IFN-γ production. NK cells were the main source of IFN-γ production, which was enhanced by IL-15. Epithelial-PBMC co-cultures showed robust IFN-γ-dependent CXCL9 and CXCL10 production by the epithelial cells following stimulation with IFN-γ and Bp. The epithelial-derived chemokines resulted in CXCR3-dependent recruitment of NK and T cells. CONCLUSIONS Inactivated Bp, but not antigens, induced potent IFN-γ production by NK cells, resulting in chemo-attraction of lymphocytes towards the respiratory epithelium. These data provide insight into the requirements for IFN-γ production and how IFN-γ enhances local immune responses to prevent Bp-mediated disease.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Immunology of Infectious Diseases and Vaccination, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Marcel A Schijf
- Department of Immunology of Infectious Diseases and Vaccination, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Guy A M Berbers
- Department of Immunology of Infectious Diseases and Vaccination, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Fiona R M van der Klis
- Department of Immunology of Infectious Diseases and Vaccination, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Anne-Marie Buisman
- Department of Immunology of Infectious Diseases and Vaccination, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
83
|
Elias-Kirma S, Artzy-Schnirman A, Das P, Heller-Algazi M, Korin N, Sznitman J. In situ-Like Aerosol Inhalation Exposure for Cytotoxicity Assessment Using Airway-on-Chips Platforms. Front Bioeng Biotechnol 2020; 8:91. [PMID: 32154228 PMCID: PMC7044134 DOI: 10.3389/fbioe.2020.00091] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/31/2020] [Indexed: 12/02/2022] Open
Abstract
Lung exposure to inhaled particulate matter (PM) is known to injure the airway epithelium via inflammation, a phenomenon linked to increased levels of global morbidity and mortality. To evaluate physiological outcomes following PM exposure and concurrently circumvent the use of animal experiments, in vitro approaches have typically relied on traditional assays with plates or well inserts. Yet, these manifest drawbacks including the inability to capture physiological inhalation conditions and aerosol deposition characteristics relative to in vivo human conditions. Here, we present a novel airway-on-chip exposure platform that emulates the epithelium of human bronchial airways with critical cellular barrier functions at an air-liquid interface (ALI). As a proof-of-concept for in vitro lung cytotoxicity testing, we recapitulate a well-characterized cell apoptosis pathway, induced through exposure to 2 μm airborne particles coated with αVR1 antibody that leads to significant loss in cell viability across the recapitulated airway epithelium. Notably, our in vitro inhalation assays enable simultaneous aerosol exposure across multiple airway chips integrated within a larger bronchial airway tree model, under physiological respiratory airflow conditions. Our findings underscore in situ-like aerosol deposition outcomes where patterns depend on respiratory flows across the airway tree geometry and gravitational orientation, as corroborated by concurrent numerical simulations. Our airway-on-chips not only highlight the prospect of realistic in vitro exposure assays in recapitulating characteristic local in vivo deposition outcomes, such platforms open opportunities toward advanced in vitro exposure assays for preclinical cytotoxicity and drug screening applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Josué Sznitman
- Department of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
84
|
Louhaichi S, Mlika M, Hamdi B, Hamzaoui K, Hamzaoui A. Sputum IL-26 Is Overexpressed in Severe Asthma and Induces Proinflammatory Cytokine Production and Th17 Cell Generation: A Case-Control Study of Women. J Asthma Allergy 2020; 13:95-107. [PMID: 32099415 PMCID: PMC7006858 DOI: 10.2147/jaa.s229522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Asthma inflammation is a complex pathway involving numerous mediators. Interleukin-26 (IL-26), a member of the IL-10 cytokine family, is abundant in human airways and induces the production of proinflammatory cytokines. Our aim was to investigate the possible role of IL-26 in severe asthma. We analysed the expression of IL-26 in severe asthma both in peripheral blood and induced sputum. Patients and Methods A total of 50 adult women with severe asthma were recruited and compared to 30 healthy controls (HC). Serum and sputum fluid (SF) levels of IL-26 and IL-17 were defined by ELISA. IL-26 mRNA expression and IL-26 protein were analysed using RT-PCR and Western blot. In vitro, we studied the effect of recombinant IL-26 (rIL-26) and SF-IL-26 on cultured CD4+ T cells and monocytes, comparing patients and controls. Results Concentrations of IL-26 are higher in serum and induced sputum of asthmatic patients than in HC. Moreover, IL-26 protein and mRNA expression were significantly elevated in asthma sputum cells compared to PBMCs. We observed a positive correlation between body mass index (BMI) and sputum fluid IL-26, while the correlation between IL-26 and lung function tests (FEV1% and FEV1/FVC ratio) was negative. IL-17A was highly expressed in SF and correlated positively with IL-26. In patients’ sputum IL-26 and IL-17A were significantly associated with neutrophils. Stimulation of cultured CD4+ T cells with monocytes by recombinant IL-26 promoted the generation of RORγt+ Th17+ cells inducing the production of IL-17A, IL-1β, IL-6 and TNF-α cytokines. IL-26 expressed in SF was biologically active and induced IL-17 secretion in the presence of IL-1β and IL-6 cytokines. Conclusion These findings show that IL-26 is highly produced in asthmatic sputum, induces pro-inflammatory cytokine secretion by monocytes/macrophages, and favours Th17 cell generation. IL-26 thereby appears as a novel pro-inflammatory cytokine, produced locally in the airways that may constitute a promising target to treat asthma inflammatory process.
