1
|
Hailu S, Hurst C, Cyphers G, Thottunkal S, Harley D, Viney K, Irwin A, Dean J, Nourse C. Prevalence of extra-pulmonary tuberculosis in Africa: A systematic review and meta-analysis. Trop Med Int Health 2024; 29:257-265. [PMID: 38263374 DOI: 10.1111/tmi.13970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
OBJECTIVE The burden of extra-pulmonary tuberculosis (EPTB) is not well quantified in TB endemic countries such as those in sub-Saharan Africa. This study aimed to quantify that burden via a systematic review of the prevalence of EPTB in African countries. METHODS Studies were retrieved by searching five databases; 105 studies published between 1990 and 2023 were included. The studies described the prevalence of EPTB among the general population (4 studies), TB patients (68) and patients with other conditions, including HIV (15), meningitis (3), renal failure (3) and other comorbidities, some of which are cancer (12). Due to the low number of studies reporting EPTB in patients with conditions other than TB, the meta-analysis was performed on studies reporting on EPTB among TB patients (68 studies). Meta-analysis was performed on the 68 studies (271,073 participants) using a random-effects model to estimate the pooled prevalence of EPTB. Meta-regression was used to explore possible explanations for heterogeneity according to regions and time periods. RESULTS The pooled prevalence of EPTB among TB patients was 26% (95% CI 23-29%). There was substantial heterogeneity of prevalence for the five African regions. The Eastern region had the highest prevalence of 32% (95% CI 28-37%) and the lowest in Western Africa, 16% (95% CI 10-24%). There was no significant difference in the prevalence of EPTB between the 3 eleven-year time periods. CONCLUSIONS Our systematic review and meta-analysis give insight into the burden of EPTB in Africa. This review could inform clinical and programmatic practices-a higher suspicion index for clinicians and more effort for better services. This could contribute to efforts aiming to end TB, which have historically been focused on PTB.Coordinated efforts that target both EPTB and PTB are needed.
Collapse
Affiliation(s)
- Semira Hailu
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Cameron Hurst
- Molly Wardaguga Research Centre, Charles Darwin University, Brisbane, Australia
| | - Griffin Cyphers
- Faculty of Medicine, School of Public Health, The University of Queensland, Brisbane, Australia
| | - Stefan Thottunkal
- ANU College of Health and Medicine, Australian National University, Canberra, Australia
| | - David Harley
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Kerri Viney
- ANU College of Health and Medicine, Australian National University, Canberra, Australia
- School of Public Health, University of Sydney, Camperdown, Australia
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Adam Irwin
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Infection Management and Prevention Service, Queensland Children's Hospital, Brisbane, Australia
| | - Judith Dean
- Faculty of Medicine, School of Public Health, The University of Queensland, Brisbane, Australia
- UQ Poche Centre for Indigenous Health, The University of Queensland, Brisbane, Australia
| | - Clare Nourse
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Infection Management and Prevention Service, Queensland Children's Hospital, Brisbane, Australia
| |
Collapse
|
2
|
Barclay AM, Ninaber DK, van Veen S, Hiemstra PS, Ottenhoff THM, van der Does AM, Joosten SA. Airway epithelial cells mount an early response to mycobacterial infection. Front Cell Infect Microbiol 2023; 13:1253037. [PMID: 37822359 PMCID: PMC10562574 DOI: 10.3389/fcimb.2023.1253037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023] Open
Abstract
Lung epithelial cells represent the first line of host defence against foreign inhaled components, including respiratory pathogens. Their responses to these exposures may direct subsequent immune activation to these pathogens. The epithelial response to mycobacterial infections is not well characterized and may provide clues to why some mycobacterial infections are cleared, while others are persistent and pathogenic. We have utilized an air-liquid interface model of human primary bronchial epithelial cells (ALI-PBEC) to investigate the epithelial response to infection with a variety of mycobacteria: Mycobacterium tuberculosis (Mtb), M. bovis (BCG), M. avium, and M. smegmatis. Airway epithelial cells were found to be infected by all four species, albeit at low frequencies. The proportion of infected epithelial cells was lowest for Mtb and highest for M. avium. Differential gene expression analysis revealed a common epithelial host response to mycobacteria, including upregulation of BIRC3, S100A8 and DEFB4, and downregulation of BPIFB1 at 48 h post infection. Apical secretions contained predominantly pro-inflammatory cytokines, while basal secretions contained tissue growth factors and chemokines. Finally, we show that neutrophils were attracted to both apical and basal secretions of infected ALI-PBEC. Neutrophils were attracted in high numbers to apical secretions from PBEC infected with all mycobacteria, with the exception of secretions from M. avium-infected ALI-PBEC. Taken together, our results show that airway epithelial cells are differentially infected by mycobacteria, and react rapidly by upregulation of antimicrobials, and increased secretion of inflammatory cytokines and chemokines which directly attract neutrophils. Thus, the airway epithelium may be an important immunological component in controlling and regulating mycobacterial infections.
Collapse
Affiliation(s)
- Amy M. Barclay
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Dennis K. Ninaber
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anne M. van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
3
|
Sabo AN, Filaudeau E, Da Silva S, Becker G, Monassier L, Kemmel V. Flavoured and nicotine-containing e-liquids impair homeostatic properties of an alveolar-capillary cell model. Food Chem Toxicol 2023; 174:113650. [PMID: 36758787 DOI: 10.1016/j.fct.2023.113650] [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: 08/29/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
Most people consider that electronic cigarettes are safer than tobacco and are marketed as quit-smoking products. The e-liquid, which usually contains propylene glycol (PG) and vegetable glycerin (VG) in different ratios, nicotine and a wide variety of flavours, is heated by a coil and the aerosol droplets are primarily delivered to the alveolar area where nicotine and other molecules cross the alveolar-capillary barrier (ACB). However, e-cigarettes effects on the ACB are not yet established. In our study, a well-characterised in vitro model of the ACB was exposed to PG and VG and to five flavoured e-liquids with and without nicotine. The vehicles, due to their hypertonic properties, modulated the ACB integrity by modifying occludin expression. Below a 10% concentration, the vehicles did not trigger oxidative stress or cell death. Different results were observed between flavoured e-liquids: while red fruits and mint-eucalyptus disrupted ACB integrity, triggered oxidative stress and cell death, blond tobacco had no worse effect compared to the vehicles. However, the addition of nicotine in the latter e-liquid increased oxidative stress and cell death compared to the vehicles. Finally, mint-eucalyptus e-liquid increased some inflammation markers. Our results revealed that e-liquids alter ACB homeostasis, depending on flavour and nicotine presence.
Collapse
Affiliation(s)
- Amelia-Naomi Sabo
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France; Laboratoire de Biochimie et Biologie Moléculaire, Hôpitaux Universitaires de Strasbourg, 67200, Strasbourg, France.
| | - Emma Filaudeau
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France.
| | - Sylvia Da Silva
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France.
| | - Guillaume Becker
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France; Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Pôle Pharmacie-Pharmacologie, 67200, France.
| | - Laurent Monassier
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France.
| | - Véronique Kemmel
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, UR 7296, Faculté de Médecine de Maïeutique et des Métiers de la Santé, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 67085, Strasbourg, France; Laboratoire de Biochimie et Biologie Moléculaire, Hôpitaux Universitaires de Strasbourg, 67200, Strasbourg, France.
| |
Collapse
|
4
|
Gan Y, Hu Q, Li A, Gu L, Guo S. Estradiol inhibits autophagy of Mycobacterium tuberculosis‑infected 16HBE cells and controls the proliferation of intracellular Mycobacterium tuberculosis. Mol Med Rep 2022; 25:196. [PMID: 35425995 PMCID: PMC9052002 DOI: 10.3892/mmr.2022.12712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/21/2022] [Indexed: 11/08/2022] Open
Abstract
Tracheobronchial tuberculosis (TBTB) is most common in young, middle-aged females. Despite adequate anti-tuberculosis therapy, >90% of patients develop tracheobronchial stenosis, which has a high rate of resulting in disability. The present study aimed to explore the effect of estradiol on the development of TBTB. Estrogen receptor (ER) expression in granulomatous tissue was assessed via immunofluorescence. In order to determine whether estrogen affected the proliferation of intracellular Mycobacterium tuberculosis (Mtb), 16HBE cells were infected with Mtb in vitro, followed by estradiol treatment. Intracellular Mtb was quantified via colony counting. The effect of estradiol on autophagy of infected 16HBE cells was determined via western blotting and transmission electron microscopy. Necrosis assays of infected 16HBE cells were analyzed using propidium iodide staining and assessing lactate dehydrogenase (LDH) release. To determine how estradiol affects autophagy, infected 16HBE cells were treated with ER-specific and non-specific modulators. Reactive oxygen species (ROS) levels were analyzed via flow cytometry. Additionally, the protein expression levels of autophagy-associated proteins were determined via western blotting. Mtb could enter human lobar bronchial goblet cells and ciliated cells in patients with TBTB. The results also demonstrated that ERα was expressed in granulomatous tissue from patients with TBTB. Administration of 10−6 M estradiol reduced the number of intracellular Mtb colony-forming units in vitro in the 16HBE human bronchial epithelial cell line at day 3 after infection. Furthermore, cells treated with estradiol and infected with Mtb released less LDH at 72 h and exhibited reduced necrosis levels at 24 h compared with the untreated cells. In addition, autophagy of infected 16HBE cells was inhibited by estradiol. Estradiol and the specific ERα agonist had similar effects on autophagy in infected 16HBE cells. Additionally, treatment with the ERα antagonist abolished the inhibition of autophagy by estradiol in infected 16BHE cells. Compared with the untreated infected 16HBE cells, the ROS levels in the infected 16HBE cells treated with estradiol and the ERα agonist significantly decreased. The levels of phosphorylated (p)-mTOR and p-AKT notably increased in estradiol- and ERα agonist-treated infected 16HBE cells. In summary, estradiol may serve a key role in the development of TBTB through binding to ERα.
