1
|
Lian E, Belardinelli JM, De K, Pandurangan AP, Angala SK, Palčeková Z, Grzegorzewicz AE, Bryant JM, Blundell TL, Parkhill J, Floto RA, Wheat WH, Jackson M. Cell envelope polysaccharide modifications alter the surface properties and interactions of Mycobacterium abscessus with innate immune cells in a morphotype-dependent manner. mBio 2025; 16:e0032225. [PMID: 40084888 PMCID: PMC11980365 DOI: 10.1128/mbio.00322-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Mycobacterium abscessus is one of the leading causes of pulmonary infections caused by non-tuberculous mycobacteria. The ability of M. abscessus to establish a chronic infection in the lung relies on a series of adaptive mutations impacting, in part, global regulators and cell envelope biosynthetic enzymes. One of the genes under strong evolutionary pressure during host adaptation is ubiA, which participates in the elaboration of the arabinan domains of two major cell envelope polysaccharides: arabinogalactan (AG) and lipoarabinomannan (LAM). We here show that patient-derived UbiA mutations not only cause alterations in the AG, LAM, and mycolic acid contents of M. abscessus but also tend to render the bacterium more prone to forming biofilms while evading uptake by innate immune cells and enhancing their pro-inflammatory properties. The fact that the effects of UbiA mutations on the physiology and pathogenicity of M. abscessus were impacted by the rough or smooth morphotype of the strain suggests that the timing of their selection relative to morphotype switching may be key to their ability to promote chronic persistence in the host.IMPORTANCEMultidrug-resistant pulmonary infections caused by Mycobacterium abscessus and subspecies are increasing in the U.S.A. and globally. Little is known of the mechanisms of pathogenicity of these microorganisms. We have identified single-nucleotide polymorphisms (SNPs) in a gene involved in the biosynthesis of two major cell envelope polysaccharides, arabinogalactan and lipoarabinomannan, in lung-adapted isolates from 13 patients. Introduction of these individual SNPs in a reference M. abscessus strain allowed us to study their impact on the physiology of the bacterium and its interactions with immune cells. The significance of our work is in identifying some of the mechanisms used by M. abscessus to colonize and persist in the human lung, which will facilitate the early detection of potentially more virulent clinical isolates and lead to new therapeutic strategies. Our findings may further have broader biomedical impacts, as the ubiA gene is conserved in other tuberculous and non-tuberculous mycobacterial pathogens.
Collapse
Affiliation(s)
- Elena Lian
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Juan M. Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Kavita De
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Arun Prasad Pandurangan
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Trumpington, Cambridge, UK
- Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
| | - Shiva K. Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Zuzana Palčeková
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Anna E. Grzegorzewicz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | | | - Tom L. Blundell
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Trumpington, Cambridge, UK
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - R. Andres Floto
- Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Trumpington, Cambridge, UK
| | - William H. Wheat
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
2
|
Bahlool AZ, Cavanagh B, Sullivan AO, MacLoughlin R, Keane J, Sullivan MPO, Cryan SA. Microfluidics produced ATRA-loaded PLGA NPs reduced tuberculosis burden in alveolar epithelial cells and enabled high delivered dose under simulated human breathing pattern in 3D printed head models. Eur J Pharm Sci 2024; 196:106734. [PMID: 38417586 DOI: 10.1016/j.ejps.2024.106734] [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: 09/26/2023] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is second only to COVID-19 as the top infectious disease killer worldwide. Multi-drug resistant TB (MDR-TB) may arise because of poor patient adherence to medications due to lengthy treatment duration and side effects. Delivering novel host directed therapies (HDT), like all trans retinoic acid (ATRA) may help to improve drug regimens and reduce the incidence of MDR-TB. Local delivery of ATRA to the site of infection leads to higher bioavailability and reduced systemic side effects. ATRA is poorly soluble in water and has a short half-life in plasma. Therefore, it requires a formulation step before it can be administered in vivo. ATRA loaded PLGA nanoparticles suitable for nebulization were manufactured and optimized using a scalable nanomanufacturing microfluidics (MF) mixing approach (MF-ATRA-PLGA NPs). MF-ATRA-PLGA NPs demonstrated a dose dependent inhibition of Mtb growth in TB-infected A549 alveolar epithelial cell model while preserving cell viability. The MF-ATRA-PLGA NPs were nebulized with the Aerogen Solo vibrating mesh nebulizer, with aerosol droplet size characterized using laser diffraction and the estimated delivered dose was determined. The volume median diameter (VMD) of the MF-ATRA-PLGA NPs was 3.00 ± 0.18 μm. The inhaled dose delivered in adult and paediatric 3D printed head models under a simulated normal adult and paediatric breathing pattern was found to be 47.05 ± 3 % and 20.15 ± 3.46 % respectively. These aerosol characteristics of MF-ATRA-PLGA NPs supports its suitability for delivery to the lungs via inhalation. The data generated on the efficacy of an inhalable, scalable and regulatory friendly ATRA-PLGA NPs formulation provides a foundation on which further pre-clinical testing can be built. Overall, the results of this project are promising for future research into ATRA loaded NPs formulations as inhaled host directed therapies for TB.