Collapse
Affiliation(s)
- Sabrine Louhaichi
- Research Laboratory 19SP02 "Chronic Pulmonary Pathologies: From Genome to Management", Abderrahman Mami Hospital, Ariana, Tunisia.,Medicine Faculty of Tunis, Department of Basic Sciences, Tunis El Manar University, Tunis, Tunisia.,Department of Paediatric and Respiratory Diseases, Abderrahman Mami Hospital, Pavillon B, Ariana, Tunisia
| | - Mona Mlika
- Medicine Faculty of Tunis, Department of Basic Sciences, Tunis El Manar University, Tunis, Tunisia.,Pathology Department, Abderrahman Mami Hospital, Ariana, Tunisia
| | - Besma Hamdi
- Research Laboratory 19SP02 "Chronic Pulmonary Pathologies: From Genome to Management", Abderrahman Mami Hospital, Ariana, Tunisia.,Medicine Faculty of Tunis, Department of Basic Sciences, Tunis El Manar University, Tunis, Tunisia.,Department of Paediatric and Respiratory Diseases, Abderrahman Mami Hospital, Pavillon B, Ariana, Tunisia
| | - Kamel Hamzaoui
- Research Laboratory 19SP02 "Chronic Pulmonary Pathologies: From Genome to Management", Abderrahman Mami Hospital, Ariana, Tunisia.,Medicine Faculty of Tunis, Department of Basic Sciences, Tunis El Manar University, Tunis, Tunisia
| | - Agnès Hamzaoui
- Research Laboratory 19SP02 "Chronic Pulmonary Pathologies: From Genome to Management", Abderrahman Mami Hospital, Ariana, Tunisia.,Medicine Faculty of Tunis, Department of Basic Sciences, Tunis El Manar University, Tunis, Tunisia.,Department of Paediatric and Respiratory Diseases, Abderrahman Mami Hospital, Pavillon B, Ariana, Tunisia
| |
Collapse
|
85
|
Tyagi N, Singh DK, Dash D, Singh R. Curcumin Modulates Paraquat-Induced Epithelial to Mesenchymal Transition by Regulating Transforming Growth Factor-β (TGF-β) in A549 Cells. Inflammation 2020; 42:1441-1455. [PMID: 31028577 DOI: 10.1007/s10753-019-01006-0] [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] [Indexed: 02/04/2023]
Abstract
Paraquat (PQ), a widely used potent herbicide, generates superoxide anions and other free radicals, leading to severe toxicity and acute lung injury. PQ induces pulmonary fibrosis through epithelial to mesenchymal transition (EMT) characterized by increased number of myofibroblasts. Time-dependent PQ-induced EMT has been evaluated in present investigation where intracellular ROS levels were significantly enhanced after 24 h of PQ intoxication. Anti-inflammatory effects of curcumin have been studied where alveolar epithelial cells (A549 cells) were incubated with curcumin (30 μΜ) for 1 and 3 h before PQ intoxication (700 μM). Western blot and immunocytochemistry studies revealed that pretreatment of A549 cells with curcumin for 3 h before PQ exposure has maintained E-cadherin expression and inhibited PQ induced α-smooth-muscle actin (α-SMA) expression. Transforming growth factor-β (TGF-β) that seems to be involved in PQ-induced EMT was enhanced after PQ intoxication, but curcumin pretreatment has effectively inhibited its expression. Immunostaining studies have shown that curcumin pretreatment has significantly reduced matrix metalloproteinase-9 (MMP-9) expressions, which were elevated after PQ intoxication. These results demonstrate that curcumin can regulate PQ-induced EMT by regulating the expression of TGF-β.
Collapse
Affiliation(s)
- Namitosh Tyagi
- Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India
| | - D K Singh
- Department of Physics, Udai Pratap Autonomous College, Varanasi, 221002, India
| | - D Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Rashmi Singh
- Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
86
|
Gohy S, Hupin C, Ladjemi MZ, Hox V, Pilette C. Key role of the epithelium in chronic upper airways diseases. Clin Exp Allergy 2019; 50:135-146. [PMID: 31746062 DOI: 10.1111/cea.13539] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022]
Abstract
The respiratory epithelium of the upper airways is a first-line defence against inhaled irritants, pathogens and allergens. It ensures a physical barrier provided by apical junctions and mucociliary clearance to avoid excessive activation of the immune system. The epithelium also forms a chemical and immunological barrier, extensively equipped to protect the airways against external aggressions before the adaptive immune system is required. Under normal circumstances, the epithelium is capable of recovering rapidly after damage. This manuscript reviews these main properties of the upper airway epithelium as well as its reported impairments in chronic inflammatory diseases. The knowledge on normal epithelial functions and their dysregulation in upper airway diseases should help to design new epithelial-targeted treatments.