Collapse
Affiliation(s)
- Yiling Gan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qianfang Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Anmao Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lei Gu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
5
|
de Waal AM, Hiemstra PS, Ottenhoff TH, Joosten SA, van der Does AM. Lung epithelial cells interact with immune cells and bacteria to shape the microenvironment in tuberculosis. Thorax 2022; 77:408-416. [PMID: 35017314 PMCID: PMC8938665 DOI: 10.1136/thoraxjnl-2021-217997] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022]
Abstract
The lung epithelium has long been overlooked as a key player in tuberculosis disease. In addition to acting as a direct barrier to Mycobacterium tuberculosis (Mtb), epithelial cells (EC) of the airways and alveoli act as first responders during Mtb infections; they directly sense and respond to Mtb by producing mediators such as cytokines, chemokines and antimicrobials. Interactions of EC with innate and adaptive immune cells further shape the immune response against Mtb. These three essential components, epithelium, immune cells and Mtb, are rarely studied in conjunction, owing in part to difficulties in coculturing them. Recent advances in cell culture technologies offer the opportunity to model the lung microenvironment more closely. Herein, we discuss the interplay between lung EC, immune cells and Mtb and argue that modelling these interactions is of key importance to unravel early events during Mtb infection.
Collapse
Affiliation(s)
- Amy M de Waal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Hm Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne M van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Affinity of Mycobacterium tuberculosis strains for M059K microglial cells after migration through A549 alveolar epithelium. Eur J Clin Microbiol Infect Dis 2021; 40:1881-1889. [PMID: 33834319 DOI: 10.1007/s10096-021-04226-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/14/2021] [Indexed: 10/21/2022]
Abstract
Tuberculosis (TB) remains a major threat worldwide while central nervous system TB (CNS-TB) is one of the most severe forms of extrapulmonary TB. CNS-TB develops as a secondary infection during the hematogenous spread of Mycobacterium tuberculosis (M. tuberculosis) from the lungs to the CNS. Factors influencing the dissemination of the bacilli to the CNS have not been studied extensively. This study evaluated the transmigration ability through the alveolar epithelium and adhesion and invasion capacity of glial cells of M. tuberculosis strains of varying drug susceptibility and genotype profiles using an in vitro co-culture model. A549 alveolar epithelial cells and M059K glial cells were co-cultured in a Transwell plate with A549 cells cultured in the upper chamber and M059K glial cells in the lower chamber. A549 epithelial cells were infected with F15/LAM4/KZN (susceptible, MDR, XDR), Beijing (susceptible, XDR), F11 (susceptible), F28 (MDR), and H37Rv strains of M. tuberculosis. The transmigration of an A549 monolayer and subsequent adhesion and invasion rates of M059K cells were established. The susceptible and XDR variants of the F15/LAM4/KZN strain transmigrate the alveolar epithelial cell monolayer more efficiently than the MDR variant. The Beijing-XDR variant showed a high transmigration rate, while the susceptible variant showed no transmigration ability. Similar to the MDR F15/LAM4/KZN, the F28 and F11 strains showed a low dissemination ability. The bacteria were still capable to adhere to M059K glial cells after passage through the A549 cells. We conclude that M. tuberculosis isolates that passed through a monolayer of A549 alveolar epithelium by transcellular migration can still adhere to M059K glial cells. There is no genetic link between resistance and transmigration.
Collapse
|
7
|
Lee DF, Stewart GR, Chambers MA. Modelling early events in Mycobacterium bovis infection using a co-culture model of the bovine alveolus. Sci Rep 2020; 10:18495. [PMID: 33116165 PMCID: PMC7595104 DOI: 10.1038/s41598-020-75113-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/21/2020] [Indexed: 01/01/2023] Open
Abstract
Bovine tuberculosis (bTB), a zoonosis mainly caused by Mycobacterium bovis has severe socio-economic consequences and impact on animal health. Host-pathogen interactions during M. bovis infection are poorly understood, especially early events which are difficult to follow in vivo. This study describes the utilisation of an in vitro co-culture model, comprising immortalised bovine alveolar type II (BATII) epithelial cells and bovine pulmonary arterial endothelial cells (BPAECs). When cultured at air-liquid interface, it was possible to follow the migration of live M. bovis Bacille Calmette-Guérin (BCG) and to observe interactions with each cell type, alongside cytokine release. Infection with BCG was shown to exert a detrimental effect primarily upon epithelial cells, with corresponding increases in IL8, TNFα, IL22 and IL17a cytokine release, quantified by ELISA. BCG infection increased expression of CD54, MHC Class I and II molecules in endothelial but not epithelial cells, which exhibited constitutive expression. The effect of peripheral blood mononuclear cell conditioned medium from vaccinated cattle upon apical-basolateral migration of BCG was examined by quantifying recovered BCG from the apical, membrane and basolateral fractions over time. The numbers of recovered BCG in each fraction were unaffected by the presence of PBMC conditioned medium, with no observable differences between vaccinated and naïve animals.
Collapse
Affiliation(s)
- Diane Frances Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, UK.
| | | | - Mark Andrew Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, UK
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, UK
| |
Collapse
|
8
|
Reedoy KS, Loots DT, Beukes D, Reenen MV, Pillay B, Pillay M. Mycobacterium tuberculosis curli pili (MTP) is associated with significant host metabolic pathways in an A549 epithelial cell infection model and contributes to the pathogenicity of Mycobacterium tuberculosis. Metabolomics 2020; 16:116. [PMID: 33084984 DOI: 10.1007/s11306-020-01736-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION A clear understanding of the metabolome of Mycobacterium tuberculosis and its target host cell during infection is fundamental for the development of novel diagnostic tools, effective drugs and vaccines required to combat tuberculosis. The surface-located Mycobacterium tuberculosis curli pili (MTP) adhesin forms initial contact with the host cell and is therefore important for the establishment of infection. OBJECTIVE The aim of this investigation was to determine the role of MTP in modulating pathogen and host metabolic pathways in A549 epithelial cells infected with MTP proficient and deficient strains of M. tuberculosis. METHODS Uninfected A549 epithelial cells, and those infected with M. tuberculosis V9124 wild-type strain, Δmtp and the mtp-complemented strains, were subjected to metabolite extraction, two-dimensional gas chromatography time-of-flight mass spectrometry (GCxGC-TOFMS) and bioinformatic analyses. Univariate and multivariate statistical tests were used to identify metabolites that were significantly differentially produced in the WT-infected and ∆mtp-infected A549 epithelial cell models, comparatively. RESULTS A total of 46 metabolites occurred in significantly lower relative concentrations in the Δmtp-infected cells, indicating a reduction in nucleic acid synthesis, amino acid metabolism, glutathione metabolism, oxidative stress, lipid metabolism and peptidoglycan, compared to those cells infected with the WT strain. CONCLUSION The absence of MTP was associated with significant changes to the host metabolome, suggesting that this adhesin is an important contributor to the pathogenicity of M. tuberculosis, and supports previous findings of its potential as a suitable drug, vaccine and diagnostic target.
Collapse
Affiliation(s)
- K S Reedoy
- Medical Microbiology School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 1st Floor, Congella, Private Bag 7, Durban, 4013, South Africa
| | - D T Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag x6001, Box 269, Potchefstroom, 2531, South Africa
| | - D Beukes
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag x6001, Box 269, Potchefstroom, 2531, South Africa
| | - M van Reenen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag x6001, Box 269, Potchefstroom, 2531, South Africa
| | - B Pillay
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - M Pillay
- Medical Microbiology School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 1st Floor, Congella, Private Bag 7, Durban, 4013, South Africa.
| |
Collapse
|
9
|
Moule MG, Cirillo JD. Mycobacterium tuberculosis Dissemination Plays a Critical Role in Pathogenesis. Front Cell Infect Microbiol 2020; 10:65. [PMID: 32161724 PMCID: PMC7053427 DOI: 10.3389/fcimb.2020.00065] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/07/2020] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium tuberculosis is primarily a respiratory pathogen. However, 15% of infections worldwide occur at extrapulmonary sites causing additional complications for diagnosis and treatment of the disease. In addition, dissemination of M. tuberculosis out of the lungs is thought to be more than just a rare event leading to extrapulmonary tuberculosis, but rather a prerequisite step that occurs during all infections, producing secondary lesions that can become latent or productive. In this review we will cover the clinical range of extrapulmonary infections and the process of dissemination including evidence from both historical medical literature and animal experiments for dissemination and subsequent reseeding of the lungs through the lymphatic and circulatory systems. While the mechanisms of M. tuberculosis dissemination are not fully understood, we will discuss the various models that have been proposed to address how this process may occur and summarize the bacterial virulence factors that facilitate M. tuberculosis dissemination.
Collapse
Affiliation(s)
- Madeleine G. Moule
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
10
|
Ryndak MB, Laal S. Mycobacterium tuberculosis Primary Infection and Dissemination: A Critical Role for Alveolar Epithelial Cells. Front Cell Infect Microbiol 2019; 9:299. [PMID: 31497538 PMCID: PMC6712944 DOI: 10.3389/fcimb.2019.00299] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/02/2019] [Indexed: 12/28/2022] Open
Abstract
Globally, tuberculosis (TB) has reemerged as a major cause of morbidity and mortality, despite the use of the Mycobacterium bovis BCG vaccine and intensive attempts to improve upon BCG or develop new vaccines. Two lacunae in our understanding of the Mycobacterium tuberculosis (M. tb)-host pathogenesis have mitigated the vaccine efforts; the bacterial-host interaction that enables successful establishment of primary infection and the correlates of protection against TB. The vast majority of vaccine efforts are based on the premise that cell-mediated immunity (CMI) is the predominating mode of protection against TB. However, studies in animal models and in humans demonstrate that post-infection, a period of several weeks precedes the initiation of CMI during which the few inhaled bacteria replicate dramatically and disseminate systemically. The “Trojan Horse” mechanism, wherein M. tb is phagocytosed and transported across the alveolar barrier by infected alveolar macrophages has been long postulated as the sole, primary M. tb:host interaction. In the current review, we present evidence from our studies of transcriptional profiles of M. tb in sputum as it emerges from infectious patients where the bacteria are in a quiescent state, to its adaptations in alveolar epithelial cells where the bacteria transform to a highly replicative and invasive phenotype, to its maintenance of the invasive phenotype in whole blood to the downregulation of invasiveness upon infection of epithelial cells at an extrapulmonary site. Evidence for this alternative mode of infection and dissemination during primary infection is supported by in vivo, in vitro cell-based, and transcriptional studies from multiple investigators in recent years. The proposed alternative mechanism of primary infection and dissemination across the alveolar barrier parallels our understanding of infection and dissemination of other Gram-positive pathogens across their relevant mucosal barriers in that barrier-specific adhesins, toxins, and enzymes synergize to facilitate systemic establishment of infection prior to the emergence of CMI. Further exploration of this M. tb:non-phagocytic cell interaction can provide alternative approaches to vaccine design to prevent infection with M. tb and not only decrease clinical disease but also decrease the overwhelming reservoir of latent TB infection.