Collapse
Affiliation(s)
- Ahmad Z Bahlool
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland RCSI, Dublin 2, Ireland
| | - Andrew O' Sullivan
- Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Mary P O' Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI and Trinity College Dublin, Dublin, Ireland; SFI Centre for Research in Medical Devices (CÚRAM), NUIG & RCSI, Dublin, Ireland.
| |
Collapse
|
3
|
Ivana Romina S, Benjamín DLCT, Melina Mara M, Gladys Ester G. Quantification of rifampicin loaded into inhaled polymeric nanoparticles by reversed phase high-performance liquid chromatography in pulmonary nonphagocytic cellular uptake. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1908-1915. [PMID: 38494905 DOI: 10.1039/d4ay00234b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Rifampicin is an antibiotic effective against both Gram-negative and Gram-positive bacteria and is commonly used as a first-line treatment for tuberculosis caused by Mycobacterium tuberculosis. In this study, a reversed-phase high-performance liquid chromatography method was developed and validated to assess rifampicin, either free or combined with ascorbic acid, loaded into chitosan/Tween 80-coated alginate nanoparticles. The method utilized a reversed-phase C18 Restek column with specific chromatographic conditions: a mobile phase of 60 : 40 ratios of methanol/buffer phosphate (pH 7.0), at a flow rate of 0.8 mL min-1, and an injection volume of 15 μL. rifampicin and the internal standard (rifamycin) had retention times of 4.0 and 2.5 min, respectively, and were detected at 334 nm. The method demonstrated the stability of stored samples after freezing-thawing cycles and specificity for rifampicin, even in the presence of degradation products from stress conditions. The high-performance liquid chromatography method was found to be specific, precise, robust, and sensitive. Results indicated that rifampicin accumulation and uptake kinetics varied based on cell type, formulation (free or loaded in nanoparticles), rifampicin concentration, and incubation time. Confocal fluorescence microscopy images supported these findings, showing isothiocyanate fluorescein nanoparticles distribution in different intracellular regions depending on the cell type used. The societal impact of this research lies in its potential to advance the treatment of respiratory infectious diseases, such as tuberculosis, through the development of more effective and specific drug delivery methods. By optimizing the way drugs, particularly rifampicin in this case, interact with lung cells, we aim to achieve greater treatment efficacy and alleviate the overall burden of disease. Furthermore, our study offers novel insights into the intracellular behavior of rifampin from polymeric nanoparticles, paving the way for personalized medicine approaches in the treatment of respiratory infections. This dual focus on social impact and innovation underscores our commitment to improving global health outcomes and addressing pressing public health challenges.
Collapse
Affiliation(s)
- Scolari Ivana Romina
- Unidad de Investigación Y Desarrollo en Tecnología Farmacéutica (UNITEFA - CONICET), Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, X5000HUA, Argentina.
| | - De La Cruz-Thea Benjamín
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Córdoba, Argentina
| | - Musri Melina Mara
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Córdoba, Argentina
- Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Granero Gladys Ester
- Unidad de Investigación Y Desarrollo en Tecnología Farmacéutica (UNITEFA - CONICET), Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, X5000HUA, Argentina.