Collapse
Affiliation(s)
- Sophie Gohy
- Pole of Pneumology, ENT and Dermatology, Université catholique de Louvain (UCL), Brussels, Belgium.,Department of Pneumology, Cliniques universitaires, Brussels, Belgium
| | - Cloé Hupin
- Pole of Pneumology, ENT and Dermatology, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Maha Zohra Ladjemi
- Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Hox
- Department of Otorhinolaryngology, Cliniques universitaires, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT and Dermatology, Université catholique de Louvain (UCL), Brussels, Belgium.,Department of Pneumology, Cliniques universitaires, Brussels, Belgium
| |
Collapse
|
87
|
Tatsuta M, Kan-O K, Ishii Y, Yamamoto N, Ogawa T, Fukuyama S, Ogawa A, Fujita A, Nakanishi Y, Matsumoto K. Effects of cigarette smoke on barrier function and tight junction proteins in the bronchial epithelium: protective role of cathelicidin LL-37. Respir Res 2019; 20:251. [PMID: 31706310 PMCID: PMC6842552 DOI: 10.1186/s12931-019-1226-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/31/2019] [Indexed: 11/10/2022] Open
Abstract
Background Airway epithelial barrier function is maintained by the formation of tight junctions (TJs) and adherens junctions (AJs). Inhalation of cigarette smoke causes airway epithelial barrier dysfunction and may contribute to the pathogenesis of chronic lung diseases such as asthma and chronic obstructive pulmonary disease (COPD). We assessed the effects of cigarette smoke on barrier function and expression of multiple TJ and AJ proteins in the bronchial epithelium. We also examined whether treatment with glucocorticosteroids (GCSs), long-acting β2-agonists (LABAs), and human cathelicidin LL-37 can protect against cigarette smoke extract (CSE)-induced barrier dysfunction. Methods Calu-3 cells cultured at the air-liquid interface were pretreated with or without GCSs, LABAs, GCSs plus LABAs, or LL-37, and subsequently exposed to CSE. Barrier function was assessed by transepithelial electronic resistance (TEER) measurements. Gene and protein expression levels of TJ and AJ proteins were analyzed by quantitative PCR and western blotting, respectively. Immunofluorescence staining of TJ and AJ proteins was performed. Results CSE decreased TEER and increased permeability in a concentration-dependent manner. CSE suppressed gene expression of claudin-1, claudin-3, claudin-4, claudin-7, claudin-15, occludin, E-cadherin, junctional adhesion molecule-A (JAM-A) and zonula occludens-1 (ZO-1) within 12 h post-CSE exposure, while suppressed protein expression levels of occludin at 12 h. CSE-treated cells exhibited discontinuous or attenuated immunostaining for claudin-1, claudin-3, claudin-4, occludin, ZO-1, and E-cadherin compared with untreated cells. GCS treatment partially restored CSE-induced TEER reduction, while LABA treatment had no effect. GCS and LABA combination treatment had no additive effect on CSE-induced TEER reduction and gene suppression of TJ and AJ proteins. Human cathelicidin LL-37 counteracted CSE-induced TEER reduction and prevented disruption of occludin and ZO-1. LL-37 also attenuated CSE-induced decreases in gene and protein expression levels of occludin. Conclusions CSE caused airway epithelial barrier dysfunction and simultaneously downregulated multiple TJ and AJ proteins. GCS and LABA combination treatment had no additive effect on CSE-induced TEER reduction. LL-37 counteracted CSE-induced TEER reduction and prevented disruption of occludin and ZO-1. Use of LL-37 to counteract airway epithelial barrier dysfunction may have significant benefits for respiratory diseases such as asthma and COPD.
Collapse
Affiliation(s)
- Miyoko Tatsuta
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Respiratory Medicine, National Hospital Organization Omuta National Hospital, Fukuoka, 837-0911, Japan
| | - Keiko Kan-O
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Endoscopic Diagnostics and Therapeutics, Kyushu University Hospital, Fukuoka, 812-8582, Japan.