Collapse
Affiliation(s)
- Michelle B Ryndak
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Suman Laal
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
11
|
Lee D, Chambers M. A bilayer tissue culture model of the bovine alveolus. F1000Res 2019; 8:357. [PMID: 31448101 PMCID: PMC6685456 DOI: 10.12688/f1000research.18696.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2019] [Indexed: 08/02/2024] Open
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture bilayer model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and the bilayer cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the bilayer model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their culture as a bilayer in conjunction with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The bilayer model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
12
|
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the co-culture model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their co-culture with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The co-culture model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
13
|
Hasan S, Sebo P, Osicka R. A guide to polarized airway epithelial models for studies of host-pathogen interactions. FEBS J 2018; 285:4343-4358. [PMID: 29896776 DOI: 10.1111/febs.14582] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/21/2018] [Accepted: 06/11/2018] [Indexed: 11/28/2022]
Abstract
Mammalian lungs are organs exhibiting the cellular and spatial complexity required for gas exchange to support life. The respiratory epithelium internally lining the airways is susceptible to infections due to constant exposure to inhaled microbes. Biomedical research into respiratory bacterial infections in humans has been mostly carried out using small mammalian animal models or two-dimensional, submerged cultures of undifferentiated epithelial cells. These experimental model systems have considerable limitations due to host specificity of bacterial pathogens and lack of cellular and morphological complexity. This review describes the in vitro differentiated and polarized airway epithelial cells of human origin that are used as a model to study respiratory bacterial infections. Overall, these models recapitulate key aspects of the complexity observed in vivo and can help in elucidating the molecular details of disease processes observed during respiratory bacterial infections.
Collapse
Affiliation(s)
- Shakir Hasan
- Institute of Microbiology of the CAS, v. v. i., Prague, Czech Republic
| | - Peter Sebo
- Institute of Microbiology of the CAS, v. v. i., Prague, Czech Republic
| | - Radim Osicka
- Institute of Microbiology of the CAS, v. v. i., Prague, Czech Republic
| |
Collapse
|
14
|
Svensson M, Chen P. Human Organotypic Respiratory Models. Curr Top Microbiol Immunol 2018:29-54. [PMID: 29808337 DOI: 10.1007/82_2018_91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Biomedical research aiming to understand the molecular basis of human lung tissue development, homeostasis and disease, or to develop new therapies for human respiratory diseases, requires models that faithfully recapitulate the human condition. This has stimulated biologists and engineers to develop in vitro organotypic models mimicking human respiratory tissues. In this chapter, we provide examples of different types of model systems ranging from simple unicellular cultures to more complex multicellular systems. The models contain, in varying degree, cell types present in real tissue in combination with different extracellular matrix components that can critically affect cell phenotype and function. We also describe how organotypic respiratory models can be combined with human innate immune cells, to better recapitulate tissue inflammation, a key component in, for example, infectious diseases. These models have the potential to provide new insights into lung physiology, tissue infection and inflammation, disease mechanisms, as well as provide a platform for identification of novel targets and screening of candidate drugs in human lung disorders.
Collapse
Affiliation(s)
- Mattias Svensson
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden.
| | - Puran Chen
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden
| |
Collapse
|
15
|
Novel Models to Study Stromal Cell-Leukocyte Interactions in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:131-146. [PMID: 30155626 DOI: 10.1007/978-3-319-78127-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To study human immunology in general and stromal immunology in particular, it is highly motivated to move from monolayers to 3D cultures, such as organotypic models, that better mimic the function of living tissue. These models can potentially contain most if not all cell types present in tissues, in combination with different extracellular matrix components that can critically affect cell phenotype. Besides their well-established use in studies of tissue-specific cells, such as epithelial cells, endothelial cells and stromal fibroblasts in combination with extracellular components, these models have also been shown to be valuable to study how tissue participates in the regulation of leukocyte differentiation and function. Organotypic models with leukocytes represent novel powerful tools to study human stromal immunology and mechanisms involved in the regulation of leukocyte functions and inflammatory processes in human health and disease. In particular, these models are robust, long-lived and reproducible and allow monitoring of disease progression in real time, as well as the mixing of cellular constituents from healthy and pathological tissues. These models are also easy to manipulate, either genetically or by adding external stimulants, such as cytokines and pathogens, to mimic pathological conditions. It is thus not surprising that these models are proposed to be useful in toxicology screening assays, evaluating therapeutic efficacy of drugs and antibiotics, as well as in personalized medicine. Within this chapter, the most recent developments in creating organotypic models for the purpose of study of human leukocyte and stromal cell interactions, in health and disease, will be discussed, in particular focusing on live imaging. Special emphasis will be given on an organotypic model resembling human lung and its usefulness in studying the fine control of physiological and pathological processes in human health and disease. Using these models in studies on human stromal cell and leukocyte interactions will likely help identifying novel disease traits and may point out new potential targets to monitor and treat human diseases.
Collapse
|
16
|
Pellowe AS, Lauridsen HM, Matta R, Gonzalez AL. Ultrathin Porated Elastic Hydrogels As a Biomimetic Basement Membrane for Dual Cell Culture. J Vis Exp 2017. [PMID: 29364202 DOI: 10.3791/56384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The basement membrane is a critical component of cellular bilayers that can vary in stiffness, composition, architecture, and porosity. In vitro studies of endothelial-epithelial bilayers have traditionally relied on permeable support models that enable bilayer culture, but permeable supports are limited in their ability to replicate the diversity of human basement membranes. In contrast, hydrogel models that require chemical synthesis are highly tunable and allow for modifications of both the material stiffness and the biochemical composition via incorporation of biomimetic peptides or proteins. However, traditional hydrogel models are limited in functionality because they lack pores for cell-cell contacts and functional in vitro migration studies. Additionally, due to the thickness of traditional hydrogels, incorporation of pores that span the entire thickness of hydrogels has been challenging. In the present study, we use poly-(ethylene-glycol) (PEG) hydrogels and a novel zinc oxide templating method to address the previous shortcomings of biomimetic hydrogels. As a result, we present an ultrathin, basement membrane-like hydrogel that permits the culture of confluent cellular bilayers on a customizable scaffold with variable pore architectures, mechanical properties, and biochemical composition.
Collapse
|
17
|
Fonseca KL, Rodrigues PNS, Olsson IAS, Saraiva M. Experimental study of tuberculosis: From animal models to complex cell systems and organoids. PLoS Pathog 2017; 13:e1006421. [PMID: 28817682 PMCID: PMC5560521 DOI: 10.1371/journal.ppat.1006421] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is a devastating disease to mankind that has killed more people than any other infectious disease. Despite many efforts and successes from the scientific and health communities, the prospect of TB elimination remains distant. On the one hand, sustainable public health programs with affordable and broad implementation of anti-TB measures are needed. On the other hand, achieving TB elimination requires critical advances in three areas: vaccination, diagnosis, and treatment. It is also well accepted that succeeding in advancing these areas requires a deeper knowledge of host—pathogen interactions during infection, and for that, better experimental models are needed. Here, we review the potential and limitations of different experimental approaches used in TB research, focusing on animal and human-based cell culture models. We highlight the most recent advances in developing in vitro 3D models and introduce the potential of lung organoids as a new tool to study Mycobacterium tuberculosis infection. Tuberculosis (TB) is the number 1 killer in the world due to a bacterial infection. The study of this disease through clinical and epidemiological data and through the use of different experimental models has provided important knowledge on the role of the immune response generated during infection. This is critical for the development of novel vaccines and therapeutic strategies. However, in spite of the advances made, it is well accepted that better models are needed to study TB. This review discusses the different models used to study TB, highlighting the advantages and disadvantages of the available animal and cellular models and introducing recently developed state-of-the-art approaches based on human-based cell culture systems. These new advances are integrated in a road map for future study of TB, converging for the potential of lung organoids in TB research.
Collapse
Affiliation(s)
- Kaori L. Fonseca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro N. S. Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - I. Anna S. Olsson
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
18
|
Strains of Mycobacterium tuberculosis differ in affinity for human osteoblasts and alveolar cells in vitro. SPRINGERPLUS 2016; 5:163. [PMID: 27026860 PMCID: PMC4766163 DOI: 10.1186/s40064-016-1819-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/15/2016] [Indexed: 12/29/2022]
Abstract
Although the lung is the primary site of infection of tuberculosis, Mycobacterium tuberculosis is capable of causing infection at other sites. In 5–10 % such extra-pulmonary tuberculosis is located in bone tissue of the spine. It is unknown whether host or microbial factors are responsible for the site where extra-pulmonary tuberculosis manifests itself. One MDR isolate belonging to strain F28, one susceptible F11 and one isolate each of susceptible, MDR and XDR F15/LAM4/KZN were cultured in Middlebrook 7H9 media. Human osteoblasts (SaOS-2) and human alveolar epithelial cells (A549) were exposed to these different isolates of M. tuberculosis and invasion capacity and intra-cellular multiplication rates were established. Mouse macrophage (MHS) cells exposed to M. tuberculosis H37Rv served as control. The invasion capacity of F15/LAM4/KZN representatives increased with the level of resistance. The F28 MDR strain showed similar invasion capacity as the XDR F15/LAM4/KZN for pulmonary epthelial cells, whilst the fully susceptible F11 strain displayed a propensity for osteoblasts. The differences observed may in part explain why certain strains are able to cause infection at specific extra-pulmonary sites. We postulated that the development of extra-pulmonary tuberculosis depends on the ability of the microbe to pass effectively through the alveolar epithelial lining and its affinity for cells other than those in pulmonary tissue.