| |
Collapse
|
4
|
Alves-Barroco C, Botelho AMN, Américo MA, Fracalanzza SEL, de Matos APA, Guimaraes MA, Ferreira-Carvalho BT, Figueiredo AMS, Fernandes AR. Assessing in vivo and in vitro biofilm development by Streptococcus dysgalactiae subsp. dysgalactiae using a murine model of catheter-associated biofilm and human keratinocyte cell. Front Cell Infect Microbiol 2022; 12:874694. [PMID: 35928206 PMCID: PMC9343579 DOI: 10.3389/fcimb.2022.874694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus dysgalactiae subsp. dysgalactiae (SDSD) is an important agent of bovine mastitis. This infection causes an inflammatory reaction in udder tissue, being the most important disease-causing significant impact on the dairy industry. Therefore, it leads to an increase in dairy farming to meet commercial demands. As a result, there is a major impact on both the dairy industry and the environment including global warming. Recurrent mastitis is often attributed to the development of bacterial biofilms, which promote survival of sessile cells in hostile environments, and resistance to the immune system defense and antimicrobial therapy. Recently, we described the in vitro biofilm development on abiotic surfaces by bovine SDSD. In that work we integrated microbiology, imaging, and computational methods to evaluate the biofilm production capability of SDSD isolates on abiotic surfaces. Additionally, we reported that bovine SDSD can adhere and internalize human cells, including human epidermal keratinocyte (HEK) cells. We showed that the adherence and internalization rates of bovine SDSD isolates in HEK cells are higher than those of a SDSD DB49998-05 isolated from humans. In vivo, bovine SDSD can cause invasive infections leading to zebrafish morbidity and mortality. In the present work, we investigated for the first time the capability of bovine SDSD to develop biofilm in vivo using a murine animal model and ex-vivo on human HEK cells. Bovine SDSD isolates were selected based on their ability to form weak, moderate, or strong biofilms on glass surfaces. Our results showed that SDSD isolates displayed an increased ability to form biofilms on the surface of catheters implanted in mice when compared to in vitro biofilm formation on abiotic surface. A greater ability to form biofilm in vitro after animal passage was observed for the VSD45 isolate, but not for the other isolates tested. Besides that, in vitro scanning electron microscopy demonstrated that SDSD biofilm development was visible after 4 hours of SDSD adhesion to HEK cells. Cell viability tests showed an important reduction in the number of HEK cells after the formation of SDSD biofilms. In this study, the expression of genes encoding BrpA-like (biofilm regulatory protein), FbpA (fibronectin-binding protein A), HtrA (serine protease), and SagA (streptolysin S precursor) was higher for biofilm grown in vivo than in vitro, suggesting a potential role for these virulence determinants in the biofilm-development, host colonization, and SDSD infections. Taken together, these results demonstrate that SDSD can develop biofilms in vivo and on the surface of HEK cells causing important cellular damages. As SDSD infections are considered zoonotic diseases, our data contribute to a better understanding of the role of biofilm accumulation during SDSD colonization and pathogenesis not only in bovine mastitis, but they also shed some lights on the mechanisms of prosthesis-associated infection and cellulitis caused by SDSD in humans, as well.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO - Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Caparica, Portugal
- i4HB, Associate Laboratory - Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ana Maria Nunes Botelho
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marco Antonio Américo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - António P. Alves de Matos
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz - Cooperativa de Ensino Superior CRL, Quinta da Granja, Portugal
| | - Márcia Aparecida Guimaraes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Agnes Marie Sá Figueiredo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Alexandra R. Fernandes, ; Agnes Marie Sá Figueiredo,
| | - Alexandra R. Fernandes
- UCIBIO - Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Caparica, Portugal
- i4HB, Associate Laboratory - Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- *Correspondence: Alexandra R. Fernandes, ; Agnes Marie Sá Figueiredo,
| |
Collapse
|
5
|
Paredes-Rojas A, Palma-Ramos A, Castrillón-Rivera LE, Mendoza-Pérez F, Navarro-González MDC, Arenas-Guzmán R, Castañeda-Sánchez JI, Luna-Herrera J. Keratinocyte Response to Infection with Sporothrix schenckii. J Fungi (Basel) 2022; 8:jof8050437. [PMID: 35628694 PMCID: PMC9143681 DOI: 10.3390/jof8050437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/27/2022] Open
