1
|
Sinha V, Singh A, Singh A, Saraswati SSK, Rana AK, Kalra K, Natarajan K. Potassium ion channel Kir2.1 negatively regulates protective responses to Mycobacterium bovis BCG. J Leukoc Biol 2024; 116:644-656. [PMID: 38489665 DOI: 10.1093/jleuko/qiae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tuberculosis caused by the pathogen Mycobacterium tuberculosis leads to increased mortality and morbidity worldwide. The prevalence of highly drug-resistant strains has reinforced the need for greater understanding of host-pathogen interactions at the cellular and molecular levels. Our previous work demonstrated critical roles of calcium ion channels in regulating protective responses to mycobacteria. In this report, we deciphered the roles of inwardly rectifying K+ ion channel Kir2.1 in epithelial cells. Data showed that infection of epithelial cells (and macrophages) increases the surface expression of Kir2.1. This increased expression of Kir2.1 results in higher intracellular mycobacterial survival, as either inhibiting or knocking down Kir2.1 results in mounting of a higher oxidative burst leading to a significant attenuation of mycobacterial survival. Further, inhibiting Kir2.1 also led to increased expression of T cell costimulatory molecules accompanied with increased activation of MAP kinases and transcription factors nuclear factor κB and phosphorylated CREB. Furthermore, inhibiting Kir2.1 induced increased autophagy and apoptosis that could also contribute to decreased bacterial survival. Interestingly, an increased association of heat shock protein 70 kDa with Kir2.1 was observed. These results showed that mycobacteria modulate the expression and function of Kir2.1 in epithelial cells to its advantage.
Collapse
Affiliation(s)
- Vishal Sinha
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Akshita Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Aarti Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Shakuntala Surender Kumar Saraswati
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Ankush Kumar Rana
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Kanika Kalra
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Krishnamurthy Natarajan
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| |
Collapse
|
2
|
Bertuzzi M, Howell GJ, Thomson DD, Fortune-Grant R, Möslinger A, Dancer P, Van Rhijn N, Motsi N, Codling A, Bignell EM. Epithelial uptake leads to fungal killing in vivo and is aberrant in COPD-derived epithelial cells. iScience 2024; 27:109939. [PMID: 38846001 PMCID: PMC11154633 DOI: 10.1016/j.isci.2024.109939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/07/2023] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Hundreds of spores of Aspergillus fumigatus (Af) are inhaled daily by human beings, representing a constant, possibly fatal, threat to respiratory health. The small size of Af spores suggests that interactions with alveolar epithelial cells (AECs) are frequent; thus, we hypothesized that spore uptake by AECs is important for driving fungal killing and susceptibility to Aspergillus-related disease. Using single-cell approaches to measure spore uptake and its outcomes in vivo, we demonstrate that Af spores are internalized and killed by AECs during whole-animal infection. Moreover, comparative analysis of primary human AECs from healthy and chronic obstructive pulmonary disease (COPD) donors revealed significant alterations in the uptake and killing of spores in COPD-derived AECs. We conclude that AECs contribute to the killing of Af spores and that dysregulation of curative AEC responses in COPD may represent a driver of Aspergillus-related diseases.
Collapse
Affiliation(s)
- Margherita Bertuzzi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Gareth J. Howell
- Flow Cytometry Core Facility, Faculty of Biology, Medicine and Health, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Darren D. Thomson
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Rachael Fortune-Grant
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Anna Möslinger
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Patrick Dancer
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Norman Van Rhijn
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Natasha Motsi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Alice Codling
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| | - Elaine M. Bignell
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
3
|
Fallert L, Urigoitia-Asua A, Cipitria A, Jimenez de Aberasturi D. Dynamic 3D in vitro lung models: applications of inorganic nanoparticles for model development and characterization. NANOSCALE 2024; 16:10880-10900. [PMID: 38787741 DOI: 10.1039/d3nr06672j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Being a vital organ exposed to the external environment, the lung is susceptible to a plethora of pathogens and pollutants. This is reflected in high incidences of chronic respiratory diseases, which remain a leading cause of mortality world-wide and pose a persistent global burden. It is thus of paramount importance to improve our understanding of these pathologies and provide better therapeutic options. This necessitates the development of representative and physiologically relevant in vitro models. Advances in bioengineering have enabled the development of sophisticated models that not only capture the three-dimensional architecture of the cellular environment but also incorporate the dynamics of local biophysical stimuli. However, such complex models also require novel approaches that provide reliable characterization. Within this review we explore how 3D bioprinting and nanoparticles can serve as multifaceted tools to develop such dynamic 4D printed in vitro lung models and facilitate their characterization in the context of pulmonary fibrosis and breast cancer lung metastasis.
Collapse
Affiliation(s)
- Laura Fallert
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
| | - Ane Urigoitia-Asua
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
- POLYMAT, Basque Centre for Macromolecular Design and Engineering, 20018 Donostia-San Sebastián, Spain
| | - Amaia Cipitria
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Dorleta Jimenez de Aberasturi
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), 20014 Donostia-San Sebastián, Spain
| |
Collapse
|
4
|
Olmo-Fontánez AM, Scordo JM, Schami A, Garcia-Vilanova A, Pino PA, Hicks A, Mishra R, Jose Maselli D, Peters JI, Restrepo BI, Nargan K, Naidoo T, Clemens DL, Steyn AJC, Thacker VV, Turner J, Schlesinger LS, Torrelles JB. Human alveolar lining fluid from the elderly promotes Mycobacterium tuberculosis intracellular growth and translocation into the cytosol of alveolar epithelial cells. Mucosal Immunol 2024; 17:155-168. [PMID: 38185331 PMCID: PMC11034793 DOI: 10.1016/j.mucimm.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/15/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
The elderly population is highly susceptible to developing respiratory diseases, including tuberculosis, a devastating disease caused by the airborne pathogen Mycobacterium tuberculosis (M.tb) that kills one person every 18 seconds. Once M.tb reaches the alveolar space, it contacts alveolar lining fluid (ALF), which dictates host-cell interactions. We previously determined that age-associated dysfunction of soluble innate components in human ALF leads to accelerated M.tb growth within human alveolar macrophages. Here we determined the impact of human ALF on M.tb infection of alveolar epithelial type cells (ATs), another critical lung cellular determinant of infection. We observed that elderly ALF (E-ALF)-exposed M.tb had significantly increased intracellular growth with rapid replication in ATs compared to adult ALF (A-ALF)-exposed bacteria, as well as a dampened inflammatory response. A potential mechanism underlying this accelerated growth in ATs was our observation of increased bacterial translocation into the cytosol, a compartment that favors bacterial replication. These findings in the context of our previous studies highlight how the oxidative and dysfunctional status of the elderly lung mucosa determines susceptibility to M.tb infection, including dampening immune responses and favoring bacterial replication within alveolar resident cell populations, including ATs, the most abundant resident cell type within the alveoli.
Collapse
Affiliation(s)
- Angélica M Olmo-Fontánez
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA; Integrated Biomedical Sciences Program, University of Texas Health Science Center at San Antonio, Texas, USA.
| | - Julia M Scordo
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA; Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Alyssa Schami
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA; Integrated Biomedical Sciences Program, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Andreu Garcia-Vilanova
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Paula A Pino
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Amberlee Hicks
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Richa Mishra
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Diego Jose Maselli
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Jay I Peters
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Blanca I Restrepo
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA; University of Texas Health Science Center at Houston, School of Public Health, Brownsville campus, Brownsville, Texas, USA; South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Kievershen Nargan
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Threnesan Naidoo
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa; Department of Laboratory Medicine and Pathology, Walter Sisulu University, Mthatha, South Africa
| | - Daniel L Clemens
- University of California, Los Angeles Health Sciences, Los Angeles, California, USA
| | - Adrie J C Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa; Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Vivek V Thacker
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Department of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Joanne Turner
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Larry S Schlesinger
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jordi B Torrelles
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA; International Center for the Advancement of Research and Education (I●CARE), Texas Biomedical Research Institute, San Antonio, TX, US.
| |
Collapse
|
5
|
Suman SK, Chandrasekaran N, Priya Doss CG. Micro-nanoemulsion and nanoparticle-assisted drug delivery against drug-resistant tuberculosis: recent developments. Clin Microbiol Rev 2023; 36:e0008823. [PMID: 38032192 PMCID: PMC10732062 DOI: 10.1128/cmr.00088-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Tuberculosis (TB) is a major global health problem and the second most prevalent infectious killer after COVID-19. It is caused by Mycobacterium tuberculosis (Mtb) and has become increasingly challenging to treat due to drug resistance. The World Health Organization declared TB a global health emergency in 1993. Drug resistance in TB is driven by mutations in the bacterial genome that can be influenced by prolonged drug exposure and poor patient adherence. The development of drug-resistant forms of TB, such as multidrug resistant, extensively drug resistant, and totally drug resistant, poses significant therapeutic challenges. Researchers are exploring new drugs and novel drug delivery systems, such as nanotechnology-based therapies, to combat drug resistance. Nanodrug delivery offers targeted and precise drug delivery, improves treatment efficacy, and reduces adverse effects. Along with nanoscale drug delivery, a new generation of antibiotics with potent therapeutic efficacy, drug repurposing, and new treatment regimens (combinations) that can tackle the problem of drug resistance in a shorter duration could be promising therapies in clinical settings. However, the clinical translation of nanomedicines faces challenges such as safety, large-scale production, regulatory frameworks, and intellectual property issues. In this review, we present the current status, most recent findings, challenges, and limiting barriers to the use of emulsions and nanoparticles against drug-resistant TB.
Collapse
Affiliation(s)
- Simpal Kumar Suman
- School of Bio Sciences & Technology (SBST), Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nano Biotechnology (CNBT), Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - C. George Priya Doss
- Laboratory for Integrative Genomics, Department of Integrative Biology, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
6
|
Albers GJ, Amouret A, Ciupka K, Montes-Cobos E, Feldmann C, Reichardt HM. Glucocorticoid Nanoparticles Show Full Therapeutic Efficacy in a Mouse Model of Acute Lung Injury and Concomitantly Reduce Adverse Effects. Int J Mol Sci 2023; 24:16843. [PMID: 38069173 PMCID: PMC10705980 DOI: 10.3390/ijms242316843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Glucocorticoids (GCs) are widely used to treat inflammatory disorders such as acute lung injury (ALI). Here, we explored inorganic-organic hybrid nanoparticles (IOH-NPs) as a new delivery vehicle for GCs in a mouse model of ALI. Betamethasone (BMZ) encapsulated into IOH-NPs (BNPs) ameliorated the massive infiltration of neutrophils into the airways with a similar efficacy as the free drug. This was accompanied by a potent inhibition of pulmonary gene expression and secretion of pro-inflammatory mediators, whereas the alveolar-capillary barrier integrity was only restored by BMZ in its traditional form. Experiments with genetically engineered mice identified myeloid cells and alveolar type II (AT II) cells as essential targets of BNPs in ALI therapy, confirming their high cell-type specificity. Consequently, adverse effects were reduced when using IOH-NPs for GC delivery. BNPs did not alter T and B cell numbers in the blood and also prevented the induction of muscle atrophy after three days of treatment. Collectively, our data suggest that IOH-NPs target GCs to myeloid and AT II cells, resulting in full therapeutic efficacy in the treatment of ALI while being associated with reduced adverse effects.