| | - Yumiko Ishii
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Norio Yamamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Ogawa
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoru Fukuyama
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Aimi Ogawa
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akitaka Fujita
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
88
|
Sadikot RT, Bedi B, Li J, Yeligar SM. Alcohol-induced mitochondrial DNA damage promotes injurious crosstalk between alveolar epithelial cells and alveolar macrophages. Alcohol 2019; 80:65-72. [PMID: 31307864 DOI: 10.1016/j.alcohol.2018.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/20/2022]
Abstract
Excessive alcohol users have a higher risk for developing respiratory infections compared to individuals who do not chronically misuse alcohol, due to impaired host immune defense. In the lung, alveolar epithelial cells play a critical role in host immune defense against invading pathogens in the lower respiratory tract due to their capacity to maintain barrier integrity, and alveolar macrophages play a key role in pulmonary innate immunity by phagocytizing and clearing infiltrating microbes. Chronic alcohol misuse induces mitochondrial damage that results in release of mitochondrial DNA (mtDNA) in exosomes. We hypothesized that alcohol-induced cellular damage leads to release of exosomes containing damaged mtDNA, which can mediate injurious crosstalk between lung epithelial cells and macrophages. The mouse alveolar epithelial cell line, MLE-12, and the mouse alveolar macrophage cell line, MH-S, were transfected with a damaged mtDNA overexpression plasmid or exposed to ethanol in vitro. Overexpression of damaged mtDNA impaired MLE-12 barrier function and MH-S phagocytic capacity. Ethanol induced damage of mtDNA in MLE-12 and MH-S cells, and promoted release of exosomes enriched with damaged mtDNA from these cells. Exosomes from ethanol-exposed MH-S cells caused mtDNA damage and barrier dysfunction in MLE-12 cells, and exosomes from ethanol-exposed MLE-12 cells caused mtDNA damage and phagocytic dysfunction in MH-S cells. Collectively, these data show that ethanol-induced mtDNA damage in MLE-12 and MH-S cells stimulates release of damaged mtDNA-enriched exosomes and contributes to injurious crosstalk between the alveolar epithelium and macrophages, potentially leading to impaired host immune defense against respiratory infections.
Collapse
Affiliation(s)
- Ruxana T Sadikot
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, United States; Emory University, Atlanta, GA, 30322, United States
| | - Brahmchetna Bedi
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, United States; Emory University, Atlanta, GA, 30322, United States
| | - Juan Li
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, United States; Emory University, Atlanta, GA, 30322, United States
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, United States; Emory University, Atlanta, GA, 30322, United States.
| |
Collapse
|
89
|
Rynning I, Neca J, Vrbova K, Libalova H, Rossner P, Holme JA, Gützkow KB, Afanou AKJ, Arnoldussen YJ, Hruba E, Skare Ø, Haugen A, Topinka J, Machala M, Mollerup S. In Vitro Transformation of Human Bronchial Epithelial Cells by Diesel Exhaust Particles: Gene Expression Profiling and Early Toxic Responses. Toxicol Sci 2019; 166:51-64. [PMID: 30010986 PMCID: PMC6204768 DOI: 10.1093/toxsci/kfy183] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Occupational exposure to diesel exhaust may cause lung cancer in humans. Mechanisms include DNA-damage and inflammatory responses. Here, the potential of NIST SRM2975 diesel exhaust particles (DEP) to transform human bronchial epithelial cells (HBEC3) in vitro was investigated. Long-term exposure of HBEC3 to DEP led to increased colony growth in soft agar. Several DEP-transformed cell lines were established and based on the expression of epithelial-to-mesenchymal-transition (EMT) marker genes, one of them (T2-HBEC3) was further characterized. T2-HBEC3 showed a mesenchymal/fibroblast-like morphology, reduced expression of CDH1, and induction of CDH2 and VIM. T2-HBEC3 had reduced migration potential compared with HBEC3 and little invasion capacity. Gene expression profiling showed baseline differences between HBEC3 and T2-HBEC3 linked to lung carcinogenesis. Next, to assess differences in sensitivity to DEP between parental HBEC3 and T2-HBEC3, gene expression profiling was carried out after DEP short-term exposure. Results revealed changes in genes involved in metabolism of xenobiotics and lipids, as well as inflammation. HBEC3 displayed a higher steady state of IL1B gene expression and release of IL-1β compared with T2-HBEC3. HBEC3 and T2-HBEC3 showed similar susceptibility towards DEP-induced genotoxic effects. Liquid-chromatography-tandem-mass-spectrometry was used to measure secretion of eicosanoids. Generally, major prostaglandin species were released in higher concentrations from T2-HBEC3 than from HBEC3 and several analytes were altered after DEP-exposure. In conclusion, long-term exposure to DEP-transformed human bronchial epithelial cells in vitro. Differences between HBEC3 and T2-HBEC3 regarding baseline levels and DEP-induced changes of particularly CYP1A1, IL-1β, PGE2, and PGF2α may have implications for acute inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Iselin Rynning