Abstract
Sporotrichosis is a subacute, or chronic mycosis caused by traumatic inoculation of material contaminated with the fungus Sporothrix schenckii which is part of the Sporothrix spp. complex. The infection is limited to the skin, although its progression to more severe systemic or disseminated forms remains possible. Skin is the tissue that comes into contact with Sporothrix first, and the role of various cell lines has been described with regard to infection control. However, there is little information on the response of keratinocytes. In this study, we used the human keratinocyte cell line (HaCaT) and evaluated different aspects of infection from modifications in the cytoskeleton to the expression of molecules of the innate response during infection with conidia and yeast cells of Sporothrix schenckii. We found that during infection with both phases of the fungus, alterations of the actin cytoskeleton, formation of membrane protuberances, and loss of stress fibers were induced. We also observed an overexpression of the surface receptors MR, TLR6, CR3 and TLR2. Cytokine analysis showed that both phases of the fungus induced the production of elevated levels of the chemokines MCP-1 and IL-8, and proinflammatory cytokines IFN-α, IFN-γ and IL-6. In contrast, TNF-α production was significant only with conidial infection. In late post-infection, cytokine production was observed with immunoregulatory activity, IL-10, and growth factors, G-CSF and GM-CSF. In conclusion, infection of keratinocytes with conidia and yeast cells of Sporothrix schenckii induces an inflammatory response and rearrangements of the cytoskeleton.
Collapse
Affiliation(s)
- Araceli Paredes-Rojas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Alejandro Palma-Ramos
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Laura Estela Castrillón-Rivera
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - María del Carmen Navarro-González
- Laboratorio de Investigación en Enfermedades Reumáticas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Roberto Arenas-Guzmán
- Sección de Micología, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico;
| | - Jorge Ismael Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| | - Julieta Luna-Herrera
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| |
Collapse
|
6
|
Shantal CJN, Juan CC, Lizbeth BUS, Carlos HGJ, Estela GPB. Candida glabrata is a successful pathogen: an artist manipulating the immune response. Microbiol Res 2022; 260:127038. [DOI: 10.1016/j.micres.2022.127038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
|
7
|
Wen D, Cui J, Li P, Xiong Q, Chen G, Wu C. Syndecan-4 assists Mycobacterium tuberculosis entry into lung epithelial cells by regulating the Cdc42, N-WASP, and Arp2/3 signaling pathways. Microbes Infect 2022; 24:104931. [PMID: 35026388 DOI: 10.1016/j.micinf.2022.104931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 09/14/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022]
Abstract
Syndecan-4 (SDC4) is a transmembrane heparin sulfate proteoglycan that regulates inflammatory responses, cell motility, cell adhesion and intracellular signaling. In this study, we found that overexpression of SDC4 promoted the infection efficiency of Mycobacterium tuberculosis (Mtb), whereas knockdown of SDC4 reduced the infection efficiency, suggesting that SDC4 assisted Mtb infection of epithelial cells. We also observed that Mtb infection affected the F-actin/G-actin ratio, which was also correlated with SDC4 expression levels. Analysis of the Cdc42, N-WASP, and Arp2/3 signaling pathways during Mtb infection revealed that knockdown of Cdc42 and N-WASP or the addition of ZCL278, Wiskostatin or CK636 (blockers of Cdc42, N-WASP, and Arp2/3, respectively) significantly exacerbated Mtb infection in lung epithelial cells. Taken together, our data indicate that SDC4 assists Mtb infection of epithelial cells by regulating the Cdc42, N-WASP, and Arp2/3 signaling pathways, which regulate the polymerization of the actin cytoskeleton.
Collapse
Affiliation(s)
- Da Wen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Jia Cui
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; Department of Microbiology, Changzhi Medical College, Changzhi 046000, China
| | - Ping Li
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Qiuhong Xiong
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Guangxin Chen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| | - Changxin Wu
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; Key Lab of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan 030006, China; The Provincial Key Laboratories for Prevention and Treatment of Major Infectious Diseases, Taiyuan 030006, Shanxi, China.