Collapse
Affiliation(s)
- Gesa J. Albers
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Agathe Amouret
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Katrin Ciupka
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Elena Montes-Cobos
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany;
| | - Holger M. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
7
|
Grubwieser P, Hilbe R, Gehrer CM, Grander M, Brigo N, Hoffmann A, Seifert M, Berger S, Theurl I, Nairz M, Weiss G. Klebsiella pneumoniae manipulates human macrophages to acquire iron. Front Microbiol 2023; 14:1223113. [PMID: 37637102 PMCID: PMC10451090 DOI: 10.3389/fmicb.2023.1223113] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Background Klebsiella pneumoniae (KP) is a major cause of hospital-acquired infections, such as pneumonia. Moreover, it is classified as a pathogen of concern due to sprawling anti-microbial resistance. During infection, the gram-negative pathogen is capable of establishing an intracellular niche in macrophages by altering cellular metabolism. One factor critically affecting the host-pathogen interaction is the availability of essential nutrients, like iron, which is required for KP to proliferate but which also modulates anti-microbial immune effector pathways. We hypothesized, that KP manipulates macrophage iron homeostasis to acquire this crucial nutrient for sustained proliferation. Methods We applied an in-vitro infection model, in which human macrophage-like PMA-differentiated THP1 cells were infected with KP (strain ATCC 43816). During a 24-h course of infection, we quantified the number of intracellular bacteria via serial plating of cell lysates and evaluated the effects of different stimuli on intracellular bacterial numbers and iron acquisition. Furthermore, we analyzed host and pathogen specific gene and protein expression of key iron metabolism molecules. Results Viable bacteria are recovered from macrophage cell lysates during the course of infection, indicative of persistence of bacteria within host cells and inefficient pathogen clearing by macrophages. Strikingly, following KP infection macrophages strongly induce the expression of the main cellular iron importer transferrin-receptor-1 (TFR1). Accordingly, intracellular KP proliferation is further augmented by the addition of iron loaded transferrin. The induction of TFR1 is mediated via the STAT-6-IL-10 axis, and pharmacological inhibition of this pathway reduces macrophage iron uptake, elicits bacterial iron starvation, and decreases bacterial survival. Conclusion Our results suggest, that KP manipulates macrophage iron metabolism to acquire iron once confined inside the host cell and enforces intracellular bacterial persistence. This is facilitated by microbial mediated induction of TFR1 via the STAT-6-IL-10 axis. Mechanistic insights into immune metabolism will provide opportunities for the development of novel antimicrobial therapies.
Collapse
Affiliation(s)
- Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Clemens Michael Gehrer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel Grander
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Sylvia Berger
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pulmonology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
van der Geest R, Fan H, Peñaloza HF, Bain WG, Xiong Z, Kohli N, Larson E, Sullivan MLG, Franks JM, Stolz DB, Ito R, Chen K, Doi Y, Harriff MJ, Lee JS. Phagocytosis is a primary determinant of pulmonary clearance of clinical Klebsiella pneumoniae isolates. Front Cell Infect Microbiol 2023; 13:1150658. [PMID: 37056705 PMCID: PMC10086180 DOI: 10.3389/fcimb.2023.1150658] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction Klebsiella pneumoniae (Kp) is a common cause of hospital-acquired pneumonia. Although previous studies have suggested that evasion of phagocytic uptake is a virulence determinant of Kp, few studies have examined phagocytosis sensitivity in clinical Kp isolates. Methods We screened 19 clinical respiratory Kp isolates that were previously assessed for mucoviscosity for their sensitivity to macrophage phagocytic uptake, and evaluated phagocytosis as a functional correlate of in vivo Kp pathogenicity. Results The respiratory Kp isolates displayed heterogeneity in the susceptibility to macrophage phagocytic uptake, with 14 out of 19 Kp isolates displaying relative phagocytosis-sensitivity compared to the reference Kp strain ATCC 43816, and 5 out of 19 Kp isolates displaying relative phagocytosis-resistance. Intratracheal infection with the non-mucoviscous phagocytosis-sensitive isolate S17 resulted in a significantly lower bacterial burden compared to infection with the mucoviscous phagocytosis-resistant isolate W42. In addition, infection with S17 was associated with a reduced inflammatory response, including reduced bronchoalveolar lavage fluid (BAL) polymorphonuclear (PMN) cell count, and reduced BAL TNF, IL-1β, and IL-12p40 levels. Importantly, host control of infection with the phagocytosis-sensitive S17 isolate was impaired in alveolar macrophage (AM)-depleted mice, whereas AM-depletion had no significant impact on host defense against infection with the phagocytosis-resistant W42 isolate. Conclusion Altogether, these findings show that phagocytosis is a primary determinant of pulmonary clearance of clinical Kp isolates.
Collapse
Affiliation(s)
- Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hongye Fan
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hernán F. Peñaloza
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - William G. Bain
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Veterans Affairs (VA) Pittsburgh Health Care System, Pittsburgh, PA, United States
| | - Zeyu Xiong
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Naina Kohli
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Emily Larson
- Veterans Affairs (VA) Portland Health Care System, Portland, OR, United States
| | - Mara L. G. Sullivan
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jonathan M. Franks
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, United States
| | - Donna B. Stolz
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ryota Ito
- Department of Respiratory Medicine, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Kong Chen
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yohei Doi
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Departments of Microbiology and Infectious Diseases, Fujita Health University, Toyoake, Japan
| | - Melanie J. Harriff
- Veterans Affairs (VA) Portland Health Care System, Portland, OR, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health State University, Portland, OR, United States
| | - Janet S. Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
9
|
Christenson JL, Williams MM, Richer JK. The underappreciated role of resident epithelial cell populations in metastatic progression: contributions of the lung alveolar epithelium. Am J Physiol Cell Physiol 2022; 323:C1777-C1790. [PMID: 36252127 PMCID: PMC9744653 DOI: 10.1152/ajpcell.00181.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Metastatic cancer is difficult to treat and is responsible for the majority of cancer-related deaths. After cancer cells initiate metastasis and successfully seed a distant site, resident cells in the tissue play a key role in determining how metastatic progression develops. The lung is the second most frequent site of metastatic spread, and the primary site of metastasis within the lung is alveoli. The most abundant cell type in the alveolar niche is the epithelium. This review will examine the potential contributions of the alveolar epithelium to metastatic progression. It will also provide insight into other ways in which alveolar epithelial cells, acting as immune sentinels within the lung, may influence metastatic progression through their various interactions with cells in the surrounding microenvironment.
Collapse
Affiliation(s)
- Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michelle M Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
10
|
Do T, Synan L, Ali G, Gappa-Fahlenkamp H. 3D tissue-engineered lung models to study immune responses following viral infections of the small airways. Stem Cell Res Ther 2022; 13:464. [PMID: 36071442 PMCID: PMC9449944 DOI: 10.1186/s13287-022-03134-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/12/2022] [Indexed: 11/10/2022] Open
Abstract
Small airway infections caused by respiratory viruses are some of the most prevalent causes of illness and death. With the recent worldwide pandemic due to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is currently a push in developing models to better understand respiratory diseases. Recent advancements have made it possible to create three-dimensional (3D) tissue-engineered models of different organs. The 3D environment is crucial to study physiological, pathophysiological, and immunomodulatory responses against different respiratory conditions. A 3D human tissue-engineered lung model that exhibits a normal immunological response against infectious agents could elucidate viral and host determinants. To create 3D small airway lung models in vitro, resident epithelial cells at the air-liquid interface are co-cultured with fibroblasts, myeloid cells, and endothelial cells. The air-liquid interface is a key culture condition to develop and differentiate airway epithelial cells in vitro. Primary human epithelial and myeloid cells are considered the best 3D model for studying viral immune responses including migration, differentiation, and the release of cytokines. Future studies may focus on utilizing bioreactors to scale up the production of 3D human tissue-engineered lung models. This review outlines the use of various cell types, scaffolds, and culture conditions for creating 3D human tissue-engineered lung models. Further, several models used to study immune responses against respiratory viruses, such as the respiratory syncytial virus, are analyzed, showing how the microenvironment aids in understanding immune responses elicited after viral infections.
Collapse
Affiliation(s)
- Taylor Do
- Edward Bartlett Chair, School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA
| | - Lilly Synan
- Edward Bartlett Chair, School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA
| | - Gibran Ali
- Edward Bartlett Chair, School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA
| | - Heather Gappa-Fahlenkamp
- Edward Bartlett Chair, School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA.
| |
Collapse
|
11
|
Sempere J, Rossi SA, Chamorro-Herrero I, González-Camacho F, de Lucas MP, Rojas-Cabañeros JM, Taborda CP, Zaragoza Ó, Yuste J, Zambrano A. Minilungs from Human Embryonic Stem Cells to Study the Interaction of Streptococcus pneumoniae with the Respiratory Tract. Microbiol Spectr 2022; 10:e0045322. [PMID: 35695525 PMCID: PMC9241785 DOI: 10.1128/spectrum.00453-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/20/2022] [Indexed: 11/20/2022] Open
Abstract
The new generation of organoids derived from human pluripotent stem cells holds a promising strategy for modeling host-bacteria interaction studies. Organoids recapitulate the composition, diversity of cell types, and, to some extent, the functional features of the native organ. We generated lung bud organoids derived from human embryonic stem cells to study the interaction of Streptococcus pneumoniae (pneumococcus) with the alveolar epithelium. Invasive pneumococcal disease is an important health problem that may occur as a result of the spread of pneumococcus from the lower respiratory tract to sterile sites. We show here an efficient experimental approach to model the main events of the pneumococcal infection that occur in the human lung, exploring bacterial adherence to the epithelium and internalization and triggering an innate response that includes the interaction with surfactant and the expression of representative cytokines and chemokines. Thus, this model, based on human minilungs, can be used to study pneumococcal virulence factors and the pathogenesis of different serotypes, and it will allow therapeutic interventions in a reliable human context. IMPORTANCE Streptococcus pneumoniae is responsible for high morbidity and mortalities rates worldwide, affecting mainly children and adults older than 65 years. Pneumococcus is also the most common etiologic agent of bacterial pneumonia and nonepidemic meningitis, and it is a frequent cause of bacterial sepsis. Although the introduction of pneumococcal vaccines has decreased the burden of pneumococcal disease, the rise of antibiotic-resistant strains and nonvaccine types by serotype replacement is worrisome. To study the biology of pneumococcus and to establish a reliable human model for pneumococcal pathogenesis, we generated human minilungs from embryonic stem cells. The results show that these organoids can be used to model some events occurring during the interaction of pneumococcus with the lung, such as adherence, internalization, and the initial alveolar innate response. This model also represents a great alternative for studying virulence factors involved in pneumonia, drug screening, and other therapeutic interventions.
Collapse
Affiliation(s)
- Julio Sempere
- Biotechnology of Stem Cells and Organoids, Chronic Diseases Program, Instituto de Salud Carlos III, Madrid, Spain
- Spanish Pneumococcal Reference Laboratory, Centro Nacional de Microbiología, and CIBER of Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Suélen Andreia Rossi
- Biotechnology of Stem Cells and Organoids, Chronic Diseases Program, Instituto de Salud Carlos III, Madrid, Spain
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, Brazil
- Mycology Reference Laboratory, Centro Nacional de Microbiología and CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Chamorro-Herrero
- Biotechnology of Stem Cells and Organoids, Chronic Diseases Program, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando González-Camacho
- Spanish Pneumococcal Reference Laboratory, Centro Nacional de Microbiología, and CIBER of Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Pilar de Lucas
- Cellular Biology Unit, Chronic Diseases Program and CIBER of Cancer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - José María Rojas-Cabañeros
- Cellular Biology Unit, Chronic Diseases Program and CIBER of Cancer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Pelleschi Taborda
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Óscar Zaragoza
- Mycology Reference Laboratory, Centro Nacional de Microbiología and CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José Yuste
- Spanish Pneumococcal Reference Laboratory, Centro Nacional de Microbiología, and CIBER of Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Zambrano
- Biotechnology of Stem Cells and Organoids, Chronic Diseases Program, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Hong JH, Lee YC. Anti-Inflammatory Effects of Cicadidae Periostracum Extract and Oleic Acid through Inhibiting Inflammatory Chemokines Using PCR Arrays in LPS-Induced Lung inflammation In Vitro. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060857. [PMID: 35743888 PMCID: PMC9225349 DOI: 10.3390/life12060857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/26/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022]
Abstract
In this study, we aimed to evaluate the anti-inflammatory effects and mechanisms of CP and OA treatments in LPS-stimulated lung epithelial cells on overall chemokines and their receptors using PCR arrays. In addition, we aimed to confirm those effects and mechanisms in LPS-stimulated lung macrophages on some chemokines and cytokines. In our study, CP treatments significantly inhibited the inflammatory mediators CCL2, CCL3, CCL4, CCL5, CCL6, CCL9, CCL11, CCL17, CCL20, CXCL1, CXCL2, CXCL3, CXCL5, CXCL7, CXCL10, TNF-α, and IL-6, while markedly suppressing NF-κB p65 nuclear translocation and the phosphorylations of PI3K p55, Akt, Erk1/2, p38, and NF-κB p65 in LPS-stimulated lung epithelial cells. CP treatments also significantly decreased the inflammatory mediators CCL2, CCL5, CCL17, CXCL1, and CXCL2, while markedly inhibiting phospho-PI3K p55 and iNOS expression in LPS-stimulated lung macrophages. Likewise, OA treatments significantly suppressed the inflammatory mediators CCL2, CCL3, CCL4, CCL5, CCL8, CCL11, CXCL1, CXCL3, CXCL5, CXCL7, CXCL10, CCRL2, TNF-α, and IL-6, while markedly reducing the phosphorylations of PI3K p85, PI3K p55, p38, JNK, and NF-κB p65 in LPS-stimulated lung epithelial cells. Finally, OA treatments significantly inhibited the inflammatory mediators CCL2, CCL5, CCL17, CXCL1, CXCL2, TNF-α, and IL-6, while markedly suppressing phospho-PI3K p55, iNOS, and Cox-2 in LPS-stimulated lung macrophages. These results prove that CP and OA treatments have anti-inflammatory effects on the inflammatory chemokines and cytokines by inhibiting pro-inflammatory mediators, including PI3K, Akt, MAPKs, NF-κB, iNOS, and Cox-2. These findings suggest that CP and OA are potential chemokine-based therapeutic substances for treating the lung and airway inflammation seen in allergic disorders.