Collapse
|
19
|
Lockwood SY, Meisel JE, Monsma FJ, Spence DM. A Diffusion-Based and Dynamic 3D-Printed Device That Enables Parallel in Vitro Pharmacokinetic Profiling of Molecules. Anal Chem 2016; 88:1864-70. [PMID: 26727249 PMCID: PMC5296943 DOI: 10.1021/acs.analchem.5b04270] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The process of bringing a drug to market involves many steps, including the preclinical stage, where various properties of the drug candidate molecule are determined. These properties, which include drug absorption, distribution, metabolism, and excretion, are often displayed in a pharmacokinetic (PK) profile. While PK profiles are determined in animal models, in vitro systems that model in vivo processes are available, although each possesses shortcomings. Here, we present a 3D-printed, diffusion-based, and dynamic in vitro PK device. The device contains six flow channels, each with integrated porous membrane-based insert wells. The pores of these membranes enable drugs to freely diffuse back and forth between the flow channels and the inserts, thus enabling both loading and clearance portions of a standard PK curve to be generated. The device is designed to work with 96-well plate technology and consumes single-digit milliliter volumes to generate multiple PK profiles, simultaneously. Generation of PK profiles by use of the device was initially performed with fluorescein as a test molecule. Effects of such parameters as flow rate, loading time, volume in the insert well, and initial concentration of the test molecule were investigated. A prediction model was generated from this data, enabling the user to predict the concentration of the test molecule at any point along the PK profile within a coefficient of variation of ∼ 5%. Depletion of the analyte from the well was characterized and was determined to follow first-order rate kinetics, indicated by statistically equivalent (p > 0.05) depletion half-lives that were independent of the starting concentration. A PK curve for an approved antibiotic, levofloxacin, was generated to show utility beyond the fluorescein test molecule.
Collapse
Affiliation(s)
- Sarah Y. Lockwood
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jayda E. Meisel
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | | | - Dana M. Spence
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
20
|
Jud C, Ahmed S, Müller L, Kinnear C, Vanhecke D, Umehara Y, Frey S, Liley M, Angeloni S, Petri-Fink A, Rothen-Rutishauser B. Ultrathin Ceramic Membranes as Scaffolds for Functional Cell Coculture Models on a Biomimetic Scale. Biores Open Access 2015; 4:457-68. [PMID: 26713225 PMCID: PMC4691652 DOI: 10.1089/biores.2015.0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Epithelial tissue serves as an interface between biological compartments. Many in vitro epithelial cell models have been developed as an alternative to animal experiments to answer a range of research questions. These in vitro models are grown on permeable two-chamber systems; however, commercially available, polymer-based cell culture inserts are around 10 μm thick. Since the basement membrane found in biological systems is usually less than 1 μm thick, the 10-fold thickness of cell culture inserts is a major limitation in the establishment of realistic models. In this work, an alternative insert, accommodating an ultrathin ceramic membrane with a thickness of only 500 nm (i.e., the Silicon nitride Microporous Permeable Insert [SIMPLI]-well), was produced and used to refine an established human alveolar barrier coculture model by both replacing the conventional inserts with the SIMPLI-well and completing it with endothelial cells. The structural–functional relationship of the model was evaluated, including the translocation of gold nanoparticles across the barrier, revealing a higher translocation if compared to corresponding polyethylene terephthalate (PET) membranes. This study demonstrates the power of the SIMPLI-well system as a scaffold for epithelial tissue cell models on a truly biomimetic scale, allowing construction of more functionally accurate models of human biological barriers.
Collapse
Affiliation(s)
- Corinne Jud
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland . ; Agroscope, Institute for Livestock Sciences ILS , Posieux, Switzerland
| | | | - Loretta Müller
- University Children's Hospital Basel , Basel, Switzerland
| | - Calum Kinnear
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland
| | - Dimitri Vanhecke
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland
| | - Yuki Umehara
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland
| | - Sabine Frey
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland
| | | | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg , Fribourg, Switzerland
| | | |
Collapse
|
21
|
Scordo JM, Knoell DL, Torrelles JB. Alveolar Epithelial Cells in Mycobacterium tuberculosis Infection: Active Players or Innocent Bystanders? J Innate Immun 2015; 8:3-14. [PMID: 26384325 DOI: 10.1159/000439275] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 08/10/2015] [Indexed: 11/19/2022] Open
Abstract
Tuberculosis (TB) is a disease that kills one person every 18 s. TB remains a global threat due to the emergence of drug-resistant Mycobacterium tuberculosis (M.tb) strains and the lack of an efficient vaccine. The ability of M.tb to persist in latency, evade recognition following seroconversion, and establish resistance in vulnerable populations warrants closer examination. Past and current research has primarily focused on examination of the role of alveolar macrophages and dendritic cells during M.tb infection, which are critical in the establishment of the host response during infection. However, emerging evidence indicates that the alveolar epithelium is a harbor for M.tb and critical during progression to active disease. Here we evaluate the relatively unexplored role of the alveolar epithelium as a reservoir and also its capacity to secrete soluble mediators upon M.tb exposure, which influence the extent of infection. We further discuss how the M.tb-alveolar epithelium interaction instigates cell-to-cell crosstalk that regulates the immune balance between a proinflammatory and an immunoregulatory state, thereby prohibiting or allowing the establishment of infection. We propose that consideration of alveolar epithelia provides a more comprehensive understanding of the lung environment in vivo in the context of host defense against M.tb.
Collapse
Affiliation(s)
- Julia M Scordo
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
22
|
Mycobacterium tuberculosis
infection of the ‘non‐classical immune cell’. Immunol Cell Biol 2015; 93:789-95. [PMID: 25801479 DOI: 10.1038/icb.2015.43] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/02/2015] [Accepted: 03/18/2015] [Indexed: 01/29/2023]
|
23
|
Ryndak MB, Singh KK, Peng Z, Laal S. Transcriptional profile of Mycobacterium tuberculosis replicating in type II alveolar epithelial cells. PLoS One 2015; 10:e0123745. [PMID: 25844539 PMCID: PMC4386821 DOI: 10.1371/journal.pone.0123745] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 03/05/2015] [Indexed: 12/31/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb) infection is initiated by the few bacilli inhaled into the alveolus. Studies in lungs of aerosol-infected mice provided evidence for extensive replication of M. tb in non-migrating, non-antigen-presenting cells in the alveoli during the first 2-3 weeks post-infection. Alveoli are lined by type II and type I alveolar epithelial cells (AEC) which outnumber alveolar macrophages by several hundred-fold. M. tb DNA and viable M. tb have been demonstrated in AEC and other non-macrophage cells of the kidney, liver, and spleen in autopsied tissues from latently-infected subjects from TB-endemic regions indicating systemic bacterial dissemination during primary infection. M. tb have also been demonstrated to replicate rapidly in A549 cells (type II AEC line) and acquire increased invasiveness for endothelial cells. Together, these results suggest that AEC could provide an important niche for bacterial expansion and development of a phenotype that promotes dissemination during primary infection. In the current studies, we have compared the transcriptional profile of M. tb replicating intracellularly in A549 cells to that of M. tb replicating in laboratory broth, by microarray analysis. Genes significantly upregulated during intracellular residence were consistent with an active, replicative, metabolic, and aerobic state, as were genes for tryptophan synthesis and for increased virulence (ESAT-6, and ESAT-6-like genes, esxH, esxJ, esxK, esxP, and esxW). In contrast, significant downregulation of the DevR (DosR) regulon and several hypoxia-induced genes was observed. Stress response genes were either not differentially expressed or were downregulated with the exception of the heat shock response and those induced by low pH. The intra-type II AEC M. tb transcriptome strongly suggests that AEC could provide a safe haven in which M. tb can expand dramatically and disseminate from the lung prior to the elicitation of adaptive immune responses.
Collapse
Affiliation(s)
- Michelle B. Ryndak
- Department of Pathology, New York University Langone Medical Center, New York, New York, United States of America
| | - Krishna K. Singh
- Department of Pathology, New York University Langone Medical Center, New York, New York, United States of America
| | - Zhengyu Peng
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Suman Laal
- Department of Pathology, New York University Langone Medical Center, New York, New York, United States of America
- Veterans Affairs New York Harbor Healthcare System, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
24
|
Vir P, Gupta D, Agarwal R, Verma I. Interaction of alveolar epithelial cells with CFP21, a mycobacterial cutinase-like enzyme. Mol Cell Biochem 2014; 396:187-99. [PMID: 25091806 DOI: 10.1007/s11010-014-2154-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 07/14/2014] [Indexed: 12/17/2022]
Abstract
Mycobacterium tuberculosis (M. tb), an intracellular pathogen, has the ability to infect alveolar epithelial cells (AEC) also in addition to alveolar macrophages. The virulence of M. tb is attributed to proteins encoded by genomic regions of deletion (RD) and till date 16 such regions (RD1-RD16) have been identified. Culture filtrate protein 21 (CFP21), a RD2 secretory protein, is a cutinase-like enzyme that possesses esterase and lipolytic activity. It is hypothesized that CFP21 could be playing a role in M. tb virulence by disrupting the host cell integrity. In this study, recombinant CFP21 was expressed and purified. The in vitro exposure of type I (WI26) and type II (A549) AEC to CFP21 resulted in a significant decline in their cellular viability by inducing cell apoptosis. However, the cytotoxic effects were more pronounced in WI26 cells than in A549 cells. The analysis of immune responses in CFP21-treated AEC exhibited significant production of reactive oxygen species and anti-inflammatory cytokine TGF-β which indicated oxidative stress-mediated cell death. These results show that CFP21 could play an important role in M. tb pathogenesis by disrupting the host alveolar barrier and thereby facilitating mycobacterial dissemination.