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, N-0304 Oslo, Norway
| | - Jiri Neca
- Department of Chemistry and Toxicology, Veterinary Research Institute, 621 00 Brno, Czech Republic
| | - Kristyna Vrbova
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Helena Libalova
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Pavel Rossner
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Jørn A Holme
- Division of Infection Control, Environment and Health, Department of Air and Noise
| | - Kristine B Gützkow
- Division of Infection Control, Department of Molecular Biology, Norwegian Institute of Public Health, N-0304 Oslo, Norway
| | - Anani K Johnny Afanou
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, N-0304 Oslo, Norway
| | - Yke J Arnoldussen
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, N-0304 Oslo, Norway
| | - Eva Hruba
- Department of Chemistry and Toxicology, Veterinary Research Institute, 621 00 Brno, Czech Republic
| | - Øivind Skare
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, N-0304 Oslo, Norway
| | - Aage Haugen
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, N-0304 Oslo, Norway
| | - Jan Topinka
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, 621 00 Brno, Czech Republic
| | - Steen Mollerup
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, N-0304 Oslo, Norway
| |
Collapse
|
90
|
Sibila O, Perea L, Cantó E, Shoemark A, Cassidy D, Smith AH, Suarez-Cuartin G, Rodrigo-Troyano A, Keir HR, Oriano M, Ong S, Vidal S, Blasi F, Aliberti S, Chalmers JD. Antimicrobial peptides, disease severity and exacerbations in bronchiectasis. Thorax 2019; 74:835-842. [DOI: 10.1136/thoraxjnl-2018-212895] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 01/13/2023]
Abstract
RationaleRecently a frequent exacerbator phenotype has been described in bronchiectasis, but the underlying biological mechanisms are unknown. Antimicrobial peptides (AMPs) are important in host defence against microbes but can be proinflammatory in chronic lung disease.ObjectivesTo determine pulmonary and systemic levels of AMP and their relationship with disease severity and future risk of exacerbations in bronchiectasis.MethodsA total of 135 adults with bronchiectasis were prospectively enrolled at three European centres. Levels of cathelicidin LL-37, lactoferrin, lysozyme and secretory leucocyte protease inhibitor (SLPI) in serum and sputum were determined at baseline by ELISA. Patients were followed up for 12 months. We examined the ability of sputum AMP to predict future exacerbation risk.Measurements and main resultsAMP levels were higher in sputum than in serum, suggesting local AMP release. Patients with more severe disease at baseline had dysregulation of airway AMP. Higher LL-37 and lower SLPI levels were associated with Bronchiectasis Severity Index, lower FEV1 (forced expiratory volume in 1 s) and Pseudomonas aeruginosa infection. Low SLPI levels were also associated with the exacerbation frequency at baseline. During follow-up, higher LL-37 and lower SLPI levels were associated with a shorter time to the next exacerbation, whereas LL-37 alone predicted exacerbation frequency over the next 12 months.ConclusionsPatients with bronchiectasis showed dysregulated sputum AMP levels, characterised by elevated LL-37 and reduced SLPI levels in the frequent exacerbator phenotype. Elevated LL-37 and reduced SLPI levels are associated with Pseudomonas aeruginosa infection and can predict future risk of exacerbations in bronchiectasis.
Collapse
|
91
|
Coureuil M, Jamet A, Bille E, Lécuyer H, Bourdoulous S, Nassif X. Molecular interactions between Neisseria meningitidis and its human host. Cell Microbiol 2019; 21:e13063. [PMID: 31167044 PMCID: PMC6899865 DOI: 10.1111/cmi.13063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/21/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Neisseria meningitidis is a Gram‐negative bacterium that asymptomatically colonises the nasopharynx of humans. For an unknown reason, N. meningitidis can cross the nasopharyngeal barrier and invade the bloodstream where it becomes one of the most harmful extracellular bacterial pathogen. This infectious cycle involves the colonisation of two different environments. (a) In the nasopharynx, N. meningitidis grow on the top of mucus‐producing epithelial cells surrounded by a complex microbiota. To survive and grow in this challenging environment, the meningococcus expresses specific virulence factors such as polymorphic toxins and MDAΦ. (b) Meningococci have the ability to survive in the extra cellular fluids including blood and cerebrospinal fluid. The interaction of N. meningitidis with human endothelial cells leads to the formation of typical microcolonies that extend overtime and promote vascular injury, disseminated intravascular coagulation, and acute inflammation. In this review, we will focus on the interplay between N. meningitidis and these two different niches at the cellular and molecular level and discuss the use of inhibitors of piliation as a potent therapeutic approach.