Collapse
Affiliation(s)
| | - Young-Cheol Lee
- Correspondence: ; Tel.: +82-33-730-0672; Fax: +82-33-730-0653
| |
Collapse
|
13
|
Grubwieser P, Hoffmann A, Hilbe R, Seifert M, Sonnweber T, Böck N, Theurl I, Weiss G, Nairz M. Airway Epithelial Cells Differentially Adapt Their Iron Metabolism to Infection With Klebsiella pneumoniae and Escherichia coli In Vitro. Front Cell Infect Microbiol 2022; 12:875543. [PMID: 35663465 PMCID: PMC9157649 DOI: 10.3389/fcimb.2022.875543] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Pneumonia is often elicited by bacteria and can be associated with a severe clinical course, respiratory failure and the need for mechanical ventilation. In the alveolus, type-2-alveolar-epithelial-cells (AECII) contribute to innate immune functions. We hypothesized that AECII actively adapt cellular iron homeostasis to restrict this essential nutrient from invading pathogens - a defense strategy termed 'nutritional immunity', hitherto mainly demonstrated for myeloid cells. Methods We established an in-vitro infection model using the human AECII-like cell line A549. We infected cells with Klebsiella pneumoniae (K. pneumoniae) and Escherichia coli (E. coli), two gram-negative bacteria with different modes of infection and frequent causes of hospital-acquired pneumonia. We followed the entry and intracellular growth of these gram-negative bacteria and analyzed differential gene expression and protein levels of key inflammatory and iron metabolism molecules. Results Both, K. pneumoniae and E. coli are able to invade A549 cells, whereas only K. pneumoniae is capable of proliferating intracellularly. After peak bacterial burden, the number of intracellular pathogens declines, suggesting that epithelial cells initiate antimicrobial immune effector pathways to combat bacterial proliferation. The extracellular pathogen E. coli induces an iron retention phenotype in A549 cells, mainly characterized by the downregulation of the pivotal iron exporter ferroportin, the upregulation of the iron importer transferrin-receptor-1 and corresponding induction of the iron storage protein ferritin. In contrast, cells infected with the facultative intracellular bacterium K. pneumoniae exhibit an iron export phenotype indicated by ferroportin upregulation. This differential regulation of iron homeostasis and the pathogen-specific inflammatory reaction is likely mediated by oxidative stress. Conclusion AECII-derived A549 cells show pathogen-specific innate immune functions and adapt their iron handling in response to infection. The differential regulation of iron transporters depends on the preferential intra- or extracellular localization of the pathogen and likely aims at limiting bacterial iron availability.
Collapse
Affiliation(s)
- Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nina Böck
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Zhu Y, Yang M, Li XH, Xu WJ, Gao W, Chen YH, Li JD, Li Q. Nogo-B promotes epithelial-mesenchymal transition in lung fibrosis via PERK branch of the endoplasmic reticulum stress pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:563. [PMID: 33987261 DOI: 10.21037/atm-20-6143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a fatal chronic pulmonary fibrosis disease and pathological mechanisms of fibrogenesis in IPF are still to be elucidated. Here, we investigated the potential role of Nogo-B in pulmonary fibrogenesis. Methods A mouse model of pulmonary fibrosis was established by intratracheal injection of bleomycin (BLM). Lung epithelial cells MLE-12 and TC-1 JHU-1 were cultured for TGF-β treatment. The extent of lung fibrosis was evaluated using hematoxylin and eosin (HE) staining and Masson staining in model mice and Nogo-B knockout mice. The protein levels of Nogo-B, endoplasmic reticulum stress (ERS) sensors including PERK, IRE1α, ATF6 and epithelial-mesenchymal transition (EMT) markers including E-cadherin and N-cadherin, vimentin were assayed by Western blotting respectively after Nogo-B knockdown or overexpression with lentivirus. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate cytokine levels of TGF-β, TNF-α, IL-1β, IL-6 and IL-10 in bronchoalveolar lavage fluid (BALF). Results Nogo-B expression was up-regulated in lung tissues of fibrosis model mice and alveolar epithelial cells. Nogo-B knockdown significantly attenuated lung fibrogenesis, downregulated the levels of inflammatory cytokines, inhibited EMT as well as decreased the level of phosphor-PERK/PERK but not the levels of phosphor-IRE1α/IRE1α and c-ATF6. Additionally, a potential efficacy of PERK blockade was demonstrated in improving the extent of lung fibrosis in model mice. Conclusions This study discovered that involvement of Nogo-B in pulmonary fibrogenesis was associated with the PERK branch of ERS pathway and EMT. Nogo-B could be considered as a potential therapeutic target for the treatment of IPF.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Respiratory and Critical Care Medicine, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Meng Yang
- Department of Geriatrics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Xue-Hui Li
- Department of Respiratory and Critical Care Medicine, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wu-Jian Xu
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medical, Shanghai, China
| | - Wei Gao
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medical, Shanghai, China
| | - Yu-Han Chen
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ Failure, Affiliated Bayi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jian-Dong Li
- Department of Respiratory and Critical Care Medicine, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiang Li
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medical, Shanghai, China
| |
Collapse
|
15
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020; 177:4825-4844. [PMID: 32333398 PMCID: PMC7572451 DOI: 10.1111/bph.15082] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
| | - Paul A. Insel
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
- Department of MedicineUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
16
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020. [PMID: 32333398 DOI: 10.1111/bph.15082.10.1111/bph.15082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Paul A Insel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Li Y, Li X, Zhou W, Yu Q, Lu Y. ORMDL3 modulates airway epithelial cell repair in children with asthma under glucocorticoid treatment via regulating IL-33. Pulm Pharmacol Ther 2020; 64:101963. [PMID: 33035699 DOI: 10.1016/j.pupt.2020.101963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/30/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Study found that glucocorticoids, as first-line treatments for asthma, fails to prevent asthma recurrence. Orosomucoid-like (ORMDL) 3 is associated to childhood asthma onset and involved in the inflammation and repair of airway epithelium. We explored the functional role of ORMDL3 in glucocorticoid treatment for childhood asthma. METHODS Mice were sensitized with Ovalbumin (OVA) and treated with Dexamethasone (Dex), followed by OVA challenge to establish a mouse model of asthma. Histopathological changes in lung tissues were observed by hematoxylin-eosin and masson staining. Human bronchial epithelial (16HBE-14°) cells were transfected with ORMDL3 overexpression plasmid and siRNA-interleukin (IL)-33 alone or in combination, followed by Dex. Cell viability was measured by MTT assay. Cell migration was evaluated by wound healing assay. The expressions of E-cadherin and Vimentin and the activation of NF-κB and MAPK/ERK in 16HBE-14° cells were assessed by Western blot. The expressions of ORMDL3 and IL-33 in lung tissues and 16HBE-14° cells were analyzed by qRT-PCR or Western blot. RESULTS Dex treatment alleviated the histopathological abnormality and reversed the overexpressions of ORMDL3 and IL-33 in the lung tissues of asthmatic mice. Overexpressed ORMDL3 enhanced migration and viability, decreased E-cadherin level, increased the levels of IL-33 and Vimentin, and promoted the phosphorylation of NF-κB and MAPK/ERK in Dex-treated 16HBE-14° cells, thus reversing the effect of Dex treatment. However, siRNA-IL-33 inhibited viability and migration, increased E-cadherin level, decreased Vimentin level, and suppressed the phosphorylation of NF-κB and MAPK/ERK, thus reversing the effect of overexpressed ORMDL3 in Dex-treated 16HBE-14° cells. CONCLUSION ORMDL3 overexpression helped airway epithelial cellrepairin asthma via regulating IL-33 expression.
Collapse
Affiliation(s)
- Yaqin Li
- Department of Pediatrics, South Campus, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No.2000, Jiangyue Road, Pujiang, Minhang District, Shanghai, 201112, China
| | - Xiaoyan Li
- Department of Pediatrics, South Campus, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No.2000, Jiangyue Road, Pujiang, Minhang District, Shanghai, 201112, China
| | - Wenjing Zhou
- Department of Pediatrics, South Campus, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No.2000, Jiangyue Road, Pujiang, Minhang District, Shanghai, 201112, China
| | - Qing Yu
- Department of Pediatrics, South Campus, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No.2000, Jiangyue Road, Pujiang, Minhang District, Shanghai, 201112, China
| | - Yanming Lu
- Department of Pediatrics, South Campus, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No.2000, Jiangyue Road, Pujiang, Minhang District, Shanghai, 201112, China.
| |
Collapse
|
18
|
He W, Sun J, Zhang Q, Li Y, Fu Y, Zheng Y, Jiang X. Andrographolide exerts anti-inflammatory effects in Mycobacterium tuberculosis-infected macrophages by regulating the Notch1/Akt/NF-κB axis. J Leukoc Biol 2020; 108:1747-1764. [PMID: 32991757 DOI: 10.1002/jlb.3ma1119-584rrr] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis is a serious public health problem aggravated by the slow progress in the development of new anti-tuberculosis drugs. The hyper-reactive TB patients have suffered from chronic inflammation which could cause deleterious effects on their bodies. Therefore, it is imperative to develop an adjunctive therapy based on inflammatory modulation during Mycobacterium tuberculosis (Mtb) infection. The present study aims to investigate the immune regulatory effects of Andrographolide (Andro) on Mtb-infected macrophages and its underlying mechanisms. The results showed that Andro inhibits the production of IL-1β and other inflammatory cytokines in a dose-dependent manner. The down-regulation of IL-1β expression causes the declining expression of IL-8 and MCP-1 in lung epithelial cells which were co-cultured with Mtb-infected macrophages. The inhibition of the activation of NF-κB pathway, but not the inhibition of MAPK signaling pathway, accounts for the anti-inflammatory role of Andro. Further studies elucidated that Andro could evoke the activation of autophagy to degrade NLRP3, which ultimately inhibited inflammasome activation and subsequent IL-1β production. Finally, the relevant results demonstrated that Andro inhibited the Notch1 pathway to down-regulate the phosphorylation of Akt/mTOR and NF-κB p65 subunit. Taken together, Andro has been found to suppress the Notch1/Akt/NF-κB signaling pathway. Both Akt inhibition-induced autophagy and inhibition of the NF-κB pathway contributed to restraining the activation of NLRP3 inflammasome and subsequent IL-1β production. Then, the decreased production of IL-1β influenced chemokine expression in lung epithelial cells. Based on these results, anti-inflammatory effect of Andro in TB infection is merit further investigation.