Collapse
Affiliation(s)
- Pooja Vir
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Sector-12, Chandigarh, 160012, India
| | | | | | | |
Collapse
|
25
|
Nguyen Hoang AT, Chen P, Björnfot S, Högstrand K, Lock JG, Grandien A, Coles M, Svensson M. Technical advance: live-imaging analysis of human dendritic cell migrating behavior under the influence of immune-stimulating reagents in an organotypic model of lung. J Leukoc Biol 2014; 96:481-9. [PMID: 24899587 DOI: 10.1189/jlb.3ta0513-303r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
This manuscript describes technical advances allowing manipulation and quantitative analyses of human DC migratory behavior in lung epithelial tissue. DCs are hematopoietic cells essential for the maintenance of tissue homeostasis and the induction of tissue-specific immune responses. Important functions include cytokine production and migration in response to infection for the induction of proper immune responses. To design appropriate strategies to exploit human DC functional properties in lung tissue for the purpose of clinical evaluation, e.g., candidate vaccination and immunotherapy strategies, we have developed a live-imaging assay based on our previously described organotypic model of the human lung. This assay allows provocations and subsequent quantitative investigations of DC functional properties under conditions mimicking morphological and functional features of the in vivo parental tissue. We present protocols to set up and prepare tissue models for 4D (x, y, z, time) fluorescence-imaging analysis that allow spatial and temporal studies of human DCs in live epithelial tissue, followed by flow cytometry analysis of DCs retrieved from digested tissue models. This model system can be useful for elucidating incompletely defined pathways controlling DC functional responses to infection and inflammation in lung epithelial tissue, as well as the efficacy of locally administered candidate interventions.
Collapse
Affiliation(s)
| | - Puran Chen
- Center for Infectious Medicine, Department of Medicine, and
| | - Sofia Björnfot
- Center for Infectious Medicine, Department of Medicine, and
| | - Kari Högstrand
- Center for Infectious Medicine, Department of Medicine, and
| | - John G Lock
- Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; and
| | - Alf Grandien
- Center for Infectious Medicine, Department of Medicine, and
| | - Mark Coles
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, United Kingdom
| | | |
Collapse
|
26
|
Vir P, Gupta D, Agarwal R, Verma I. Immunomodulation of alveolar epithelial cells by Mycobacterium tuberculosis phosphatidylinositol mannosides results in apoptosis. APMIS 2013; 122:268-82. [PMID: 23919648 DOI: 10.1111/apm.12141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/22/2013] [Indexed: 11/27/2022]
Abstract
During intracellular residence in macrophages, mycobacterial lipids, namely phosphatidylinositol mannosides (PIM) and lipoarabinomannans, are expelled in the lung milieu to interact with host cells. PIM include a group of essential lipid components of Mycobacterium tuberculosis (M. tb) cell wall. Given that PIM function as mycobacterial adhesins for binding to host cells, the present study explored the consequences of interaction of M. tb PIM with alveolar epithelial cells (AEC). A 24-h PIM exposure at a concentration of 10 μg/mL to AEC conferred cytolysis to AEC via induction of apoptosis, suggesting their potential to alter alveolar epithelium integrity. The results also reflected that type I like AEC are more sensitive to cytolysis than type II AEC. PIM-treated AEC exhibited significant production of reactive oxygen species (ROS) and an immunosuppressive cytokine transforming growth factor-β (TGF-β) in the culture supernatants. Although AEC displayed constitutive mRNA transcripts for toll-like receptors (TLR2 and 4) as well as chemokines (IL-8 and MCP-1), no significant change in their expression was observed upon PIM treatment. Collectively, these results offer insights into PIM potential as M. tb virulence factor that might promote mycobacterial dissemination by causing cytolysis of AEC via increased production of ROS and TGF-β.
Collapse
Affiliation(s)
- Pooja Vir
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | |
Collapse
|
27
|
Rivero-Lezcano OM. In vitro infection of human cells with Mycobacterium tuberculosis. Tuberculosis (Edinb) 2013; 93:123-9. [DOI: 10.1016/j.tube.2012.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 08/23/2012] [Accepted: 09/20/2012] [Indexed: 11/26/2022]
|
28
|
Guo XG, Ji TX, Xia Y, Ma YY. Autophagy protects type II alveolar epithelial cells from Mycobacterium tuberculosis infection. Biochem Biophys Res Commun 2013; 432:308-13. [PMID: 23396060 DOI: 10.1016/j.bbrc.2013.01.111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 01/29/2013] [Indexed: 11/30/2022]
Abstract
This study was designed to investigate the protective effect of the autophagy signaling pathway against Mycobacterium tuberculosis infection in type II alveolar epithelial cells. An in vitro M. tuberculosis system was established using human A549 cells. Infection-induced changes in the expression of the autophagic marker LC3 were assessed by reverse transcription-PCR and Western blotting. Morphological changes in autophagosomes were detected by transmission electron microscopy (TEM). The function of the autophagy signaling pathway during infection was assessed by measuring the level of cell death and the amount of lactate dehydrogenase (LDH) released in the presence or absence of the inhibitor 3-methyladenine (3-MA). In addition, effects on LDH release were assessed after the siRNA-mediated knockdown of the essential autophagosomal structural membrane protein Atg5. LC3 mRNA expression was significantly reduced in M.tuberculosis-infected A549 cells (16888.76 ± 1576.34 vs. uninfected: 12744.29 ± 1089.37; P < 0.05). TEM revealed M.tuberculosis bacilli-containing compartments that were surrounded by double membranes characteristic of the autophagic process. M.tuberculosis-infected A549 cells released more LDH (1.45 ± 0.12 vs. uninfected: 0.45 ± 0.04; P < 0.05). The inhibition of autophagy signaling significantly enhanced M.tuberculosis-induced necrosis (3-MA: 75 ± 5% vs. untreated: 15 ± 1%; P < 0.05) and LDH release (3-MA: 2.50 ± 0.24 vs. untreated: 0.45 ± 0.04; Atg5 knockdown: 3.19 ± 0.29 vs. untreated: 1.28 ± 0.11; P < 0.05). Our results indicate that autophagy signaling pathway prevents apoptosis in type II alveolar epithelial cells infected with M.tuberculosis and may represent a molecular target for promoting cell survival during infection by respiratory pathogens.
Collapse
Affiliation(s)
- Xu-Guang Guo
- Center for Clinical Laboratory Medicine of PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | |
Collapse
|
29
|
Adlakha N, Vir P, Verma I. Effect of mycobacterial secretory proteins on the cellular integrity and cytokine profile of type II alveolar epithelial cells. Lung India 2012; 29:313-8. [PMID: 23243342 PMCID: PMC3519014 DOI: 10.4103/0970-2113.102796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Pulmonary tuberculosis (TB) is caused by Mycobacterium tuberculosis (M. tb). In lungs, alveolar macrophages and type II alveolar epithelial cells serve as a replicative niche for this pathogen. Secretory proteins released by actively replicating tubercle bacilli are known to interact with host cells at the initial stages of infection. To understand the role of these cells in TB pathogenesis, it is important to identify the mycobacterial components involved in interaction with alveolar epithelial cells. Materials and Methods: We fractionated the whole secretory proteome of M. tb H37Rv into 10 narrow molecular mass fractions (A1-A10; <20 kDa to >90 kDa) that were studied for their binding potential with A549; type II alveolar epithelial cell line. We also studied the consequences of this interaction in terms of change in epithelial cell viability by MTT assay and cytokine release by ELISA. Results: Our results show that several mycobacterial proteins bind and confer cytolysis in epithelial cells. Amongst all the fractions, proteins ranging from 35-45 kDa (A5) exhibited highest binding to A549 cells with a consequence of cytolysis of these cells. This fraction (A5) also led to release of various cytokines important in anti-mycobacterial immunity. Conclusion: Fraction A5 (35-45 kDa) of mycobacterial secretory proteome play an important role in mediating M. tb interaction with type II alveolar epithelial cells with the consequences detrimental for the TB pathogenesis. Further studies are being carried out to identify the candidate proteins from this region.
Collapse
Affiliation(s)
- Nidhi Adlakha
- Department of Biochemistry, TB Immunology and Cell Biology Lab, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
30
|
Rothen-Rutishauser B, Clift MJ, Jud C, Fink A, Wick P. Human epithelial cells in vitro – Are they an advantageous tool to help understand the nanomaterial-biological barrier interaction? ACTA ACUST UNITED AC 2012. [DOI: 10.1515/entl-2015-0004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstratThe human body can be exposed to nanomaterials through a variety of different routes. As nanomaterials get in contact with the skin, the gastrointestinal tract, and the respiratory tract, these biological compartments are acting as barriers to the passage of nano-sized materials into the organism. These structural and functional barriers are provided by the epithelia serving as an interface between biological compartments. In order to initiate the reduction, refinement and replacement of time consuming, expensive and stressful (to the animals) in vivo experimental approaches, many in vitro epithelial cell culture models have been developed during the last decades. This review therefore, focuses on the functional as well as structural aspects of epithelial cells as well as the most commonly used in vitro epithelial models of the primary biological barriers with which nanomaterials might come in contact with either occupationally, or during their manufacturing and application. The advantages and disadvantages of the different in vitro models are discussed in order to provide a clear overview as to whether or not epithelial cell cultures are an advantageous model to be used for basic mechanism and nanotoxicology research.
Collapse
|
31
|
Alveolocapillary model system to study alveolar re-epithelialization. Exp Cell Res 2012; 319:64-74. [PMID: 23022369 DOI: 10.1016/j.yexcr.2012.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/28/2012] [Accepted: 09/18/2012] [Indexed: 11/22/2022]
Abstract
In the present study an in vitro bilayer model system of the pulmonary alveolocapillary barrier was established to investigate the role of the microvascular endothelium on re-epithelialization. The model system, confluent monolayer cultures on opposing sides of a porous membrane, consisted of a human microvascular endothelial cell line (HPMEC-ST1.6R) and an alveolar type II like cell line (A549), stably expressing EGFP and mCherry, respectively. These fluorescent proteins allowed the real time assessment of the integrity of the monolayers and the automated analysis of the wound healing process after a scratch injury. The HPMECs significantly attenuated the speed of re-epithelialization, which was associated with the proximity to the A549 layer. Examination of cross-sectional transmission electron micrographs of the model system revealed protrusions through the membrane pores and close contact between the A549 cells and the HPMECs. Immunohistochemical analysis showed that these close contacts consisted of heterocellular gap-, tight- and adherens-junctions. Additional analysis, using a fluorescent probe to assess gap-junctional communication, revealed that the HPMECs and A549 cells were able to exchange the fluorophore, which could be abrogated by disrupting the gap junctions using connexin mimetic peptides. These data suggest that the pulmonary microvascular endothelium may impact the re-epithelialization process.