| |
Collapse
|
8
|
Mast cells modulate early responses to Mycobacterium bovis Bacillus Calmette-Guerin by phagocytosis and formation of extracellular traps. Cell Immunol 2021; 365:104380. [PMID: 34049012 DOI: 10.1016/j.cellimm.2021.104380] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/15/2022]
Abstract
The early interactions between the vaccine Mycobacterium bovis Bacillus Calmette Guerin (BCG) and host peripheral innate immune cells like Mast cells (MCs) may pave the way for generating appropriate protective innate and adaptive immune responses. Mice on administration of BCG by intratracheal instillation showed a massive increase in MC numbers in the infected lung. In vitro co-culture of BCG and rodent Rat Basophilic Leukaemia (RBL-2H3) MCs led to significant killing of BCG. RBL-2H3 MCs were able to phagocytose BCG, take up BCG-derived antigens by macropinocytosis, generate Reactive Oxygen Species (ROS) and degranulate. Further, a few MCs died and released MC extracellular traps (MCETs) having DNA, histones and tryptase to trap BCG. This study highlights the multi-pronged effector responses of MCs on encountering BCG. These responses or their evasion may lead to success or failure of BCG vaccine to provide long term immunity to infections.
Collapse
|
9
|
Chang YY, Enninga J, Stévenin V. New methods to decrypt emerging macropinosome functions during the host-pathogen crosstalk. Cell Microbiol 2021; 23:e13342. [PMID: 33848057 PMCID: PMC8365644 DOI: 10.1111/cmi.13342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Large volumes of liquid and other materials from the extracellular environment are internalised by eukaryotic cells via an endocytic process called macropinocytosis. It is now recognised that this fundamental and evolutionarily conserved pathway is hijacked by numerous intracellular pathogens as an entry portal to the host cell interior. Yet, an increasing number of additional cellular functions of macropinosomes in pathologic processes have been reported beyond this role for fluid internalisation. It emerges that the identity of macropinosomes can vary hugely and change rapidly during their lifetime. A deeper understanding of this important multi-faceted compartment is based on novel methods for their investigation. These methods are either imaging-based for the tracking of macropinosome dynamics, or they provide the means to extract macropinosomes at high purity for comprehensive proteomic analyses. Here, we portray these new approaches for the investigation of macropinosomes. We document how these method developments have provided insights for a new understanding of the intracellular lifestyle of the bacterial pathogens Shigella and Salmonella. We suggest that a systematic complete characterisation of macropinosome subversion with these approaches during other infection processes and pathologies will be highly beneficial for our understanding of the underlying cellular and molecular processes.
Collapse
Affiliation(s)
- Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France
| | - Virginie Stévenin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Université Paris Diderot, Ecole doctorale BioSPC, Paris, France
| |
Collapse
|
10
|
Allué-Guardia A, García JI, Torrelles JB. Evolution of Drug-Resistant Mycobacterium tuberculosis Strains and Their Adaptation to the Human Lung Environment. Front Microbiol 2021; 12:612675. [PMID: 33613483 PMCID: PMC7889510 DOI: 10.3389/fmicb.2021.612675] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
In the last two decades, multi (MDR), extensively (XDR), extremely (XXDR) and total (TDR) drug-resistant Mycobacterium tuberculosis (M.tb) strains have emerged as a threat to public health worldwide, stressing the need to develop new tuberculosis (TB) prevention and treatment strategies. It is estimated that in the next 35 years, drug-resistant TB will kill around 75 million people and cost the global economy $16.7 trillion. Indeed, the COVID-19 pandemic alone may contribute with the development of 6.3 million new TB cases due to lack of resources and enforced confinement in TB endemic areas. Evolution of drug-resistant M.tb depends on numerous factors, such as bacterial fitness, strain's genetic background and its capacity to adapt to the surrounding environment, as well as host-specific and environmental factors. Whole-genome transcriptomics and genome-wide association studies in recent years have shed some insights into the complexity of M.tb drug resistance and have provided a better understanding of its underlying molecular mechanisms. In this review, we will discuss M.tb phenotypic and genotypic changes driving resistance, including changes in cell envelope components, as well as recently described intrinsic and extrinsic factors promoting resistance emergence and transmission. We will further explore how drug-resistant M.tb adapts differently than drug-susceptible strains to the lung environment at the cellular level, modulating M.tb-host interactions and disease outcome, and novel next generation sequencing (NGS) strategies to study drug-resistant TB.