Collapse
Affiliation(s)
- Mathieu Coureuil
- Inserm, Institut Necker Enfants Malades, U1151, Paris, France.,Université de Paris, UMR_S 1151, Paris, France.,CNRS, UMR 8253, Paris, France
| | - Anne Jamet
- Inserm, Institut Necker Enfants Malades, U1151, Paris, France.,Université de Paris, UMR_S 1151, Paris, France.,CNRS, UMR 8253, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Emmanuelle Bille
- Inserm, Institut Necker Enfants Malades, U1151, Paris, France.,Université de Paris, UMR_S 1151, Paris, France.,CNRS, UMR 8253, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Hervé Lécuyer
- Inserm, Institut Necker Enfants Malades, U1151, Paris, France.,Université de Paris, UMR_S 1151, Paris, France.,CNRS, UMR 8253, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Sandrine Bourdoulous
- Université de Paris, UMR_S 1151, Paris, France.,Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France
| | - Xavier Nassif
- Inserm, Institut Necker Enfants Malades, U1151, Paris, France.,Université de Paris, UMR_S 1151, Paris, France.,CNRS, UMR 8253, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
92
|
Rao DM, Phan DT, Choo MJ, Weaver MR, Oberley-Deegan RE, Bowler RP, Gally F. Impact of fatty acid binding protein 5-deficiency on COPD exacerbations and cigarette smoke-induced inflammatory response to bacterial infection. Clin Transl Med 2019; 8:7. [PMID: 30877402 PMCID: PMC6420539 DOI: 10.1186/s40169-019-0227-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/09/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Although cigarette smoking (CS) is by far the most important risk factor of chronic obstructive pulmonary disease (COPD), repeated and sustained infections are clearly linked to disease pathogenesis and are responsible for acute inflammatory flares (i.e. COPD exacerbations). We have previously identified Fatty Acid Binding Protein 5 (FABP5) as an important anti-inflammatory protein in primary airway epithelial cells. RESULTS In this study we found decreased FABP5 mRNA and protein levels in peripheral blood mononuclear cells (PBMCs) of COPD patients, especially among those who reported episodes of COPD exacerbations. Using wildtype (WT) and FABP5-/- mice, we examined the effects of FABP5 on CS and infection-induced inflammatory responses. Similarly to what we saw in airway epithelial cells, infection increased FABP5 expression while CS decreased FABP5 expression in mouse lung tissues. CS-exposed and P. aeruginosa-infected FABP5-/- mice had significantly increased inflammation as shown by increased lung histopathological score, cell infiltration and inflammatory cytokine levels. Restoration of FABP5 in alveolar macrophages using a lentiviral approach attenuated the CS- and bacteria-induced pulmonary inflammation. And finally, while P. aeruginosa infection increased PPARγ activity, CS or FABP5 knockdown greatly reduced PPARγ activity. CONCLUSIONS These findings support a model in which CS-induced FABP5 inhibition contributes to increased inflammation in COPD exacerbations. It is interesting to speculate that the increased inflammation is a result of decreased PPARγ activity.
Collapse
Affiliation(s)
- Deviyani M. Rao
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO 80206 USA
| | - Della T. Phan
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO 80206 USA
| | - Michelle J. Choo
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO 80206 USA
| | - Michael R. Weaver
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206 USA
| | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC 6.12.392, 985870 Nebraska Medical Center, Omaha, NE 68198-5870 USA
| | - Russell P. Bowler
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206 USA
| | - Fabienne Gally
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO 80206 USA
| |
Collapse
|
93
|
Czekala L, Simms L, Stevenson M, Trelles-Sticken E, Walker P, Walele T. High Content Screening in NHBE cells shows significantly reduced biological activity of flavoured e-liquids, when compared to cigarette smoke condensate. Toxicol In Vitro 2019; 58:86-96. [PMID: 30880017 DOI: 10.1016/j.tiv.2019.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 01/05/2023]
Abstract
There is scientific agreement that the detrimental effects of cigarettes are produced by the formation of Harmful and Potentially Harmful Constituents from tobacco combustion and not by nicotine. For this reason numerous public health bodies and governments worldwide have indicated that e-cigarettes have a central role to play in tobacco harm reduction. In this study, high content screening (HCS) was used to compare the effects of neat e-liquids and 3R4F reference cigarette smoke condensate (CSC), which served as a positive control, in Normal Human Bronchial Epithelial (NHBE) cells. The endpoints measured covered cellular health, energy production and oxidative stress. Base liquids, with or without nicotine, and commercial, flavoured, nicotine-containing e-liquids (CFs), had little or no effect on cell viability and most HCS endpoints even at significantly higher concentrations (typically 100 times or higher) than 3R4F CSC. CSC induced a dose-dependent decrease of cell viability and triggered the response in all HCS endpoints. Effects of CFs were typically observed at or above 1%. CF Menthol was the most active flavour, with minimum effective concentrations 43 to 659 times higher than corresponding 3R4F CSC concentrations. Our results show a lower biological activity of e-liquids compared to cigarette smoke condensate in this experimental setting, across wide range of cellular endpoints.
Collapse
Affiliation(s)
- Lukasz Czekala
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom.
| | - Liam Simms
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Matthew Stevenson
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Edgar Trelles-Sticken
- Reemstma Cigarettenfabriken GmbH, An Imperial Brands Company, Albert Einstein Ring 7, D-22791 Hamburg, Germany
| | - Paul Walker
- Cyprotex No. 24 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, United Kingdom
| | - Tanvir Walele
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| |
Collapse
|
94
|
Increased susceptibility of airway epithelial cells from ataxia-telangiectasia to S. pneumoniae infection due to oxidative damage and impaired innate immunity. Sci Rep 2019; 9:2627. [PMID: 30796268 PMCID: PMC6385340 DOI: 10.1038/s41598-019-38901-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/19/2018] [Indexed: 12/25/2022] Open
Abstract
Respiratory disease is a major cause of morbidity and mortality in patients with ataxia-telangiectasia (A-T) who are prone to recurrent sinopulmonary infections, bronchiectasis, pulmonary fibrosis, and pulmonary failure. Upper airway infections are common in patients and S. pneumoniae is associated with these infections. We demonstrate here that the upper airway microbiome in patients with A-T is different from that to healthy controls, with S. pneumoniae detected largely in patients only. Patient-specific airway epithelial cells and differentiated air-liquid interface cultures derived from these were hypersensitive to infection which was at least in part due to oxidative damage since it was partially reversed by catalase. We also observed increased levels of the pro-inflammatory cytokines IL-8 and TNF-α (inflammasome-independent) and a decreased level of the inflammasome-dependent cytokine IL-β in patient cells. Further investigation revealed that the ASC-Caspase 1 signalling pathway was defective in A-T airway epithelial cells. These data suggest that the heightened susceptibility of these cells to S. pneumoniae infection is due to both increased oxidative damage and a defect in inflammasome activation, and has implications for lung disease in these patients.