Collapse
|
32
|
Fine KL, Metcalfe MG, White E, Virji M, Karls RK, Quinn FD. Involvement of the autophagy pathway in trafficking of Mycobacterium tuberculosis bacilli through cultured human type II epithelial cells. Cell Microbiol 2012; 14:1402-14. [PMID: 22519722 DOI: 10.1111/j.1462-5822.2012.01804.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/26/2012] [Accepted: 04/15/2012] [Indexed: 11/29/2022]
Abstract
Interactions between Mycobacterium tuberculosis bacilli and alveolar macrophages have been extensively characterized, while similar analyses in epithelial cells have not been performed. In this study, we microscopically examined endosomal trafficking of M. tuberculosis strain Erdman in A549 cells, a human type II pneumocyte cell line. Immuno-electron microscopic (IEM) analyses indicate that M. tuberculosis bacilli are internalized to a compartment labelled first with Rab5 and then with Rab7 small GTPase proteins. This suggests that, unlike macrophages, M. tuberculosis bacilli traffic to late endosomes in epithelial cells. However, fusion of lysosomes with the bacteria-containing compartment appears to be inhibited, as illustrated by IEM studies employing LAMP-2 and cathepsin-L antibodies. Examination by transmission electron microscopy and IEM revealed M. tuberculosis-containing compartments surrounded by double membranes and labelled with antibodies against the autophagy marker Lc3, providing evidence for involvement and intersection of the autophagy and endosomal pathways. Interestingly, inhibition of the autophagy pathway using 3-methyladenine improved host cell viability and decreased numbers of viable intracellular bacteria recovered after 72 h post infection. Collectively, these data suggest that trafficking patterns for M. tuberculosis bacilli in alveolar epithelial cells differ from macrophages, and that autophagy is involved this process.
Collapse
Affiliation(s)
- Kari L Fine
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Cellular bilayer models can be used to simulate many biological compartments. Here, we describe a cell culture model of the human alveolus that enables the study of early invasive pulmonary aspergillosis. The cellular bilayer is constructed with human alveolar epithelial cells and human pulmonary artery endothelial cells. The cells are grown on a semipermeable polyester membrane. This model can be used to study the pathogenesis, immunobiology and pharmacology of invasive pulmonary aspergillosis.
Collapse
|
34
|
Davidson LB, Nessar R, Kempaiah P, Perkins DJ, Byrd TF. Mycobacterium abscessus glycopeptidolipid prevents respiratory epithelial TLR2 signaling as measured by HβD2 gene expression and IL-8 release. PLoS One 2011; 6:e29148. [PMID: 22216191 PMCID: PMC3244437 DOI: 10.1371/journal.pone.0029148] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Accepted: 11/21/2011] [Indexed: 11/18/2022] Open
Abstract
Mycobacterium abscessus has emerged as an important cause of lung infection, particularly in patients with bronchiectasis. Innate immune responses must be highly effective at preventing infection with M. abscessus because it is a ubiquitous environmental saprophyte and normal hosts are not commonly infected. M. abscessus exists as either a glycopeptidolipid (GPL) expressing variant (smooth phenotype) in which GPL masks underlying bioactive cell wall lipids, or as a variant lacking GPL which is immunostimulatory and invasive in macrophage infection models. Respiratory epithelium has been increasingly recognized as playing an important role in the innate immune response to pulmonary pathogens. Respiratory epithelial cells express toll-like receptors (TLRs) which mediate the innate immune response to pulmonary pathogens. Both interleukin-8 (IL-8) and human β-defensin 2 (HβD2) are expressed by respiratory epithelial cells in response to toll-like receptor 2 (TLR2) receptor stimulation. In this study, we demonstrate that respiratory epithelial cells respond to M. abscessus variants lacking GPL with expression of IL-8 and HβD2. Furthermore, we demonstrate that this interaction is mediated through TLR2. Conversely, M. abscessus expressing GPL does not stimulate expression of IL-8 or HβD2 by respiratory epithelial cells which is consistent with "masking" of underlying bioactive cell wall lipids by GPL. Because GPL-expressing smooth variants are the predominant phenotype existing in the environment, this provides an explanation whereby initial M. abscessus colonization of abnormal lung airways escapes detection by the innate immune system.
Collapse
Affiliation(s)
- Lisa B. Davidson
- Department of Medicine, New Mexico Veterans Health Care System, Albuquerque, New Mexico, United States of America
| | - Rachid Nessar
- Universite Paris V-Descartes, Faculté de Medecine, Paris, France
| | - Prakasha Kempaiah
- The University of New Mexico School of Medicine, Center for Global Health, Albuquerque, New Mexico, United States of America
| | - Douglas J. Perkins
- The University of New Mexico School of Medicine, Center for Global Health, Albuquerque, New Mexico, United States of America
| | - Thomas F. Byrd
- Department of Medicine, New Mexico Veterans Health Care System, Albuquerque, New Mexico, United States of America
- The University of New Mexico School of Medicine, Division of Infectious Diseases, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
35
|
Nguyen Hoang AT, Chen P, Juarez J, Sachamitr P, Billing B, Bosnjak L, Dahlén B, Coles M, Svensson M. Dendritic cell functional properties in a three-dimensional tissue model of human lung mucosa. Am J Physiol Lung Cell Mol Physiol 2011; 302:L226-37. [PMID: 22101763 DOI: 10.1152/ajplung.00059.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In lung tissue, dendritic cells (DC) are found in close association with the epithelial cell layer, and there is evidence of DC regulation by the epithelium; that epithelial dysfunction leads to overzealous immune cell activation. However, dissecting basic mechanisms of DC interactions with epithelial cells in human tissue is difficult. Here, we describe a method to generate a three-dimensional organotypic model of the human airway mucosa in which we have implanted human DC. The model recapitulates key anatomical and functional features of lung mucosal tissue, including a stratified epithelial cell layer, deposition of extracellular matrix proteins, and the production of tight junction and adherence junction proteins. Labeling of fixed tissue model sections and imaging of live tissue models also revealed that DC distribute in close association with the epithelial layer. As functional properties of DC may be affected by the local tissue microenvironment, this system provides a tool to study human DC function associated with lung mucosal tissue. As an example, we report that the lung tissue model regulates the capacity of DC to produce the chemokines CCL17, CCL18, and CCL22, leading to enhanced CCL18 expression and reduced CCL17 and CCL22 expression. This novel tissue model thus provides a tool well suited for a wide range of studies, including those on the regulation of DC functional properties within the local tissue microenvironment during homeostasis and inflammatory reactions.
Collapse
Affiliation(s)
- Anh Thu Nguyen Hoang
- Center for Infectious Medicine, F59, Dept. of Medicine, Karolinska Institutet, Karolinska Univ. Hospital, Huddinge, 141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Human pulmonary microvascular endothelial cells support productive replication of highly pathogenic avian influenza viruses: possible involvement in the pathogenesis of human H5N1 virus infection. J Virol 2011; 86:667-78. [PMID: 22072765 DOI: 10.1128/jvi.06348-11] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 viruses continue to cause sporadic human infections with a high fatality rate. Respiratory failure due to acute respiratory distress syndrome (ARDS) is a complication among hospitalized patients. Since progressive pulmonary endothelial damage is the hallmark of ARDS, we investigated host responses following HPAI virus infection of human pulmonary microvascular endothelial cells. Evaluation of these cells for the presence of receptors preferred by influenza virus demonstrated that avian-like (α2-3-linked) receptors were more abundant than human-like (α2-6-linked) receptors. To test the permissiveness of pulmonary endothelial cells to virus infection, we compared the replication of selected seasonal, pandemic (2009 H1N1 and 1918), and potentially pandemic (H5N1) influenza virus strains. We observed that these cells support productive replication only of HPAI H5N1 viruses, which preferentially enter through and are released from the apical surface of polarized human endothelial monolayers. Furthermore, A/Thailand/16/2004 and A/Vietnam/1203/2004 (VN/1203) H5N1 viruses, which exhibit heightened virulence in mammalian models, replicated to higher titers than less virulent H5N1 strains. VN/1203 infection caused a significant decrease in endothelial cell proliferation compared to other subtype viruses. VN/1203 virus was also found to be a potent inducer of cytokines and adhesion molecules known to regulate inflammation during acute lung injury. Deletion of the H5 hemagglutinin (HA) multibasic cleavage site did not affect virus infectivity but resulted in decreased virus replication in endothelial cells. Our results highlight remarkable tropism and infectivity of the H5N1 viruses for human pulmonary endothelial cells, resulting in the potent induction of host inflammatory responses.
Collapse
|
37
|
de Souza Carvalho C, Kasmapour B, Gronow A, Rohde M, Rabinovitch M, Gutierrez MG. Internalization, phagolysosomal biogenesis and killing of mycobacteria in enucleated epithelial cells. Cell Microbiol 2011; 13:1234-49. [DOI: 10.1111/j.1462-5822.2011.01615.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
García-Pérez BE, Villagómez-Palatto DA, Castañeda-Sánchez JI, Coral-Vázquez RM, Ramírez-Sánchez I, Ordoñez-Razo RM, Luna-Herrera J. Innate response of human endothelial cells infected with mycobacteria. Immunobiology 2011; 216:925-35. [PMID: 21397978 DOI: 10.1016/j.imbio.2011.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/02/2010] [Accepted: 01/04/2011] [Indexed: 10/18/2022]
Abstract
Endothelial cells are susceptible to infection by several pathogens, but little is known about mycobacterial infection. We analyzed some features of mycobacteria-endothelial cell interactions and the innate response to the infection. Intracellular growth in human umbilical vein endothelial cells (HUVECs) of three Mycobacterium species: M. tuberculosis (MTB), M. abscessus (MAB) and M. smegmatis (MSM) was analyzed. M. smegmatis was eliminated; M. abscessus had an accelerate intracellular replication and M. tuberculosis did not replicate or was eliminated. M. abscessus infection induced profound cytoskeleton rearrangements, with M. tuberculosis infection changes were less marked, and with MSM were slight. Nitric oxide (NO) production was induced differentially: M. abscessus induced the highest levels followed by M. tuberculosis and M. smegmatis; the contrary was true for reactive oxygen species (ROS) production. Only M. tuberculosis infection caused beta-1 defensin over-expression. As a whole, our results describe some aspects of the innate response of HUVEC infected by mycobacteria with different virulence and suggest that a strong cytoskeleton mobilization triggers a high NO production in these cells.