Collapse
Affiliation(s)
- Weigang He
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Jinxia Sun
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Qingwen Zhang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.,Department of Inspection and Quarantine, School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, P.R. China
| | - Yinhong Li
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yan Fu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Xin Jiang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| |
Collapse
|
19
|
Kheiry M, Dianat M, Badavi M, Mard SA, Bayati V. p-Coumaric Acid Attenuates Lipopolysaccharide-Induced Lung Inflammation in Rats by Scavenging ROS Production: an In Vivo and In Vitro Study. Inflammation 2020; 42:1939-1950. [PMID: 31267276 DOI: 10.1007/s10753-019-01054-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS), known as lipoglycans and endotoxins found in the cell wall of some type of Gram-negative bacteria, causes acute lung inflammation (ALI). p-Coumaric acid (p-CA) possesses anti-inflammatory and anti-oxidative activities. The main purpose of our research was to explore the effect of p-CA on LPS-induced inflammation. In part I, 32 rats were divided into four groups: Control, LPS (5 mg/kg), p-CA (100 mg/kg), and LPS + p-CA to investigate acute lung inflammation caused by LPS. In part II, the effect of LPS-stimulated inflammatory response on A549 cells was investigated. The dosage of LPS and p-CA which used in this part was 1 μg/ml and 20 mM, respectively. ALI rats showed an elevation in antioxidant activity, TNF-alpha, IL-6, MDA, inflammatory parameters, and Nrf2 gene expression. Although pre-treatment with p-CA could return these changes approximately to normal condition in all two-part studies (in vivo and in vitro). The results of in vivo and in vitro study showed that LPS induced lung inflammation. Pre-treatment with p-CA causes modulating of oxidative stress in inflammatory condition in lung injury and A549 cell.
Collapse
Affiliation(s)
- Maryam Kheiry
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Boulevard, Ahvaz, Iran
| | - Mahin Dianat
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Boulevard, Ahvaz, Iran.
| | - Mohammad Badavi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Boulevard, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Boulevard, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
20
|
Ramírez-Labrada AG, Isla D, Artal A, Arias M, Rezusta A, Pardo J, Gálvez EM. The Influence of Lung Microbiota on Lung Carcinogenesis, Immunity, and Immunotherapy. Trends Cancer 2020; 6:86-97. [PMID: 32061309 DOI: 10.1016/j.trecan.2019.12.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Microbiota have emerged as key modulators of both the carcinogenic process and the immune response against cancer cells, and, thus, it seems to influence the efficacy of immunotherapy. While most studies have focused on analyzing the influence of gut microbiota, its composition substantially differs from that in the lung. Here, we describe how microbial life in the lungs is associated with host immune status in the lungs and, thus, how the identification of the microbial populations in the lower respiratory tract rather than in the gut might be key to understanding the lung carcinogenic process and to predict the efficacy of different treatments. Understanding the influence of lung microbiota on host immunity may identify new therapeutic targets and help to design new immunotherapy approaches to treat lung cancer.
Collapse
Affiliation(s)
- Ariel G Ramírez-Labrada
- Unidad de Nanotoxicología e Inmunotoxicología (UNATI), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Centro de Investigación Biomédica de Aragón (CIBA), Zaragoza, Spain
| | - Dolores Isla
- Medical Oncology Department, Instituto de Investigación Sanitaria Aragón, Hospital Clinico Universitario Lozano Blesa, Zaragoza, Spain
| | - Angel Artal
- Medical Oncology Department, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Maykel Arias
- Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
| | - Antonio Rezusta
- Department of Microbiology, Hospital Universitario Miguel Servet, Zaragoza, Spain; Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Julián Pardo
- Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain; Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Eva M Gálvez
- Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain.
| |
Collapse
|
21
|
Tian Y, Jin Z, Zhu P, Liu S, Zhang D, Tang M, Wang Y, Li D, Yan D, Li G, Zhu X. TRIM59: A membrane protein expressed on Bacillus Calmette-Guérin-activated macrophages that induces apoptosis of fibrosarcoma cells by direct contact. Exp Cell Res 2019; 384:111590. [DOI: 10.1016/j.yexcr.2019.111590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/25/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022]
|
22
|
Exosomal miRNAs in Lung Diseases: From Biologic Function to Therapeutic Targets. J Clin Med 2019; 8:jcm8091345. [PMID: 31470655 PMCID: PMC6781233 DOI: 10.3390/jcm8091345] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests the potential role of extracellular vesicles (EVs) in many lung diseases. According to their subcellular origin, secretion mechanism, and size, EVs are currently classified into three subpopulations: exosomes, microvesicles, and apoptotic bodies. Exosomes are released in most biofluids, including airway fluids, and play a key role in intercellular communication via the delivery of their cargo (e.g., microRNAs (miRNAs)) to target cell. In a physiological context, lung exosomes present protective effects against stress signals which allow them to participate in the maintenance of lung homeostasis. The presence of air pollution alters the composition of lung exosomes (dysregulation of exosomal miRNAs) and their homeostatic property. Indeed, besides their potential as diagnostic biomarkers for lung diseases, lung exosomes are functional units capable of dysregulating numerous pathophysiological processes (including inflammation or fibrosis), resulting in the promotion of lung disease progression. Here, we review recent studies on the known and potential role of lung exosomes/exosomal miRNAs, in the maintaining of lung homeostasis on one hand, and in promoting lung disease progression on the other. We will also discuss using exosomes as prognostic/diagnostic biomarkers as well as therapeutic tools for lung diseases.
Collapse
|
23
|
Dong Y, Glaser K, Schlegel N, Claus H, Speer CP. An underestimated pathogen: Staphylococcus epidermidis induces pro-inflammatory responses in human alveolar epithelial cells. Cytokine 2019; 123:154761. [PMID: 31226437 DOI: 10.1016/j.cyto.2019.154761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/06/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Conventionally regarded as a harmless skin commensal, Staphylococcus epidermidis accounts for the majority of neonatal late-onset sepsis and is shown to be associated with neonatal inflammatory morbidities, especially bronchopulmonary dysplasia. This study addressed the pro-inflammatory capacity of different S. epidermidis strains on human alveolar epithelial cells. METHODS A549 cell monolayers were stimulated by live bacteria of S. epidermidis RP62A strain (biofilm-positive) and ATCC 12228 strain (biofilm-negative) at a multiplicity of infection ratio of 10 for 24 h. LPS (100 ng/ml) and Pam3CSK4 (1 µg/ml) were used for comparisons. Cell viability was measured by MTT method. The mRNA and protein expression of inflammatory mediators and toll-like receptor (TLR)-2 were assessed using RT-PCR, immunoassays and immunofluorescence. RESULTS Both S. epidermidis strains induced expression of tumor necrosis factor (TNF)-α, IL-1β, interleukin (IL)-6, IL-8, monocyte chemoattractant protein (MCP)-1, interferon γ-induced protein 10 (IP-10) and intercellular adhesion molecule (ICAM)-1, but not IL-10. The stimulatory effect of RP62A exceeded that of LPS (p < 0.05). RP62A strain showed a trend towards higher induction of pro-inflammatory mediators than ATCC 12228 strain. The co-stimulation with RP62A strain decreased cell viability compared to control and TLR agonists (p < 0.05). RP62A but not ATCC 12228 stimulated mRNA and protein expression of TLR2. CONCLUSIONS S. epidermidis drives pro-inflammatory responses in lung epithelial cells in vitro. The pro-inflammatory capacity of S. epidermidis may differ between strains. Biofilm-positive S. epidermidis strain seems to induce more potent pulmonary pro-inflammation than biofilm-negative S. epidermidis strain.
Collapse
Affiliation(s)
- Ying Dong
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany.
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Nicolas Schlegel
- Department of Surgery I, University of Wuerzburg, Wuerzburg, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Wuerzburg, Germany
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
24
|
The human lung mucosa drives differential Mycobacterium tuberculosis infection outcome in the alveolar epithelium. Mucosal Immunol 2019; 12:795-804. [PMID: 30846830 PMCID: PMC6462240 DOI: 10.1038/s41385-019-0156-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/10/2019] [Accepted: 02/25/2019] [Indexed: 02/08/2023]
Abstract
Mycobacterium tuberculosis (M.tb) is deposited into the alveolus where it first encounters the alveolar lining fluid (ALF) prior contacts host cells. We demonstrated that M.tb-exposure to human ALF alters its cell surface, driving better M.tb infection control by professional phagocytes. Contrary to these findings, our results with non-professional phagocytes alveolar epithelial cells (ATs) define two distinct subsets of human ALFs; where M.tb exposure to Low (L)-ALF or High(H)-ALF results in low or high intracellular bacterial growth rates in ATs, respectively. H-ALF exposed-M.tb growth within ATs was independent of M.tb-uptake, M.tb-trafficking, and M.tb-infection induced cytotoxicity; however, it was associated with enhanced bacterial replication within LAMP-1+/ABCA1+ compartments. H-ALF exposed-M.tb infection of ATs decreased AT immune mediator production, decreased AT surface adhesion expression, and downregulated macrophage inflammatory responses. Composition analysis of H-ALF vs. L-ALF showed H-ALF with higher protein tyrosine nitration and less functional ALF-innate proteins important in M.tb pathogenesis. Replenishment of H-ALF with functional ALF-innate proteins reversed the H-ALF-M.tb growth rate to the levels observed for L-ALF-M.tb. These results indicate that dysfunctionality of innate proteins in the H-ALF phenotype promotes M.tb replication within ATs, while limiting inflammation and phagocyte activation, thus potentiating ATs as a reservoir for M.tb replication and survival.
Collapse
|
25
|
Significance of CCL2 (−2518A/G), CCR2 (190G/A) and TLR4 polymorphisms (896 A/G and 1196C/T) in tuberculosis risk in Indian population. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
26
|
Haggadone MD, Peters-Golden M. Microenvironmental Influences on Extracellular Vesicle-Mediated Communication in the Lung. Trends Mol Med 2018; 24:963-975. [DOI: 10.1016/j.molmed.2018.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/27/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022]
|
27
|
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
|
28
|
Hadifar S, Behrouzi A, Fateh A, Khatami S, Rahimi Jamnani F, Siadat SD, Vaziri F. Comparative study of interruption of signaling pathways in lung epithelial cell by two different Mycobacterium tuberculosis lineages. J Cell Physiol 2018; 234:4739-4753. [PMID: 30192006 DOI: 10.1002/jcp.27271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022]
Abstract
Alveolar epithelial cell (AEC) provides a replication niche for Mycobacterium tuberculosis. Based on the role of AEC in M. tuberculosis pathogenesis and existence of genetic diversity within this bacterium, we investigated interactions between AEC II and two different M. tuberculosis lineages. We have compared the transcriptome and cytokines/chemokines levels of A549 infected by M. tuberculosis lineage three and four using qRT-PCR and ELISA arrays, respectively. We showed different M. tuberculosis strains induced changes in different effectors that involved in TLRs and NF-κB signaling pathways. We observed different reaction of the studied lineages specifically in pathogenesis, immune evasion mechanism, IL-12/IFN-γ axis, and autophagy. Similar behavior was detected in regarding to apoptosis, necroptosis, anti-inflammatory responses, and canonical inflammasome. Our findings contribute to elucidate more details in pathogenesis, immune evasion strategies, novel target and druggable pathway for therapeutic intervention, and host directed therapy in tuberculosis infection. Also, different M. tuberculosis lineages-dependent host-pathogen interactions suggested using only one strain for this kind of research will be controversial.