Collapse
|
95
|
Gan T, Yang Y, Hu F, Chen X, Zhou J, Li Y, Xu Y, Wang H, Chen Y, Zhang M. TLR3 Regulated Poly I:C-Induced Neutrophil Extracellular Traps and Acute Lung Injury Partly Through p38 MAP Kinase. Front Microbiol 2018; 9:3174. [PMID: 30622526 PMCID: PMC6308186 DOI: 10.3389/fmicb.2018.03174] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is the leading cause of morbidity and mortality in critically ill patients. Neutrophil extracellular traps (NETs) have been well documented in the ALI model of bacterial infection. In the present study, we demonstrated that poly I:C could induce pulmonary NETs. Upon poly I:C intratracheal inoculation, neutrophil infiltration in the bronchoalveolar lavage fluid (BALF) was significantly increased. Furthermore, the inflammatory cytokines IL-1β, IL-6, and TNF-α in the lung were also significantly elevated. Neutrophil depletion abolished NETs and decreased both neutrophil infiltration and IL-1β in the lung. As expected, DNase I, an inhibitor of MPO and NADPH, decreased pulmonary inflammation and NETs. Blocking of the poly I:C receptor TLR3 reduced lung inflammation and NETs. The MAPK kinase inhibitor p38 diminished the formation of NETs and restored the expression of the tight junction protein claudin-5 in the mouse lung when challenged with poly I:C. In summary, poly I:C induced the formation of pulmonary NETs and ALI, which may be associated with the activation of p38 MAPK and the decreased expression of claudin-5.
Collapse
Affiliation(s)
- Tingting Gan
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yonglin Yang
- Department of Infectious Diseases, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China
| | - Fan Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xichen Chen
- Analysis Center, Nanjing Medical University, Nanjing, China
| | - Jiawei Zhou
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Li
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xu
- Nanjing University Medical School, The Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Huijuan Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yu Chen
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China.,Department of Immunology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
96
|
Schagen J, Sly PD, Fantino E. Characterizing well-differentiated culture of primary human nasal epithelial cells for use in wound healing assays. J Transl Med 2018; 98:1478-1486. [PMID: 30089850 DOI: 10.1038/s41374-018-0100-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/18/2022] Open
Abstract
The nasal epithelium is the initial contact between the external environment and the respiratory tract and how it responds to noxious stimuli and repairs epithelial damage is important. Growing airway epithelial cells in culture at air-liquid interface allows for a physiologically relevant model of the human upper airways. The aim of the present study was to characterize human primary nasal epithelial cells grown at the air-liquid interface and establish a model for use in wound healing assays. This study determined the time required for full differentiation of nasal epithelial cells in an air-liquid interface culture to be at least 7 weeks using the standardized B-ALI media. Also, a model was established that studied the response to wounding and the effect of EGFR inhibition on this process. Nasal epithelial cultures from healthy subjects were differentiated at air-liquid interface and manually wounded. Wounds were monitored over time to complete closure using a time lapse imaging microscope with cultures identified to have a rate of wound healing above 2.5%/h independent of initial wound size. EGFR inhibition caused the rate of wound healing to drop a significant 4.6%/h with there being no closure of the wound after 48 h. The robust model established in this study will be essential for studying factors influencing wound healing, including host disease status and environmental exposures in the future.
Collapse
Affiliation(s)
- Johanna Schagen
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| | - Peter D Sly
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia.
| | - Emmanuelle Fantino
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| |
Collapse
|
97
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
98
|
Kaczmarek JC, Kauffman KJ, Fenton OS, Sadtler K, Patel AK, Heartlein MW, DeRosa F, Anderson DG. Optimization of a Degradable Polymer-Lipid Nanoparticle for Potent Systemic Delivery of mRNA to the Lung Endothelium and Immune Cells. NANO LETTERS 2018; 18:6449-6454. [PMID: 30211557 PMCID: PMC6415675 DOI: 10.1021/acs.nanolett.8b02917] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
mRNA therapeutics hold great potential for treating a variety of diseases through protein-replacement, immunomodulation, and gene editing. However, much like siRNA therapy the majority of progress in mRNA delivery has been confined to the liver. Previously, we demonstrated that poly(β-amino esters), a class of degradable polymers, are capable of systemic mRNA delivery to the lungs in mice when formulated into nanoparticles with poly(ethylene glycol)-lipid conjugates. Using experimental design, a statistical approach to optimization that reduces experimental burden, we demonstrate herein that these degradable polymer-lipid nanoparticles can be optimized in terms of polymer synthesis and nanoparticle formulation to achieve a multiple order-of-magnitude increase in potency. Furthermore, using genetically engineered Cre reporter mice, we demonstrate that mRNA is functionally delivered to both the lung endothelium and pulmonary immune cells, expanding the potential utility of these nanoparticles.