Collapse
Affiliation(s)
- Blanca Estela García-Pérez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Z.P. 11340, México, D.F., Mexico
| | | | | | | | | | | | | |
Collapse
|
39
|
Gloede J, Scheerans C, Derendorf H, Kloft C. In vitro pharmacodynamic models to determine the effect of antibacterial drugs. J Antimicrob Chemother 2009; 65:186-201. [PMID: 20026612 DOI: 10.1093/jac/dkp434] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In vitro pharmacodynamic (PD) models are used to obtain useful quantitative information on the effect of either single drugs or drug combinations against bacteria. This review provides an overview of in vitro PD models and their experimental implementation. Models are categorized on the basis of whether the drug concentration remains constant or changes and whether there is a loss of bacteria from the system. Further subdifferentiation is based on whether bacterial loss involves dilution of the medium or is associated with dialysis or diffusion. For comprehension of the underlying principles, experimental settings are simplified and schematically illustrated, including the simulations of various in vivo routes of administration. The different model types are categorized and their (dis)advantages discussed. The application of in vitro models to special organs, infections and pathogens is comprehensively presented. Finally, the relevance and perspectives of in vitro investigations in drug discovery and clinical research are elucidated and discussed.
Collapse
Affiliation(s)
- Julia Gloede
- Department of Clinical Pharmacy, Institute of Pharmacy, Martin-Luther-Universitaet Halle-Wittenberg, 06120 Halle, Germany
| | | | | | | |
Collapse
|
40
|
Kinhikar AG, Verma I, Chandra D, Singh KK, Weldingh K, Andersen P, Hsu T, Jacobs WR, Laal S. Potential role for ESAT6 in dissemination of M. tuberculosis via human lung epithelial cells. Mol Microbiol 2009; 75:92-106. [PMID: 19906174 DOI: 10.1111/j.1365-2958.2009.06959.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
ESAT6 has recently been demonstrated to cause haemolysis and macrophage lysis. Our studies demonstrate that ESAT6 causes cytolysis of type 1 and type 2 pneumocytes. Both types of pneumocytes express membrane laminin, and ESAT6 exhibits dose-dependent binding to both cell types and to purified human laminin. While minimal ESAT6 was detected on the surface of Mycobacterium tuberculosis grown in vitro, exogenously provided ESAT6 specifically associated with the bacterial cell surface, and the bacterium-associated ESAT6 retained its cytolytic ability. esat6 transcripts were upregulated approximately 4- to approximately 13-fold in bacteria replicating in type 1 cells, and approximately 3- to approximately 5 fold in type 2 cells. In vivo, laminin is primarily concentrated at the basolateral surface of pneumocytes where they rest on the basement membrane, which is composed primarily of laminin and collagen. The upregulation of esat6 transcripts in bacteria replicating in pneumocytes, the specific association of ESAT6 with the bacterial surface, the binding of ESAT6 to laminin and the lysis of pneumocytes by free and bacterium-associated ESAT6 together suggest a scenario wherein Mycobacterium tuberculosis replicating in pneumocytes may utilize surface ESAT6 to anchor onto the basolateral laminin-expressing surface of the pneumocytes, and damage the cells and the basement membrane to directly disseminate through the alveolar wall.
Collapse
Affiliation(s)
- Arvind G Kinhikar
- Department of Pathology, New York University Langone School of Medicine, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
An open-access microfluidic model for lung-specific functional studies at an air-liquid interface. Biomed Microdevices 2009; 11:1081-9. [PMID: 19484389 DOI: 10.1007/s10544-009-9325-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In an effort to improve the physiologic relevance of existing in vitro models for alveolar cells, we present a microfluidic platform which provides an air-interface in a dynamic system combining microfluidic and suspended membrane culture systems. Such a system provides the ability to manipulate multiple parameters on a single platform along with ease in cell seeding and manipulation. The current study presents a comparison of the efficacy of the hybrid system with conventional platforms using assays analyzing the maintenance of function and integrity of A549 alveolar epithelial cell monolayer cultures. The hybrid system incorporates bio-mimetic nourishment on the basal side of the epithelial cells along with an open system on the apical side of the cells exposed to air allowing for easy access for assays.
Collapse
|
42
|
Mühlfeld C, Rothen-Rutishauser B, Blank F, Vanhecke D, Ochs M, Gehr P. Interactions of nanoparticles with pulmonary structures and cellular responses. Am J Physiol Lung Cell Mol Physiol 2008; 294:L817-29. [DOI: 10.1152/ajplung.00442.2007] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Combustion-derived and synthetic nano-sized particles (NSP) have gained considerable interest among pulmonary researchers and clinicians for two main reasons. 1) Inhalation exposure to combustion-derived NSP was associated with increased pulmonary and cardiovascular morbidity and mortality as suggested by epidemiological studies. Experimental evidence has provided a mechanistic picture of the adverse health effects associated with inhalation of combustion-derived and synthetic NSP. 2) The toxicological potential of NSP contrasts with the potential application of synthetic NSP in technological as well as medicinal settings, with the latter including the use of NSP as diagnostics or therapeutics. To shed light on this paradox, this article aims to highlight recent findings about the interaction of inhaled NSP with the structures of the respiratory tract including surfactant, alveolar macrophages, and epithelial cells. Cellular responses to NSP exposure include the generation of reactive oxygen species and the induction of an inflammatory response. Furthermore, this review places special emphasis on methodological differences between experimental studies and the caveats associated with the dose metrics and points out ways to overcome inherent methodological problems.
Collapse
|
43
|
Russell BH, Vasan R, Keene DR, Koehler TM, Xu Y. Potential dissemination of Bacillus anthracis utilizing human lung epithelial cells. Cell Microbiol 2007; 10:945-57. [PMID: 18067609 DOI: 10.1111/j.1462-5822.2007.01098.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dissemination of Bacillus anthracis spores from the lung is a critical early event in the establishment of inhalational anthrax. We recently reported that B. anthracis could adhere to and be internalized by cultured intestinal epithelial and fibroblast cells. Here, using gentamicin protection assays and/or electron microscopy, we found that Sterne strain 7702 spores were able to adhere to and subsequently be internalized by polarized A549 cells and primary human small airway epithelial cells. We showed for the first time that internalized spores were able to survive and that spores could translocate across an A549 cell barrier from the apical side to the basolateral side without disrupting the barrier integrity, suggesting a transcellular route. In addition, dormant spores of fully virulent Ames and UT500 strains were able to adhere to A549 cells at a frequency similar to that of 7702, whereas the capsule in germinated Ames and UT500 spores prevented adherence. Fluorescence microscopy also revealed that dormant Ames spores were internalized at a frequency similar to that of 7702. These findings highlight the possibility of a novel route of dissemination in which B. anthracis utilizes epithelial cells of the lung. The implications of these results to B. anthracis pathogenesis are discussed.
Collapse
Affiliation(s)
- Brooke H Russell
- Center for Extracellular Matrix Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
44
|
Villar CC, Kashleva H, Nobile CJ, Mitchell AP, Dongari-Bagtzoglou A. Mucosal tissue invasion by Candida albicans is associated with E-cadherin degradation, mediated by transcription factor Rim101p and protease Sap5p. Infect Immun 2007; 75:2126-35. [PMID: 17339363 PMCID: PMC1865768 DOI: 10.1128/iai.00054-07] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 02/03/2007] [Accepted: 02/18/2007] [Indexed: 01/12/2023] Open
Abstract
The ability of Candida albicans to invade mucosal tissues is a major virulence determinant of this organism; however, the mechanism of invasion is not understood in detail. Proteolytic breakdown of E-cadherin, the major protein in epithelial cell junctions, has been proposed as a mechanism of invasion of certain bacteria in the oral mucosa. The objectives of this study were (i) to assess whether C. albicans degrades E-cadherin expressed by oral epithelial cells in vitro; (ii) to compare the abilities of strains with different invasive potentials to degrade this protein; and (iii) to investigate fungal virulence factors responsible for E-cadherin degradation. We found that while E-cadherin gene expression was not altered, E-cadherin was proteolytically degraded during the interaction of oral epithelial cells with C. albicans. Moreover, C. albicans-mediated degradation of E-cadherin was completely inhibited in the presence of protease inhibitors. Using a three-dimensional model of the human oral mucosa, we found that E-cadherin was degraded in localized areas of tissue invasion by C. albicans. An invasion-deficient rim101-/rim101- strain was deficient in degradation of E-cadherin, and this finding suggested that proteases may depend on Rim101p for expression. Indeed, reverse transcription-PCR data indicated that expression of the SAP4, SAP5, and SAP6 genes is severely reduced in the rim101-/rim101- mutant. These SAP genes are functional Rim101p targets, because engineered expression of SAP5 in the rim101-/rim101- strain restored E-cadherin degradation and invasion in the mucosal model. Our data support the hypothesis that there is a mechanism by which C. albicans invades mucosal tissues by promoting the proteolytic degradation of E-cadherin in epithelial adherens junctions.
Collapse
Affiliation(s)
- C C Villar
- Department of Periodontology, School of Dental Medicine, University of Connecticut, 263 Farmington Ave., Farmington, CT 06030-1710, USA.
| | | | | | | | | |
Collapse
|
45
|
Birkness KA, Guarner J, Sable SB, Tripp RA, Kellar KL, Bartlett J, Quinn FD. An in vitro model of the leukocyte interactions associated with granuloma formation in Mycobacterium tuberculosis infection. Immunol Cell Biol 2007; 85:160-8. [PMID: 17199112 DOI: 10.1038/sj.icb.7100019] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The principal defense of the human host against a Mycobacterium tuberculosis infection is the formation of granulomas, organized collections of activated macrophages, including epithelioid and multinucleated giant cells, surrounded by lymphocytes. This granuloma can sequester and contain the bacteria preventing active disease, and if the granuloma is maintained, these bacteria may remain latent for a person's lifetime. Secretion of a variety of chemoattractant cytokines following phagocytosis of the bacilli by the macrophage is critical not only to the formation of the granuloma but also to its maintenance. To investigate this process of early granuloma formation, we developed an in vitro model composed entirely of human cells. Combining blood lymphocytes and autologous macrophages from healthy purified protein derivative skin test-negative individuals and mycobacteria resulted in the formation of small, rounded aggregate structures. Microscopic examination found macrophage-specific CD68(+) epithelioid macrophages and small round CD3(+) lymphocytes that in complex resembled small granulomas seen in clinical pathology specimens. Acid-fast staining bacteria were observed between and possibly within the cells composing the granulomas. Supernatants from the infected cells collected at 24 and 48 h and 5 and 9 days after infection were analyzed by a multiplexed cytokine bead-based assay using the Luminex 100 and were found to contain interleukin (IL)-6, IL-8, interferon-gamma and tumor necrosis factor-alpha, cytokines known to be involved in human granuloma formation, in quantities from two-fold to 7000-fold higher than supernatants from uninfected control cells. In addition, chemotaxis assays demonstrated that the same supernatants attracted significantly more human peripheral blood mononuclear cells than those of uninfected cells (P<0.001). This model may provide insight into the earliest stages of granuloma formation in those newly infected.