Collapse
Affiliation(s)
- Shima Hadifar
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rahimi Jamnani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
29
|
Adam L, López-González M, Björk A, Pålsson S, Poux C, Wahren-Herlenius M, Fernández C, Spetz AL. Early Resistance of Non-virulent Mycobacterial Infection in C57BL/6 Mice Is Associated With Rapid Up-Regulation of Antimicrobial Cathelicidin Camp. Front Immunol 2018; 9:1939. [PMID: 30233570 PMCID: PMC6129578 DOI: 10.3389/fimmu.2018.01939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/06/2018] [Indexed: 01/04/2023] Open
Abstract
Early clearance of tuberculosis is the successful eradication of inhaled bacteria before the development of an adaptive immune response. We previously showed, by utilizing a non-virulent mycobacteria infection model, that C57BL/6 mice are more efficient than BALB/c in their control of bacterial growth in the lungs during the first weeks of the infection. Here, we assessed early (within 1–3 days) innate immune events locally in the lungs to identify factors that may contribute to the control of non-virulent mycobacterial burden. We confirmed that C57BL/6 mice are more resistant to infection compared with BALB/c after intranasal inoculation with mycobacterium. Transcriptomic analyses revealed a remarkably silent signature in C57BL/6 mice despite effective control of bacterial growth. In contrast, BALB/c mice up-regulated genes associated with neutrophil and myeloid cell chemotaxis and migration. Flow cytometry analyses corroborated the transcriptomic analyses and demonstrated influx of both neutrophil and myeloid cell populations in BALB/c mice, while these did not increase in C57BL/6 mice. We further detected increased release of TNF-α from BALB/c lung cells but limited release from C57BL/6-derived cells. However, C57BL/6 mice showed a marked early up-regulation of the Camp gene, encoding the cathelicidin CRAMP peptide, post-mycobacterial exposure. CRAMP (LL-37 in human) expression in the lungs was confirmed using immunofluorescence staining. Altogether, these findings show that C57BL/6 mice can clear the mycobacterial infection early and that this early control is associated with high CRAMP expression in the lungs without concomitant influx of immune cells. The role of CRAMP/LL-37 during mycobacterial infection may be relevant for novel protective strategies, and warrants further studies of human cohorts.
Collapse
Affiliation(s)
- Lucille Adam
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Moisés López-González
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Albin Björk
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Pålsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Candice Poux
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Carmen Fernández
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
30
|
Bacterial Pore-Forming Toxins Promote the Activation of Caspases in Parallel to Necroptosis to Enhance Alarmin Release and Inflammation During Pneumonia. Sci Rep 2018; 8:5846. [PMID: 29643440 PMCID: PMC5895757 DOI: 10.1038/s41598-018-24210-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/26/2018] [Indexed: 12/18/2022] Open
Abstract
Pore-forming toxins are the most common virulence factor in pathogenic bacteria. They lead to membrane permeabilization and cell death. Herein, we show that respiratory epithelial cells (REC) undergoing bacterial pore-forming toxin (PFT)-induced necroptosis simultaneously experienced caspase activation independently of RIPK3. MLKL deficient REC treated with a pan-caspase inhibitor were protected in an additive manner against PFT-induced death. Subsequently, cleaved versions of caspases-2, -4 and -10 were detected within REC undergoing necroptosis by immunoblots and monoclonal antibody staining. Caspase activation was observed in lung samples from mice and non-human primates experiencing Gram-negative and Gram-positive bacterial pneumonia, respectively. During apoptosis, caspase activation normally leads to cell shrinkage, nuclear condensation, and immunoquiescent death. In contrast, caspase activity during PFT-induced necroptosis increased the release of alarmins to the extracellular milieu. Caspase-mediated alarmin release was found sufficient to activate resting macrophages, leading to Interleukin-6 production. In a mouse model of Gram-negative pneumonia, deletion of caspases -2 and -11, the mouse orthologue of caspase-4, reduced pulmonary inflammation, immune cell infiltration and lung damage. Thus, our study describes a previously unrecognized role for caspase activation in parallel to necroptosis, and indicates that their activity plays a critical pro-inflammatory role during bacterial pneumonia.
Collapse
|
31
|
Schwotzer D, Niehof M, Schaudien D, Kock H, Hansen T, Dasenbrock C, Creutzenberg O. Cerium oxide and barium sulfate nanoparticle inhalation affects gene expression in alveolar epithelial cells type II. J Nanobiotechnology 2018; 16:16. [PMID: 29463257 PMCID: PMC5819288 DOI: 10.1186/s12951-018-0343-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/13/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Understanding the molecular mechanisms of nanomaterial interacting with cellular systems is important for appropriate risk assessment. The identification of early biomarkers for potential (sub-)chronic effects of nanoparticles provides a promising approach towards cost-intensive and animal consuming long-term studies. As part of a 90-day inhalation toxicity study with CeO2 NM-212 and BaSO4 NM-220 the present investigations on gene expression and immunohistochemistry should reveal details on underlying mechanisms of pulmonary effects. The role of alveolar epithelial cells type II (AEII cells) is focused since its contribution to defense against inhaled particles and potentially resulting adverse effects is assumed. Low dose levels should help to specify particle-related events, including inflammation and oxidative stress. RESULTS Rats were exposed to clean air, 0.1, 0.3, 1.0, and 3.0 mg/m3 CeO2 NM-212 or 50.0 mg/m3 BaSO4 NM-220 and the expression of 391 genes was analyzed in AEII cells after one, 28 and 90 days exposure. A total number of 34 genes was regulated, most of them related to inflammatory mediators. Marked changes in gene expression were measured for Ccl2, Ccl7, Ccl17, Ccl22, Ccl3, Ccl4, Il-1α, Il-1ß, and Il-1rn (inflammation), Lpo and Noxo1 (oxidative stress), and Mmp12 (inflammation/lung cancer). Genes related to genotoxicity and apoptosis did not display marked regulation. Although gene expression was less affected by BaSO4 compared to CeO2 the gene pattern showed great overlap. Gene expression was further analyzed in liver and kidney tissue showing inflammatory responses in both organs and marked downregulation of oxidative stress related genes in the kidney. Increases in the amount of Ce were measured in liver but not in kidney tissue. Investigation of selected genes on protein level revealed increased Ccl2 in bronchoalveolar lavage of exposed animals and increased Lpo and Mmp12 in the alveolar epithelia. CONCLUSION AEII cells contribute to CeO2 nanoparticle caused inflammatory and oxidative stress reactions in the respiratory tract by the release of related mediators. Effects of BaSO4 exposure are low. However, overlap between both substances were detected and support identification of potential early biomarkers for nanoparticle effects on the respiratory system. Signs for long-term effects need to be further evaluated by comparison to a respective exposure setting.
Collapse
Affiliation(s)
- Daniela Schwotzer
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany.
| | - Monika Niehof
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Heiko Kock
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Clemens Dasenbrock
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Otto Creutzenberg
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| |
Collapse
|
32
|
Yang R, Yang E, Shen L, Modlin RL, Shen H, Chen ZW. IL-12+IL-18 Cosignaling in Human Macrophages and Lung Epithelial Cells Activates Cathelicidin and Autophagy, Inhibiting Intracellular Mycobacterial Growth. THE JOURNAL OF IMMUNOLOGY 2018; 200:2405-2417. [PMID: 29453279 DOI: 10.4049/jimmunol.1701073] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/25/2018] [Indexed: 11/19/2022]
Abstract
The ability of Mycobacterium tuberculosis to block host antimicrobial responses in infected cells provides a key mechanism for disease pathogenesis. The immune system has evolved to overcome this blockade to restrict the infection, but it is not clear whether two key innate cytokines (IL-12/IL-18) involved in host defense can enhance antimycobacterial mechanisms. In this study, we demonstrated that the combination of IL-12 and IL-18 triggered an antimicrobial response against mycobacteria in infected macrophages (THP-1 and human primary monocyte-derived macrophages) and pulmonary epithelial A549 cells. The inhibition of intracellular bacterial growth required p38-MAPK and STAT4 pathways, the vitamin D receptor, the vitamin D receptor-derived antimicrobial peptide cathelicidin, and autophagy, but not caspase-mediated apoptosis. Finally, the ability of IL-12+IL-18 to activate an innate antimicrobial response in human primary macrophages was dependent on the autonomous production of IFN-γ and the CAMP/autophagy pathway. Together, these data suggest that IL-12+IL-18 cosignaling can trigger the antimicrobial protein cathelicidin and autophagy, resulting in inhibition of intracellular mycobacteria in macrophages and lung epithelial cells.
Collapse
Affiliation(s)
- Rui Yang
- Unit of Anti-Tuberculosis Immunity, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - Enzhuo Yang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL 60612
| | - Ling Shen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL 60612
| | - Robert L Modlin
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095.,Division of Dermatology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095; and
| | - Hongbo Shen
- Unit of Anti-Tuberculosis Immunity, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China;
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL 60612.,Institut Pasteur of Shanghai, Shanghai 200031, China
| |
Collapse
|
33
|
Mvubu NE, Pillay B, McKinnon LR, Pillay M. Mycobacterium tuberculosis strains induce strain-specific cytokine and chemokine response in pulmonary epithelial cells. Cytokine 2017; 104:53-64. [PMID: 29032986 DOI: 10.1016/j.cyto.2017.09.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/23/2017] [Accepted: 09/23/2017] [Indexed: 02/07/2023]
Abstract
M. tuberculosis F15/LAM4/KZN has been associated with high transmission rates of drug resistant tuberculosis in the KwaZulu-Natal province of South Africa. The current study elucidated the cytokine/chemokine responses induced by representatives of the F15/LAM4/KZN and other dominant strain families in pulmonary epithelial cells. Multiplex cytokine analyses were performed at 24, 48 and 72h post infection of the A549 pulmonary epithelial cell line with the F15/LAM4/KZN, F28, F11, Beijing, Unique and H37Rv strains at an MOI of ∼10:1. Twenty-three anti- and pro-inflammatory cytokines/chemokines were detected at all-time intervals. Significantly high concentrations of IL-6, IFN-γ, TNF-α and G-CSF at 48h, and IL-8, IFN-γ, TNF-α, G-CSF and GM-CSF at 72h, were induced by the F28 and F15/LAM4/KZN strains, respectively. Lower levels of cytokines/chemokines were induced by either the Beijing or Unique strains at all three time intervals. All strains induced up-regulation of pathogen recognition receptors (PRRs) (TLR3 and TLR5) while only the F15/LAM4/KZN, F11 and F28 strains induced significant differential expression of TLR2 compared to the Beijing, Unique and H37Rv strains. The low induction of cytokines in epithelial cells by the Beijing strain correlates with its previously reported hypervirulent properties. High concentrations of cytokines and chemokines required for early protection against M. tuberculosis infections induced by the F15/LAM4/KZN and F28 strains suggests a lower virulence of these genotypes compared to the Beijing strain. These findings demonstrate the high diversity in host cytokine/chemokine response to early infection of pulmonary epithelial cells by different strains of M. tuberculosis.
Collapse
Affiliation(s)
- Nontobeko E Mvubu
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Balakrishna Pillay
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Lyle R McKinnon
- Centre for the AIDS Program of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa; Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Manormoney Pillay
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 719 Umbilo Road, South Africa.
| |
Collapse
|
34
|
Zhang D, Lee H, Haspel JA, Jin Y. Long noncoding RNA FOXD3-AS1 regulates oxidative stress-induced apoptosis via sponging microRNA-150. FASEB J 2017; 31:4472-4481. [PMID: 28655711 PMCID: PMC5602897 DOI: 10.1096/fj.201700091r] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
Abstract
The function of most human long noncoding RNAs (lncRNAs) remains unclear. Our studies identified a highly up-regulated mammalian lncRNA, FOXD3-AS1, known as linc1623 in mice, in the setting of hyperoxia/reactive oxygen species (ROS)-induced lung injury. We found that ROS induced a robust expression of FOXD3-AS1 in mouse lung tissue. Functionally, FOXD3-AS1 promoted oxidative stress-induced lung epithelial cell death. In human lung epithelial cells, the microRNA-150 (miR-150) was identified to interact with FOXD3-AS1; this finding was confirmed using the luciferase reporter assays. Consistently, mutation on the miR-150 pairing sequence in FOXD3-AS1 abolished the interactions between FOXD3-AS1 and miR-150. Additionally, miR-150 mimics suppressed the level of FOXD3-AS1. The antisense oligos of FOXD3-AS1 significantly augmented the intracellular level of miR-150, supporting the theory of sponging effects of FOXD3-AS1 on miR-150. We further investigated the cellular function of miR-150 in our lung injury models. MiR-150 conferred a cytoprotective role in lung epithelial cells after oxidative stress, whereas FOXD3-AS1 promoted cell death. Taken together, our studies indicated that FOXD3-AS1 serves as a sponge or as a competing endogenous noncoding RNA for miR-150, restricting its capability to promote cell growth and thereby exaggerating hyperoxia-induced lung epithelial cell death.-Zhang, D., Lee, H., Haspel, J. A., Jin, Y. Long noncoding RNA FOXD3-AS1 regulates oxidative stress-induced apoptosis via sponging microRNA-150.