Collapse
Affiliation(s)
- James C. Kaczmarek
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Kevin J. Kauffman
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Owen S. Fenton
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Kaitlyn Sadtler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Asha K. Patel
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2RD (UK)
| | | | | | - Daniel G. Anderson
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA (USA)
| |
Collapse
|
99
|
Epithelial Cells Attenuate Toll-Like Receptor-Mediated Inflammatory Responses in Monocyte-Derived Macrophage-Like Cells to Mycobacterium tuberculosis by Modulating the PI3K/Akt/mTOR Signaling Pathway. Mediators Inflamm 2018; 2018:3685948. [PMID: 30356420 PMCID: PMC6178170 DOI: 10.1155/2018/3685948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022] Open
Abstract
Both alveolar macrophages (AMs) and alveolar epithelial cells (AECs) are main targets of Mycobacterium tuberculosis (M. tuberculosis (Mtb)). Intercellular communications between mucosal AECs and AMs have important implications in cellular responses to exogenous insults. However, molecular mechanisms underpinning interactions responding to Mtb remain largely unknown. In this study, impacts of AECs on Toll-like receptor- (TLR-) mediated inflammatory responses of AMs to Mtb virulent strain H37Rv were interrogated using an air-liquid interface (ALI) coculture model of epithelial A549 cells and U937 monocyte-derived macrophage-like cells. Results showed that Mtb-activated TLR-mediated inflammatory responses in U937 cells were significantly alleviated when A549 cells were coinfected with H37Rv, in comparison with the infection of U937 cells alone. Mechanistically, PI3K/Akt/mTOR signaling was involved in the epithelial cell-modulated Mtb-activated TLR signaling. The epithelial cell-attenuated TLR signaling in U937s could be reversed by PI3K inhibitor LY294002 and mTOR inhibitor rapamycin, but not glycogen synthase kinase 3β inhibitor LiCl, suggesting that the epithelially modulated-TLR signaling in macrophages was in part caused by inhibiting the TLR-triggered PI3K/Akt/mTOR signaling pathway. Together, this study demonstrates that mucosal AEC-derived signals play an important role in modulating inflammatory responses of AMs to Mtb, which thus also offers an insight into cellular communications between AECs and AMs to Mtb infections.
Collapse
|
100
|
Jin L, Hu S, Tu T, Huang Z, Tang Q, Ma J, Wang X, Li X, Zhou X, Shuai S, Li M. Global Long Noncoding RNA and mRNA Expression Changes between Prenatal and Neonatal Lung Tissue in Pigs. Genes (Basel) 2018; 9:genes9090443. [PMID: 30189656 PMCID: PMC6162397 DOI: 10.3390/genes9090443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/25/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022] Open
Abstract
Lung tissue plays an important role in the respiratory system of mammals after birth. Early lung development includes six key stages, of which the saccular stage spans the pre- and neonatal periods and prepares the distal lung for alveolarization and gas-exchange. However, little is known about the changes in gene expression between fetal and neonatal lungs. In this study, we performed transcriptomic analysis of messenger RNA (mRNA) and long noncoding RNA (lncRNA) expressed in the lung tissue of fetal and neonatal piglets. A total of 19,310 lncRNAs and 14,579 mRNAs were identified and substantially expressed. Furthermore, 3248 mRNAs were significantly (FDR-adjusted p value ≤ 0.05, FDR: False Discovery Rate) differentially expressed and were mainly enriched in categories related to cell proliferation, immune response, hypoxia response, and mitochondrial activation. For example, CCNA2, an important gene involved in the cell cycle and DNA replication, was upregulated in neonatal lungs. We also identified 452 significantly (FDR-adjusted p value ≤ 0.05) differentially expressed lncRNAs, which might function in cell proliferation, mitochondrial activation, and immune response, similar to the differentially expressed mRNAs. These results suggest that differentially expressed mRNAs and lncRNAs might co-regulate lung development in early postnatal pigs. Notably, the TU64359 lncRNA might promote distal lung development by up-regulating the heparin-binding epidermal growth factor-like (HB-EGF) expression. Our research provides basic lung development datasets and will accelerate clinical researches of newborn lung diseases with pig models.
Collapse
Affiliation(s)
- Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Teng Tu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Jideng Ma
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xun Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xuewei Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xuan Zhou
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Surong Shuai
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|