Collapse
Affiliation(s)
- Kristin A Birkness
- Mycobacteriology Laboratory Branch, Division of Tuberculosis Elimination, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Mehta PK, Karls RK, White EH, Ades EW, Quinn FD. Entry and intracellular replication of Mycobacterium tuberculosis in cultured human microvascular endothelial cells. Microb Pathog 2006; 41:119-24. [PMID: 16860530 DOI: 10.1016/j.micpath.2006.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Revised: 05/18/2006] [Accepted: 05/22/2006] [Indexed: 11/25/2022]
Abstract
Establishment of pulmonary Mycobacterium tuberculosis infection requires evasion of host innate defenses. In the lung alveoli, epithelial cells naturally resist uptake by the inhaled bacilli while macrophages patrol the epithelial surface and phagocytose foreign microbes. Alveolar microvascular endothelial cells, however, have not been examined as a potential point of direct interaction with the bacilli. It has been shown with other bacterial and viral lung pathogens that the lung endothelial cells are not only a point of interaction, but a source for intracellular replication and chronic infection by the pathogen. To investigate if endothelial cells are susceptible to M. tuberculosis infection, we examined attachment, internalization, and intracellular replication of M. tuberculosis bacilli in an immortalized human lung microvascular endothelial cell line (HULEC). By 6 h post-infection, 12% of infecting bacilli were associated with the HULEC monolayer cells. This was twice the association observed following a similar infection with cells from a human foreskin microvascular endothelial cell line (HMEC-1). As measured by survival after the addition of a high extracellular concentration of the aminoglycoside amikacin, approximately one-third of the associated bacilli were internalized and unavailable to the drug in both cell lines. Using electron microscopy, large numbers of bacilli were visible in the vacuoles of HULEC cells after 48 h post-infection; the presence of bacterial septa between adjacent mycobacteria suggests intracellular replication. These in vitro findings support the hypothesis that lung endothelial cells have the potential to participate in in vivo lung infections.
Collapse
Affiliation(s)
- Parmod K Mehta
- Division of Tuberculosis Elimination, National Center for AIDS, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | | | | | | | | |
Collapse
|
47
|
Villar CC, Kashleva H, Mitchell AP, Dongari-Bagtzoglou A. Invasive phenotype of Candida albicans affects the host proinflammatory response to infection. Infect Immun 2005; 73:4588-95. [PMID: 16040970 PMCID: PMC1201248 DOI: 10.1128/iai.73.8.4588-4595.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Candida albicans is a major opportunistic pathogen in immunocompromised patients. Production of proinflammatory cytokines by host cells in response to C. albicans plays a critical role in the activation of immune cells and final clearance of the organism. Invasion of host cells and tissues is considered one of the virulence attributes of this organism. The purpose of this study was to investigate whether the ability of C. albicans to invade host cells and tissues affects the proinflammatory cytokine responses by epithelial and endothelial cells. In this study we used the invasion-deficient RIM101 gene knockout strain DAY25, the highly invasive strain SC5314, and highly invasive RIM101-complemented strain DAY44 to compare the proinflammatory cytokine responses by oral epithelial or endothelial cells. Using a high-throughput approach, we found both qualitative and quantitative differences in the overall inflammatory responses to C. albicans strains with different invasive potentials. Overall, the highly invasive strains triggered higher levels of proinflammatory cytokines in host cells than the invasion-deficient mutant triggered. Significant differences compared to the attenuated mutant were noted in interleukin-1alpha (IL-1alpha), IL-6, IL-8, and tumor necrosis factor alpha in epithelial cells and in IL-6, growth-related oncogene, IL-8, monocyte chemoattractant protein 1 (MCP-1), MCP-2, and granulocyte colony-stimulating factor in endothelial cells. Our results indicate that invasion of host cells and tissues by C. albicans enhances the host proinflammatory response to infection.
Collapse
Affiliation(s)
- C C Villar
- Department of Periodontology, School of Dental Medicine, University of Connecticut, 263 Farmington Ave., Farmington, CT 06030-1710, USA
| | | | | | | |
Collapse
|
48
|
Rothen-Rutishauser BM, Kiama SG, Gehr P. A Three-Dimensional Cellular Model of the Human Respiratory Tract to Study the Interaction with Particles. Am J Respir Cell Mol Biol 2005; 32:281-9. [PMID: 15640437 DOI: 10.1165/rcmb.2004-0187oc] [Citation(s) in RCA: 238] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A novel triple co-culture model of the human airway barrier was designed to simulate the cellular part of the air-blood barrier of the respiratory tract represented by macrophages, epithelial cells, and dendritic cells. When epithelial cells (A549 cells) were grown on filter inserts with pores of 3.0 mum in diameter in a two-chamber system, they formed monolayers with polarization into apical and basolateral domains. The epithelial cell cultures were then supplemented with human blood monocyte-derived macrophages and dendritic cells on the apical and basal aspect, respectively. The single-cell cultures as well as the triple co-cultures were characterized in terms of a number of typical features, for example, morphology of cell types, integrity of epithelial layer, and expression of specific cell surface markers (CD14 for macrophages and CD86 for dendritic cells). The interplay of epithelial cells with macrophages and dendritic cells during the uptake of polystyrene particles (1 mum in diameter) was investigated with confocal laser scanning and conventional transmission electron microscopy. Particles were found in all three cell types, although dendritic cells were not directly exposed to the particles. More investigations are needed to understand the translocation pathway.
Collapse
|
49
|
Chang B, Amemura-Maekawa J, Kura F, Kawamura I, Watanabe H. Expression of IL-6 and TNF-α in human alveolar epithelial cells is induced by invading, but not by adhering, Legionella pneumophila. Microb Pathog 2004; 37:295-302. [PMID: 15619425 DOI: 10.1016/j.micpath.2004.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2004] [Accepted: 10/08/2004] [Indexed: 02/04/2023]
Abstract
Legionella pnemophila causes atypical pneumonia in humans, especially in patients with chronic pulmonary diseases and underlying immunosuppression, and in elderly people. Several previous studies have shown that L. pneumophila induced several inflammatory cytokines in murine macrophages, but little is known about cytokine induction by the bacterium in lung epithelial cells. In this study, we investigated the ability of L. pneumophila to stimulate the production of pro-inflammatory cytokines in the human A549 alveolar epithelial cell line during 24h exposure to 10(6), 10(7), and 10(8) microbes. Infection of the wild L. pneumophila strain to A549 resulted in increased levels of interleukin-8 (IL-8), IL-6, and tumor necrosis factor alpha (TNF-alpha) mRNA, and also the secretion of their production into culture medium. In contrast, the level of mRNAs and proteins of IL-1beta and gamma interferon (IFN-gamma) remained unchanged and undetected, respectively. Production of IL-8, IL-6, and TNF-alpha in A549 decreased when an icmE multiplication-less mutant and the heat-killed L. pneumophila strain were inoculated. The treatment of cytochalasin D, which effectively inhibited invasion of L. pneumophila into A549, significantly reduced the production of IL-6 and TNF-alpha, but not IL-8. These results suggested that the induction and expression of IL-6 and TNF-alpha in the human alveolar epithelial cells especially required intracellular signaling by L. pneumophila after invasion.
Collapse
Affiliation(s)
- Bin Chang
- Department of Bacteriology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | |
Collapse
|
50
|
Hermanns MI, Unger RE, Kehe K, Peters K, Kirkpatrick CJ. Lung epithelial cell lines in coculture with human pulmonary microvascular endothelial cells: development of an alveolo-capillary barrier in vitro. J Transl Med 2004; 84:736-52. [PMID: 15077120 DOI: 10.1038/labinvest.3700081] [Citation(s) in RCA: 213] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We have established a coculture system of human distal lung epithelial cells and human microvascular endothelial cells in order to study the cellular interactions of epithelium and endothelium at the alveolocapillary barrier in both pathogenesis and recovery from acute lung injury. The aim was to determine conditions for the development of functional cellular junctions and the formation of a tight epithelial barrier similar to that observed in vivo. The in vitro coculture system consisted of monolayers of human lung epithelial cell lines (A549 or NCI H441) and primary human pulmonary microvascular endothelial cells (HPMEC) on opposite sides of a permeable filter membrane. A549 failed to show sufficient differentiation with respect to formation of a tight epithelial barrier with intact cell-cell junctions. Stimulated with dexamethasone, the cocultures of NCI H441 and HPMEC established contact-inhibited differentiated monolayers, with NCI H441 showing a continuous, circumferential immunostaining of the tight junctional protein, ZO-1 and the adherens junction protein, E-cadherin. The generation of a polarized epithelial cell monolayer with typical junctional structures was confirmed by transmission electron microscopy. Dexamethasone treatment resulted in average transbilayer electrical resistance (TER) values of 500 Omega cm(2) after 10-12 days of cocultivation and correlated with a reduced flux of the hydrophilic permeability marker, sodium-fluorescein. In addition, basolateral distribution of the proinflammatory cytokine tumour necrosis factor-alpha caused a significant reduction of TER-values after 24 h exposure. This decrease in TER could be re-established to control level by removal of the cytokine within 24 h. Thus, the coculture system of the NCI H441 with HPMEC should be a suitable in vitro model system to examine epithelial and endothelial interactions in the pathogenesis of acute lung injury, infectious lung diseases and toxic lung injury. In addition, it could be used to improve techniques of lung drug delivery that also requires a functional barrier.
Collapse
|