Collapse
Affiliation(s)
- Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Jeffrey A Haspel
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, Massachusetts, USA;
| |
Collapse
|
35
|
Reuschl AK, Edwards MR, Parker R, Connell DW, Hoang L, Halliday A, Jarvis H, Siddiqui N, Wright C, Bremang S, Newton SM, Beverley P, Shattock RJ, Kon OM, Lalvani A. Innate activation of human primary epithelial cells broadens the host response to Mycobacterium tuberculosis in the airways. PLoS Pathog 2017; 13:e1006577. [PMID: 28863187 PMCID: PMC5605092 DOI: 10.1371/journal.ppat.1006577] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 09/19/2017] [Accepted: 08/11/2017] [Indexed: 01/17/2023] Open
Abstract
Early events in the human airways determining whether exposure to Mycobacterium tuberculosis (Mtb) results in acquisition of infection are poorly understood. Epithelial cells are the dominant cell type in the lungs, but little is known about their role in tuberculosis. We hypothesised that human primary airway epithelial cells are part of the first line of defense against Mtb-infection and contribute to the protective host response in the human respiratory tract. We modelled these early airway-interactions with human primary bronchial epithelial cells (PBECs) and alveolar macrophages. By combining in vitro infection and transwell co-culture models with a global transcriptomic approach, we identified PBECs to be inert to direct Mtb-infection, yet to be potent responders within an Mtb-activated immune network, mediated by IL1β and type I interferon (IFN). Activation of PBECs by Mtb-infected alveolar macrophages and monocytes increased expression of known and novel antimycobacterial peptides, defensins and S100-family members and epithelial-myeloid interactions further shaped the immunological environment during Mtb-infection by promoting neutrophil influx. This is the first in depth analysis of the primary epithelial response to infection and offers new insights into their emerging role in tuberculosis through complementing and amplifying responses to Mtb. Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis, which remains a major public health burden today. In the majority of cases, infection is acquired by inhalation of aerosolised bacteria. Mtb is thought to target alveolar macrophages in the lower airways to establish infection. However, the cells predominantly lining the respiratory tract are epithelial cells and thus are likely crucial during the early host-pathogen interactions. We recovered primary human bronchial epithelial cells from healthy volunteers to assess their global transcriptomic response to direct Mtb-exposure and exposure to Mtb-infected myeloid cells. Our analysis revealed that, while being inert to direct Mtb-infection, epithelial cells were highly responsive to soluble mediators released by infected macrophages. The epithelial response induced by this cellular cross-talk, promoted neutrophil influx in vitro as well as the increase of antimycobaterial host responses. Our data provide novel and unexpected insights into the role of the primary human airway epithelium and define a non-redundant role for epithelial cells in shaping the local immunological environment at the site of initial Mtb infection.
Collapse
Affiliation(s)
- Ann-Kathrin Reuschl
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Michael R. Edwards
- Department of Cytopathology, Imperial College London, St Mary’s Hospital, Imperial College NHS Trust, London, United Kingdom
| | - Robert Parker
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - David W. Connell
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Long Hoang
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Alice Halliday
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Hannah Jarvis
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Nazneen Siddiqui
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Corrina Wright
- Respiratory Medicine, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, Norfolk Place, London, United Kingdom
| | - Samuel Bremang
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Sandra M. Newton
- Section of Paediatrics, Department of Medicine, St Mary’s Campus, Imperial College, London, United Kingdom
| | - Peter Beverley
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Robin J. Shattock
- Department of Medicine, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Onn Min Kon
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Ajit Lalvani
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, St Mary’s Campus, London, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Wirsdörfer F, Jendrossek V. Modeling DNA damage-induced pneumopathy in mice: insight from danger signaling cascades. Radiat Oncol 2017; 12:142. [PMID: 28836991 PMCID: PMC5571607 DOI: 10.1186/s13014-017-0865-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/07/2017] [Indexed: 02/08/2023] Open
Abstract
Radiation-induced pneumonitis and fibrosis represent severe and dose-limiting side effects in the radiotherapy of thorax-associated neoplasms leading to decreased quality of life or - as a consequence of treatment with suboptimal radiation doses - to fatal outcomes by local recurrence or metastatic disease. It is assumed that the initial radiation-induced damage to the resident cells triggers a multifaceted damage-signalling cascade in irradiated normal tissues including a multifactorial secretory program. The resulting pro-inflammatory and pro-angiogenic microenvironment triggers a cascade of events that can lead within weeks to a pronounced lung inflammation (pneumonitis) or after months to excessive deposition of extracellular matrix molecules and tissue scarring (pulmonary fibrosis).The use of preclinical in vivo models of DNA damage-induced pneumopathy in genetically modified mice has helped to substantially advance our understanding of molecular mechanisms and signalling molecules that participate in the pathogenesis of radiation-induced adverse late effects in the lung. Herein, murine models of whole thorax irradiation or hemithorax irradiation nicely reproduce the pathogenesis of the human disease with respect to the time course and the clinical symptoms. Alternatively, treatment with the radiomimetic DNA damaging chemotherapeutic drug Bleomycin (BLM) has frequently been used as a surrogate model of radiation-induced lung disease. The advantage of the BLM model is that the symptoms of pneumonitis and fibrosis develop within 1 month.Here we summarize and discuss published data about the role of danger signalling in the response of the lung tissue to DNA damage and its cross-talk with the innate and adaptive immune systems obtained in preclinical studies using immune-deficient inbred mouse strains and genetically modified mice. Interestingly we observed differences in the role of molecules involved in damage sensing (TOLL-like receptors), damage signalling (MyD88) and immune regulation (cytokines, CD73, lymphocytes) for the pathogenesis and progression of DNA damage-induced pneumopathy between the models of pneumopathy induced by whole thorax irradiation or treatment with the radiomimetic drug BLM. These findings underline the importance to pursue studies in the radiation model(s) if we are to unravel the mechanisms driving radiation-induced adverse late effects.A better understanding of the cross-talk of danger perception and signalling with immune activation and repair mechanisms may allow a modulation of these processes to prevent or treat radiation-induced adverse effects. Vice-versa an improved knowledge of the normal tissue response to injury is also particularly important in view of the increasing interest in combining radiotherapy with immune checkpoint blockade or immunotherapies to avoid exacerbation of radiation-induced normal tissue toxicity.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 173, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 173, Essen, Germany.
| |
Collapse
|
37
|
Oczypok EA, Perkins TN, Oury TD. Alveolar Epithelial Cell-Derived Mediators: Potential Direct Regulators of Large Airway and Vascular Responses. Am J Respir Cell Mol Biol 2017; 56:694-699. [PMID: 28080134 DOI: 10.1165/rcmb.2016-0151ps] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchial epithelial cells and pulmonary endothelial cells are thought to be the primary modulators of conducting airways and vessels, respectively. However, histological examination of both mouse and human lung tissue reveals that alveolar epithelial cells (AECs) line the adventitia of large airways and vessels and thus are also in a position to directly regulate these structures. The primary purpose of this perspective is to highlight the fact that AECs coat the adventitial surface of every vessel and airway in the lung parenchyma. This localization is ideal for transmitting signals that can contribute to physiologic and pathologic responses in vessels and airways. A few examples of mediators produced by AECs that may contribute to vascular and airway responses are provided to illustrate some of the potential effects that AECs may modulate.
Collapse
Affiliation(s)
- Elizabeth A Oczypok
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Lee SM, Park HY, Suh YS, Yoon EH, Kim J, Jang WH, Lee WS, Park SG, Choi IW, Choi I, Kang SW, Yun H, Teshima T, Kwon B, Seo SK. Inhibition of acute lethal pulmonary inflammation by the IDO-AhR pathway. Proc Natl Acad Sci U S A 2017; 114:E5881-E5890. [PMID: 28673995 PMCID: PMC5530642 DOI: 10.1073/pnas.1615280114] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The lung is a prototypic organ that was evolved to reduce immunopathology during the immune response to potentially hazardous endogenous and exogenous antigens. In this study, we show that donor CD4+ T cells transiently induced expression of indoleamine 2,3-dioxygenase (IDO) in lung parenchyma in an IFN-γ-dependent manner early after allogeneic hematopoietic stem cell transplantation (HSCT). Abrogation of host IDO expression by deletion of the IDO gene or the IFN-γ gene in donor T cells or by FK506 treatment resulted in acute lethal pulmonary inflammation known as idiopathic pneumonia syndrome (IPS). Interestingly, IL-6 strongly induced IDO expression in an IFN-γ-independent manner when deacetylation of STAT3 was inhibited. Accordingly, a histone deacetylase inhibitor (HDACi) could reduce IPS in the state where IFN-γ expression was suppressed by FK506. Finally, l-kynurenine produced by lung epithelial cells and alveolar macrophages during IPS progression suppresses the inflammatory activities of lung epithelial cells and CD4+ T cells through the aryl hydrocarbon receptor pathway. Taken together, our results reveal that IDO is a critical regulator of acute pulmonary inflammation and that regulation of IDO expression by HDACi may be a therapeutic approach for IPS after HSCT.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/immunology
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Female
- Graft vs Host Disease
- Hematopoietic Stem Cell Transplantation/mortality
- Histone Deacetylase Inhibitors/pharmacology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Kynurenine/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Pneumonia/drug therapy
- Pneumonia/metabolism
- Receptors, Aryl Hydrocarbon/immunology
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/immunology
- Tacrolimus/pharmacology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Soung-Min Lee
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Ha Young Park
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Young-Sill Suh
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Eun Hye Yoon
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Juyang Kim
- Biomedical Research Center and Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Won Hee Jang
- Department of Biochemistry, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Won-Sik Lee
- Department of Hemato/Oncology, Busan Paik Hospital, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Sae-Gwang Park
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Il-Whan Choi
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Inhak Choi
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea
- Advanced Research Center for Multiple Myeloma, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Sun-Woo Kang
- Department of Nephrology, Busan Paik Hospital, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - Hwayoung Yun
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Byungsuk Kwon
- Biomedical Research Center and Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Su-Kil Seo
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Republic of Korea;
| |
Collapse
|
39
|
Scheiermann J, Klinman DM. Three distinct pneumotypes characterize the microbiome of the lung in BALB/cJ mice. PLoS One 2017; 12:e0180561. [PMID: 28683098 PMCID: PMC5500332 DOI: 10.1371/journal.pone.0180561] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/16/2017] [Indexed: 01/05/2023] Open
Abstract
Bacteria can rarely be isolated from normal healthy lungs using conventional culture techniques, supporting the traditional belief that the lungs are sterile. Yet recent studies using next generation sequencing report that bacterial DNA commonly found in the upper respiratory tract (URT) is present at lower levels in the lungs. Interpretation of that finding is complicated by the technical limitations and potential for contamination introduced when dealing with low biomass samples. The current work sought to overcome those limitations to clarify the number, type and source of bacteria present in the lungs of normal mice. Results showed that the oral microbiome is diverse and highly conserved whereas murine lung samples fall into three distinct patterns. 33% of the samples were sterile, as they lacked culturable bacteria and their bacterial DNA content did not differ from background. 9% of samples contained comparatively higher amounts of bacterial DNA whose composition mimicked that detected in the URT. A final group (58%) contained smaller amounts of microbial DNA whose composition was correlating to that of rodent chow and cage bedding, likely acquired by inspiration of food and bedding fragments. By analyzing each sample independently rather than working with group averages, this work eliminated the bias introduced by aspiration-contaminated samples to establish that three distinct microbiome pneumotypes are present in normal murine lungs.