Collapse
Affiliation(s)
- Anna Allué-Guardia
- Population Health Program, Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| | | | - Jordi B. Torrelles
- Population Health Program, Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
11
|
Glebov OO. Understanding SARS-CoV-2 endocytosis for COVID-19 drug repurposing. FEBS J 2020; 287:3664-3671. [PMID: 32428379 PMCID: PMC7276759 DOI: 10.1111/febs.15369] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
The quest for the effective treatment against coronavirus disease 2019 pneumonia caused by the severe acute respiratory syndrome (SARS)‐coronavirus 2(CoV‐2) coronavirus is hampered by the lack of knowledge concerning the basic cell biology of the infection. Given that most viruses use endocytosis to enter the host cell, mechanistic investigation of SARS‐CoV‐2 infection needs to consider the diversity of endocytic pathways available for SARS‐CoV‐2 entry in the human lung epithelium. Taking advantage of the well‐established methodology of membrane trafficking studies, this research direction allows for the rapid characterisation of the key cell biological mechanism(s) responsible for SARS‐CoV‐2 infection. Furthermore, 11 clinically approved generic drugs are identified as potential candidates for repurposing as blockers of several potential routes for SARS‐CoV‐2 endocytosis. More broadly, the paradigm of targeting a fundamental aspect of human cell biology to protect against infection may be advantageous in the context of future pandemic outbreaks.
Collapse
Affiliation(s)
- Oleg O Glebov
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China.,Department of Old Age Psychiatry, The Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England, UK
| |
Collapse
|
12
|
Schwarz DGG, Pena JL, Carvalho IA, Silva Júnior A, Moreira MAS. Inhibition of Escherichia coli invasion into bovine mammary epithelial cells previously infected by Mycobacterium avium subsp. paratuberculosis. Vet Q 2020; 40:43-50. [PMID: 31939335 PMCID: PMC7734034 DOI: 10.1080/01652176.2020.1716278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background The coinfection process of Escherichia coli, an etiological agent of clinical mastitis and Mycobacterium avium subsp. paratuberculosis (MAP), a non-mastitic etiological agent in the bovine mammary gland is not fully known. Objective Verify the ability of MAP to interfere with the invasion and translocation of E. coli in bovine mammary epithelial cell line (MAC-T). Methods For the invasion assay, MAC-T cells were challenged with MAP K10 for 2 h and then challenged with E. coli for 10, 30 and 120 min. For the translocation assay, the trans well plates were used and the challenge sequence was repeated as previously described. The amount of E. coli in the assays was determined by counting colony forming units (CFU) in Luria-Bertani medium. Quantitative real-time PCR was used to quantify MAP in MAC-T cells. To verify the viability of the MAC-T cells, the MTT assay was performed. MAP culture supernatant was also evaluated at different percentages for E. coli growth. Results Previous MAP infection in MAC-T cells inhibited E. coli invasion in 10, 30 and 120 min. No significant interference of MAP in the translocation of E. coli from the apical-basal direction was verified. Quantity of MAP DNA inside the MAC-T cells was statistically similar. Neither reduction in MAC-T cells viability was detected during the experiment nor MAP-released factor in the supernatant inhibited E. coli invasion. Conclusion These findings suggest that MAP-positive cows could be more resistant to E. coli infection, but when infected, could rapidly translocate E. coli to the subepithelial region.