Collapse
Affiliation(s)
- Julia Scheiermann
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland, United States of America
| | - Dennis M. Klinman
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Brazee PL, Soni PN, Tokhtaeva E, Magnani N, Yemelyanov A, Perlman HR, Ridge KM, Sznajder JI, Vagin O, Dada LA. FXYD5 Is an Essential Mediator of the Inflammatory Response during Lung Injury. Front Immunol 2017; 8:623. [PMID: 28620381 PMCID: PMC5451504 DOI: 10.3389/fimmu.2017.00623] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022] Open
Abstract
The alveolar epithelium secretes cytokines and chemokines that recruit immune cells to the lungs, which is essential for fighting infections but in excess can promote lung injury. Overexpression of FXYD5, a tissue-specific regulator of the Na,K-ATPase, in mice, impairs the alveolo-epithelial barrier, and FXYD5 overexpression in renal cells increases C-C chemokine ligand-2 (CCL2) secretion in response to lipopolysaccharide (LPS). The aim of this study was to determine whether FXYD5 contributes to the lung inflammation and injury. Exposure of alveolar epithelial cells (AEC) to LPS increased FXYD5 levels at the plasma membrane, and FXYD5 silencing prevented both the activation of NF-κB and the secretion of cytokines in response to LPS. Intratracheal instillation of LPS into mice increased FXYD5 levels in the lung. FXYD5 overexpression increased the recruitment of interstitial macrophages and classical monocytes to the lung in response to LPS. FXYD5 silencing decreased CCL2 levels, number of cells, and protein concentration in bronchoalveolar lavage fluid (BALF) after LPS treatment, indicating that FXYD5 is required for the NF-κB-stimulated epithelial production of CCL2, the influx of immune cells, and the increase in alveolo-epithelial permeability in response to LPS. Silencing of FXYD5 also prevented the activation of NF-κB and cytokine secretion in response to interferon α and TNF-α, suggesting that pro-inflammatory effects of FXYD5 are not limited to the LPS-induced pathway. Furthermore, in the absence of other stimuli, FXYD5 overexpression in AEC activated NF-κB and increased cytokine production, while FXYD5 overexpression in mice increased cytokine levels in BALF, indicating that FXYD5 is sufficient to induce the NF-κB-stimulated cytokine secretion by the alveolar epithelium. The FXYD5 overexpression also increased cell counts in BALF, which was prevented by silencing the CCL2 receptor (CCR2), or by treating mice with a CCR2-blocking antibody, confirming that FXYD5-induced CCL2 production leads to the recruitment of monocytes to the lung. Taken together, the data demonstrate that FXYD5 is a key contributor to inflammatory lung injury.
Collapse
Affiliation(s)
- Patricia L Brazee
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Pritin N Soni
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Elmira Tokhtaeva
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Natalia Magnani
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alex Yemelyanov
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Harris R Perlman
- Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Karen M Ridge
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob I Sznajder
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Laura A Dada
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
41
|
Klaßen C, Karabinskaya A, Dejager L, Vettorazzi S, Van Moorleghem J, Lühder F, Meijsing SH, Tuckermann JP, Bohnenberger H, Libert C, Reichardt HM. Airway Epithelial Cells Are Crucial Targets of Glucocorticoids in a Mouse Model of Allergic Asthma. THE JOURNAL OF IMMUNOLOGY 2017; 199:48-61. [PMID: 28515280 DOI: 10.4049/jimmunol.1601691] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 04/25/2017] [Indexed: 11/19/2022]
Abstract
Although glucocorticoids (GCs) are a mainstay in the clinical management of asthma, the target cells that mediate their therapeutic effects are unknown. Contrary to our expectation, we found that GC receptor (GR) expression in immune cells was dispensable for successful therapy of allergic airway inflammation (AAI) with dexamethasone. Instead, GC treatment was compromised in mice expressing a defective GR in the nonhematopoietic compartment or selectively lacking the GR in airway epithelial cells. Further, we found that an intact GR dimerization interface was a prerequisite for the suppression of AAI and airway hyperresponsiveness by GCs. Our observation that the ability of dexamethasone to modulate gene expression in airway epithelial cells coincided with its potency to resolve AAI supports a crucial role for transcriptional regulation by the GR in this cell type. Taken together, we identified an unknown mode of GC action in the treatment of allergic asthma that might help to develop more specific therapies in the future.
Collapse
Affiliation(s)
- Carina Klaßen
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Anna Karabinskaya
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Lien Dejager
- Inflammation Research Center, Flanders Institute for Biotechnology, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, University of Ghent, 9052 Ghent, Belgium
| | - Sabine Vettorazzi
- Institute of Comparative Endocrinology, University of Ulm, 89081 Ulm, Germany
| | | | - Fred Lühder
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | | | - Jan P Tuckermann
- Institute of Comparative Endocrinology, University of Ulm, 89081 Ulm, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Claude Libert
- Inflammation Research Center, Flanders Institute for Biotechnology, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, University of Ghent, 9052 Ghent, Belgium
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany;
| |
Collapse
|
42
|
Cardani A, Boulton A, Kim TS, Braciale TJ. Alveolar Macrophages Prevent Lethal Influenza Pneumonia By Inhibiting Infection Of Type-1 Alveolar Epithelial Cells. PLoS Pathog 2017; 13:e1006140. [PMID: 28085958 PMCID: PMC5268648 DOI: 10.1371/journal.ppat.1006140] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/26/2017] [Accepted: 12/19/2016] [Indexed: 12/21/2022] Open
Abstract
The Influenza A virus (IAV) is a major human pathogen that produces significant morbidity and mortality. To explore the contribution of alveolar macrophages (AlvMΦs) in regulating the severity of IAV infection we employed a murine model in which the Core Binding Factor Beta gene is conditionally disrupted in myeloid cells. These mice exhibit a selective deficiency in AlvMΦs. Following IAV infection these AlvMΦ deficient mice developed severe diffuse alveolar damage, lethal respiratory compromise, and consequent lethality. Lethal injury in these mice resulted from increased infection of their Type-1 Alveolar Epithelial Cells (T1AECs) and the subsequent elimination of the infected T1AECs by the adaptive immune T cell response. Further analysis indicated AlvMΦ-mediated suppression of the cysteinyl leukotriene (cysLT) pathway genes in T1AECs in vivo and in vitro. Inhibition of the cysLT pathway enzymes in a T1AECs cell line reduced the susceptibility of T1AECs to IAV infection, suggesting that AlvMΦ-mediated suppression of this pathway contributes to the resistance of T1AECs to IAV infection. Furthermore, inhibition of the cysLT pathway enzymes, as well as blockade of the cysteinyl leukotriene receptors in the AlvMΦ deficient mice reduced the susceptibility of their T1AECs to IAV infection and protected these mice from lethal infection. These results suggest that AlvMΦs may utilize a previously unappreciated mechanism to protect T1AECs against IAV infection, and thereby reduce the severity of infection. The findings further suggest that the cysLT pathway and the receptors for cysLT metabolites represent potential therapeutic targets in severe IAV infection. A primary feature of lethal influenza infection is viral pneumonia. Influenza viral pneumonia is caused by the direct infection of alveolar epithelial cells, which subsequently causes extensive alveolar inflammation and injury. Clinically this pathology manifests as diffuse alveolar damage leading to acute respiratory distress syndrome. As alveolar macrophages are positioned in the alveoli, they are the ideally localized to be a first-line of defense against alveolar invading pathogens, such as influenza. To explore the contribution of alveolar macrophages to the development of lethal influenza pneumonia, we generated a novel mouse model with a selective deficiency in alveolar macrophages. As a result of the alveolar macrophage deficiency, these mice developed severe diffuse alveolar damage and lethal respiratory compromise after influenza infection. Lethal injury resulted from increased infection of type-1 alveolar epithelial cells, and the elimination of these infected cells by effector T cells. Further analysis indicated that in order to render type 1 cells resistant to influenza infection, alveolar macrophages suppress leukotrieneD4 production and autocrine-signaling in type 1 cells. These results suggest that alveolar macrophages play a previously unappreciated role in protecting type 1 alveolar epithelial cells against IAV infection, and thus the severity of infection.
Collapse
Affiliation(s)
- Amber Cardani
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Adam Boulton
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Taeg S. Kim
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
43
|
Mvubu NE, Pillay B, Gamieldien J, Bishai W, Pillay M. Mycobacterium tuberculosis strains exhibit differential and strain-specific molecular signatures in pulmonary epithelial cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 65:321-329. [PMID: 27497873 DOI: 10.1016/j.dci.2016.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/31/2016] [Accepted: 07/31/2016] [Indexed: 06/06/2023]
Abstract
Although pulmonary epithelial cells are integral to innate and adaptive immune responses during Mycobacterium tuberculosis infection, global transcriptomic changes in these cells remain largely unknown. Changes in gene expression induced in pulmonary epithelial cells infected with M. tuberculosis F15/LAM4/KZN, F11, F28, Beijing and Unique genotypes were investigated by RNA sequencing (RNA-Seq). The Illumina HiSeq 2000 platform generated 50 bp reads that were mapped to the human genome (Hg19) using Tophat (2.0.10). Differential gene expression induced by the different strains in infected relative to the uninfected cells was quantified and compared using Cufflinks (2.1.0) and MeV (4.0.9), respectively. Gene expression varied among the strains with the total number of genes as follows: F15/LAM4/KZN (1187), Beijing (1252), F11 (1639), F28 (870), Unique (886) and H37Rv (1179). A subset of 292 genes was commonly induced by all strains, where 52 genes were down-regulated while 240 genes were up-regulated. Differentially expressed genes were compared among the strains and the number of induced strain-specific gene signatures were as follows: F15/LAM4/KZN (138), Beijing (52), F11 (255), F28 (55), Unique (186) and H37Rv (125). Strain-specific molecular gene signatures associated with functional pathways were observed only for the Unique and H37Rv strains while certain biological functions may be associated with other strain signatures. This study demonstrated that strains of M. tuberculosis induce differential gene expression and strain-specific molecular signatures in pulmonary epithelial cells. Specific signatures induced by clinical strains of M. tuberculosis can be further explored for novel host-associated biomarkers and adjunctive immunotherapies.
Collapse
Affiliation(s)
- Nontobeko Eunice Mvubu
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, South Africa.
| | - Balakrishna Pillay
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, South Africa.
| | - Junaid Gamieldien
- South African National Bioinformatics Institute/MRC Unit for Bioinformatics Capacity Development, University of the Western Cape, South Africa.
| | - William Bishai
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, United States.
| | - Manormoney Pillay
- Medical Microbiology and Infection Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, South Africa.
| |
Collapse
|
44
|
Altube MJ, Selzer SM, de Farias MA, Portugal RV, Morilla MJ, Romero EL. Surviving nebulization-induced stress: dexamethasone in pH-sensitive archaeosomes. Nanomedicine (Lond) 2016; 11:2103-17. [DOI: 10.2217/nnm-2016-0165] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To increase the subcellular delivery of dexamethasone phosphate (DP) and stability to nebulization stress, pH-sensitive nanoliposomes (LpH) exhibiting archaeolipids, acting as ligands for scavenger receptors (pH-sensitive archaeosomes [ApH]), were prepared. Materials & methods: The anti-inflammatory effect of 0.18 mg DP/mg total lipid, 100–150 nm DP-containing ApH (dioleylphosphatidylethanolamine: Halorubrum tebenquichense total polar archaeolipids:cholesteryl hemisuccinate 4.2:2.8:3 w:w) was tested on different cell lines. Size and HPTS retention of ApH and conventional LpH (dioleylphosphatidylethanolamine:cholesteryl hemisuccinate 7:3 w:w) before and after nebulization were determined. Results & conclusion: DP-ApH suppressed IL-6 and TNF-α on phagocytic cells. Nebulized after 6-month storage, LpH increased size and completely lost its HPTS while ApH3 conserved size and polydispersity, fully retaining its original HPTS content.