Collapse
Affiliation(s)
- David Germano G Schwarz
- Veterinary Medicine, Universidade Federal do Piauí (UFPI), Campus Cinobelina Elvas (CPCE), Bom Jesus, PI, Brazil
| | - Junnia L Pena
- Departament of Veterinary, Sector of Veterinary Medicine and Public Health, Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | - Isabel A Carvalho
- Department of Pathology, Universidade Estadual do Maranhão, São Luís, MA, Brazil
| | - Abelardo Silva Júnior
- Departament of Veterinary, Sector of Veterinary Medicine and Public Health, Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | - Maria Aparecida S Moreira
- Departament of Veterinary, Sector of Veterinary Medicine and Public Health, Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| |
Collapse
|
13
|
Bao Y, Zhao X, Wang L, Qian W, Sun J. Morphology-based classification of mycobacteria-infected macrophages with convolutional neural network: reveal EsxA-induced morphologic changes indistinguishable by naked eyes. Transl Res 2019; 212:1-13. [PMID: 31287998 PMCID: PMC6755059 DOI: 10.1016/j.trsl.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/17/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
EsxA is an essential virulence factor for Mycobacterium tuberculosis (Mtb) pathogenesis as well as an important biomarker for Mtb detection. In this study, we use light microscopy and deep learning-based image analysis to classify the morphologic changes of macrophages infected by Mycobacterium marinum (Mm), a surrogate model for Mtb. Macrophages were infected either with the mCherry-expressing Mm wild type strain (Mm(WT)), or a mutant strain with deletion of the esxA-esxB operon (Mm(ΔEsxA:B)). The mCherry serves as an infection marker to train the convolution neural network (CNN) and to validate the classification results. Data show that CNN can distinguish the Mm(WT)-infected cells from uninfected cells with an accuracy of 92.4% at 2 hours postinfection (hpi). However, the accuracy at 12 and 24 hpi is decreased to ∼75% and ∼83%, respectively, suggesting dynamic morphologic changes through different stages of infection. The accuracy of discriminating Mm(ΔEsxA:B)-infected cells from uninfected cells is lower than 80% at all time, which is consistent to attenuated virulence of Mm(ΔEsxA:B). Interestingly, CNN distinguishes Mm(WT)-infected cells from Mm(ΔEsxA:B)-infected cells with ∼90% accuracy, implicating EsxA induces unique morphologic changes in macrophages. Deconvolutional analysis successfully reconstructed the morphologic features used by CNN for classification, which are indistinguishable to naked eyes and distinct from intracellular mycobacteria. This study presents a deep learning-aided imaging analytical tool that can accurately detect virulent mycobacteria-infected macrophages by cellular morphologic changes. The observed morphologic changes induced by EsxA warrant further studies to fill the gap from molecular actions of bacterial virulence factors to cellular morphology.
Collapse
Affiliation(s)
- Yanqing Bao
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Xinzhuo Zhao
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas; Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Lin Wang
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Wei Qian
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas
| | - Jianjun Sun
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| |
Collapse
|
14
|
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: 49] [Impact Index Per Article: 8.2] [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
|
15
|
Hadifar S, Behrouzi A, Fateh A, Khatami S, Rahimi Jamnani F, Siadat SD, Vaziri F. Comparative study of interruption of signaling pathways in lung epithelial cell by two different Mycobacterium tuberculosis lineages. J Cell Physiol 2019; 234:4739-4753. [PMID: 30192006 DOI: 10.1002/jcp.27271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022]
Abstract
Alveolar epithelial cell (AEC) provides a replication niche for Mycobacterium tuberculosis. Based on the role of AEC in M. tuberculosis pathogenesis and existence of genetic diversity within this bacterium, we investigated interactions between AEC II and two different M. tuberculosis lineages. We have compared the transcriptome and cytokines/chemokines levels of A549 infected by M. tuberculosis lineage three and four using qRT-PCR and ELISA arrays, respectively. We showed different M. tuberculosis strains induced changes in different effectors that involved in TLRs and NF-κB signaling pathways. We observed different reaction of the studied lineages specifically in pathogenesis, immune evasion mechanism, IL-12/IFN-γ axis, and autophagy. Similar behavior was detected in regarding to apoptosis, necroptosis, anti-inflammatory responses, and canonical inflammasome. Our findings contribute to elucidate more details in pathogenesis, immune evasion strategies, novel target and druggable pathway for therapeutic intervention, and host directed therapy in tuberculosis infection. Also, different M. tuberculosis lineages-dependent host-pathogen interactions suggested using only one strain for this kind of research will be controversial.