Collapse
Affiliation(s)
- Maria Julia Altube
- Nanomedicine Research Program, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes. Roque Saenz Peña 352, Bernal B1876BXD, Argentina
| | - Solange Mailen Selzer
- Nanomedicine Research Program, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes. Roque Saenz Peña 352, Bernal B1876BXD, Argentina
| | - Marcelo Alexandre de Farias
- Brazilian Nanotechnology National Laboratory, CNPEM, Caixa Postal 6192, CEP 13.083–970, Campinas, São Paulo, Brazil
| | - Rodrigo Villares Portugal
- Brazilian Nanotechnology National Laboratory, CNPEM, Caixa Postal 6192, CEP 13.083–970, Campinas, São Paulo, Brazil
| | - Maria Jose Morilla
- Nanomedicine Research Program, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes. Roque Saenz Peña 352, Bernal B1876BXD, Argentina
| | - Eder Lilia Romero
- Nanomedicine Research Program, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes. Roque Saenz Peña 352, Bernal B1876BXD, Argentina
| |
Collapse
|
45
|
Speth JM, Bourdonnay E, Penke LRK, Mancuso P, Moore BB, Weinberg JB, Peters-Golden M. Alveolar Epithelial Cell-Derived Prostaglandin E2 Serves as a Request Signal for Macrophage Secretion of Suppressor of Cytokine Signaling 3 during Innate Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 196:5112-20. [PMID: 27183597 DOI: 10.4049/jimmunol.1502153] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/15/2016] [Indexed: 01/22/2023]
Abstract
Preservation of gas exchange mandates that the pulmonary alveolar surface restrain unnecessarily harmful inflammatory responses to the many challenges to which it is exposed. These responses reflect the cross-talk between alveolar epithelial cells (AECs) and resident alveolar macrophages (AMs). We recently determined that AMs can secrete suppressor of cytokine signaling (SOCS) proteins within microparticles. Uptake of these SOCS-containing vesicles by epithelial cells inhibits cytokine-induced STAT activation. However, the ability of epithelial cells to direct AM release of SOCS-containing vesicles in response to inflammatory insults has not been studied. In this study, we report that SOCS3 protein was elevated in bronchoalveolar lavage fluid of both virus- and bacteria-infected mice, as well as in an in vivo LPS model of acute inflammation. In vitro studies revealed that AEC-conditioned medium (AEC-CM) enhanced AM SOCS3 secretion above basal levels. Increased amounts of PGE2 were present in AEC-CM after LPS challenge, and both pharmacologic inhibition of PGE2 synthesis in AECs and neutralization of PGE2 in AEC-CM implicated this prostanoid as the major AEC-derived factor mediating enhanced AM SOCS3 secretion. Moreover, pharmacologic blockade of PGE2 synthesis or genetic deletion of a PGE2 synthase similarly attenuated the increase in bronchoalveolar lavage fluid SOCS3 noted in lungs of mice challenged with LPS in vivo. These results demonstrate a novel tunable form of cross-talk in which AECs use PGE2 as a signal to request SOCS3 from AMs to dampen their endogenous inflammatory responses during infection.
Collapse
Affiliation(s)
- Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Emilie Bourdonnay
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Loka Raghu Kumar Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jason B Weinberg
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and Division of Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109;
| |
Collapse
|
46
|
Ma Y, Han F, Liang J, Yang J, Shi J, Xue J, Yang L, Li Y, Luo M, Wang Y, Wei J, Liu X. A species-specific activation of Toll-like receptor signaling in bovine and sheep bronchial epithelial cells triggered by Mycobacterial infections. Mol Immunol 2016; 71:23-33. [DOI: 10.1016/j.molimm.2016.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/04/2016] [Indexed: 01/29/2023]
|
47
|
Su F, Wang Y, Liu G, Ru K, Liu X, Yu Y, Liu J, Wu Y, Quan F, Guo Z, Zhang Y. Generation of transgenic cattle expressing human β-defensin 3 as an approach to reducing susceptibility toMycobacterium bovisinfection. FEBS J 2016; 283:776-90. [DOI: 10.1111/febs.13641] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 12/19/2015] [Accepted: 01/04/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Feng Su
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
- College of Animal Science and Veterinary Medicine; Shandong Agricultural University; Taian Shandong China
| | - Yongsheng Wang
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Guanghui Liu
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Kun Ru
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Xin Liu
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Yuan Yu
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Jun Liu
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Yongyan Wu
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Fusheng Quan
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Zekun Guo
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| | - Yong Zhang
- College of Veterinary Medicine; Northwest A&F University; Yangling Shaanxi China
| |
Collapse
|
48
|
miR-26a suppresses EMT by disrupting the Lin28B/let-7d axis: potential cross-talks among miRNAs in IPF. J Mol Med (Berl) 2016; 94:655-65. [PMID: 26787543 DOI: 10.1007/s00109-016-1381-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/20/2015] [Accepted: 01/11/2016] [Indexed: 12/31/2022]
Abstract
UNLABELLED Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and highly lethal fibrotic lung disease with unknown cause or cure. Although some microRNAs (miRNAs), such as miR-26a and let-7d, have been confirmed, the contribution to the pathophysiological processes of IPF, the roles of miRNAs and intrinsic links between them in fibrotic lung diseases are not yet well understood. In this study, we found that Lin28B could induce the process of epithelial-mesenchymal transition (EMT) by inhibiting let-7d, whereas inhibition of Lin28B mitigated TGF-β1-induced fibrogenesis and attenuated EMT in both cultured A549 cells and MLE-12 cells. More importantly, over-expression of miR-26a could simultaneously enhance the expression of let-7d in A549 cells, and further study confirmed that Lin28B was one of the direct targets of miR-26a, which mediates, at least in part, the regulatory effects of miR-26a on the biogenesis of let-7d. Finally, we constructed a regulatory network among miRNAs involved in the progression of IPF. Taken together, our study deciphered the essential role of Lin28B in the pathogenesis of EMT, and unraveled a novel mechanism that miR-26a is a modulator of let-7d. This study also defines the miRNAs network involved in IPF, which may have implications for developing new strategies for pulmonary fibrosis. KEY MESSAGE Upregulation of Lin28B contributes to idiopathic pulmonary fibrosis. Lin28B causes epithelial-mesenchymal transition (EMT) by inhibition of let-7d. Lin28B is one of the targets of microRNA-26a. miR-26a enhances the expression of let-7d via targeting regulation of Lin28B. A regulatory network among miRNAs involved in the progression of IPF.
Collapse
|
49
|
Mvubu NE, Pillay B, Gamieldien J, Bishai W, Pillay M. Canonical pathways, networks and transcriptional factor regulation by clinical strains of Mycobacterium tuberculosis in pulmonary alveolar epithelial cells. Tuberculosis (Edinb) 2015; 97:73-85. [PMID: 26980499 DOI: 10.1016/j.tube.2015.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/30/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Limited knowledge exists on pathways, networks and transcriptional factors regulated within epithelial cells by diverse Mycobacterium tuberculosis genotypes. This study aimed to elucidate these mechanisms induced in A549 epithelial cells by dominant clinical strains in KwaZulu-Natal, South Africa. RNA for sequencing was extracted from epithelial cells at 48 h post-infection with 5 strains at a multiplicity of infection of approximately 10:1. Bioinformatics analysis performed with the RNA-Seq Tuxedo pipeline identified differentially expressed genes. Changes in pathways, networks and transcriptional factors were identified using Ingenuity Pathway Analysis (IPA). The interferon signalling and hepatic fibrosis/hepatic stellate cell activation pathways were among the top 5 canonical pathways in all strains. Hierarchical clustering for enrichment of cholesterol biosynthesis and immune associated pathways revealed similar patterns for Beijing and Unique; F15/LAM4/KZN and F11; and, F28 and H37Rv strains, respectively. However, the induction of top scoring networks varied among the strains. Among the transcriptional factors, only EHL, IRF7, PML, STAT1, STAT2 and VDR were induced by all clinical strains. Activation of the different pathways, networks and transcriptional factors revealed in the current study may be an underlying mechanism that results in the differential host response by clinical strains of M. tuberculosis.
Collapse
Affiliation(s)
- Nontobeko E Mvubu
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Balakrishna Pillay
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Junaid Gamieldien
- South African National Bioinformatics Institute/MRC Unit for Bioinformatics Capacity Development, University of the Western Cape, Bellville 7530, South Africa.
| | - William Bishai
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, 1550 Orleans St., Baltimore, MD, United State of America.
| | - Manormoney Pillay
- Medical Microbiology and Infection Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 719 Umbilo Road, Private Bag 7, Congella 4013, Durban, South Africa.
| |
Collapse
|
50
|
Kobayashi T, Tanaka K, Fujita T, Umezawa H, Amano H, Yoshioka K, Naito Y, Hatano M, Kimura S, Tatsumi K, Kasuya Y. Bidirectional role of IL-6 signal in pathogenesis of lung fibrosis. Respir Res 2015; 16:99. [PMID: 26289430 PMCID: PMC4546032 DOI: 10.1186/s12931-015-0261-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/12/2015] [Indexed: 02/08/2023] Open
Abstract
Background Various signals are known to participate in the pathogenesis of lung fibrosis. Our aim was to determine which signal is predominantly mobilized in the early inflammatory phase and thereafter modulates the development of lung fibrosis. Methods Mice received a single dose of 3 mg/kg body weight of bleomycin (BLM) and were sacrificed at designated days post-instillation (dpi). Lung homogenates and sections from mice in the early inflammatory phase were subjected to phospho-protein array analysis and immunofluorescence studies, respectively. Bronchoalveolar lavage fluid (BALF) from mice was subjected to an enzyme-linked immunosorbent assay (EIA) for interleukin (IL)-6 and evaluation of infiltrated cell populations. The effects of endogenous and exogenous IL-6 on the BLM-induced apoptotic signal in A549 cells and type 2 pneumocytes were elucidated. In addition, the effect of IL-6-neutralizing antibody on BLM-induced lung injury was evaluated. Results Phospho-protein array revealed that BLM induced phosphorylation of molecules downstream of the IL-6 receptor such as Stat3 and Akt in the lung at 3 dpi. At 3 dpi, immunofluorescence studies showed that signals of phospho-Stat3 and -Akt were localized in type 2 pneumocytes, and that BLM-induced IL-6-like immunoreactivity was predominantly observed in type 2 pneumocytes. Activation of caspases in BLM-treated A549 cells and type 2 pneumocytes was augmented by application of IL-6-neutralizing antibody, a PI3K inhibitor or a Stat3 inhibitor. EIA revealed that BLM-induced IL-6 in BALF was biphasic, with the first increase from 0.5 to 3 dpi followed by the second increase from 8 to 10 dpi. Blockade of the first increase of IL-6 by IL-6-neutralizing antibody enhanced apoptosis of type 2 pneumocytes and neutrophilic infiltration and markedly accelerated fibrosis in the lung. In contrast, blockade of the second increase of IL-6 by IL-6-neutralizing antibody ameliorated lung fibrosis. Conclusions The present study demonstrated that IL-6 could play a bidirectional role in the pathogenesis of lung fibrosis. In particular, upregulation of IL-6 at the early inflammatory stage of BLM-injured lung has antifibrotic activity through regulating the cell fate of type 2 pneumocytes in an autocrine/paracrine manner. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0261-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takeshi Kobayashi
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Kensuke Tanaka
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Tetsuo Fujita
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Hiroki Umezawa
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Hiroyuki Amano
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Kento Yoshioka
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Yusuke Naito
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Masahiko Hatano
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Sadao Kimura
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Yoshitoshi Kasuya
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|