Collapse
Affiliation(s)
- Shima Hadifar
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rahimi Jamnani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
16
|
Bertuzzi M, Hayes GE, Bignell EM. Microbial uptake by the respiratory epithelium: outcomes for host and pathogen. FEMS Microbiol Rev 2019; 43:145-161. [PMID: 30657899 PMCID: PMC6435450 DOI: 10.1093/femsre/fuy045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Intracellular occupancy of the respiratory epithelium is a useful pathogenic strategy facilitating microbial replication and evasion of professional phagocytes or circulating antimicrobial drugs. A less appreciated but growing body of evidence indicates that the airway epithelium also plays a crucial role in host defence against inhaled pathogens, by promoting ingestion and quelling of microorganisms, processes that become subverted to favour pathogen activities and promote respiratory disease. To achieve a deeper understanding of beneficial and deleterious activities of respiratory epithelia during antimicrobial defence, we have comprehensively surveyed all current knowledge on airway epithelial uptake of bacterial and fungal pathogens. We find that microbial uptake by airway epithelial cells (AECs) is a common feature of respiratory host-microbe interactions whose stepwise execution, and impacts upon the host, vary by pathogen. Amidst the diversity of underlying mechanisms and disease outcomes, we identify four key infection scenarios and use best-characterised host-pathogen interactions as prototypical examples of each. The emergent view is one in which effi-ciency of AEC-mediated pathogen clearance correlates directly with severity of disease outcome, therefore highlighting an important unmet need to broaden our understanding of the antimicrobial properties of respiratory epithelia and associated drivers of pathogen entry and intracellular fate.
Collapse
Affiliation(s)
- Margherita Bertuzzi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
- Lydia Becker Institute of Immunology and Inflammation, Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre
| | - Gemma E Hayes
- Northern Devon Healthcare NHS Trust, North Devon District Hospital, Raleigh Park, Barnstaple EX31 4JB, UK
| | - Elaine M Bignell
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
- Lydia Becker Institute of Immunology and Inflammation, Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre
| |
Collapse
|
17
|
Edwardsiella piscicida Enters Nonphagocytic Cells via a Macropinocytosis-Involved Hybrid Mechanism. J Bacteriol 2019; 201:JB.00548-18. [PMID: 30530518 DOI: 10.1128/jb.00548-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
Edwardsiella piscicida is an important pathogen that infects a wide range of hosts from fish to human. Recent studies demonstrated that E. piscicida can invade and survive within multiple nonphagocytic cells, but the internalization mechanism remains poorly understood. Here, we used HeLa cells as a nonphagocytic cell model to investigate the endocytic strategy used by the pathogenic E. piscicida isolate EIB202. Using a combination of optical and electron microscopy, we observed obvious membrane ruffles and F-actin rearrangements in HeLa cells after EIB202 infection. We also revealed that EIB202 internalization significantly depended on the activity of Na+/H+ exchangers and multiple intracellular signaling events related to macropinocytosis, suggesting that E. piscicida utilizes the host macropinocytosis pathway to enter HeLa cells. Further, using inhibitory drugs and shRNAs to block specific endocytic pathways, we found that a caveolin-dependent but not clathrin-dependent pathway is involved in E. piscicida entry and that its entry requires dynamin and membrane cholesterol. Together, these data suggest that E. piscicida enters nonphagocytic cells via macropinocytosis and caveolin-dependent endocytosis involving cholesterol and dynamin, improving the understanding of how E. piscicida interacts with nonphagocytic cells.IMPORTANCE Bacterial internalization is the first step in breaking through the host cell defense. Therefore, studying the mechanism of bacterial internalization improves the understanding of the pathogenic mechanism of bacteria. In this study, the internalization process on nonphagocytic cells by Edwardsiella piscicida was evaluated. Our results showed that E. piscicida can be internalized into nonphagocytic cells via macropinocytosis and caveolin-mediated endocytosis, and that cholesterol and dynamin are involved in this process. These results reveal a new method for inhibiting E. piscicida infection, providing a foundation for further studies of bacterial pathogenicity.
Collapse
|
18
|
Abhishek S, Saikia UN, Gupta A, Bansal R, Gupta V, Singh N, Laal S, Verma I. Transcriptional Profile of Mycobacterium tuberculosis in an in vitro Model of Intraocular Tuberculosis. Front Cell Infect Microbiol 2018; 8:330. [PMID: 30333960 PMCID: PMC6175983 DOI: 10.3389/fcimb.2018.00330] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/28/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Intraocular tuberculosis (IOTB), an extrapulmonary manifestation of tuberculosis of the eye, has unique and varied clinical presentations with poorly understood pathogenesis. As it is a significant cause of inflammation and visual morbidity, particularly in TB endemic countries, it is essential to study the pathogenesis of IOTB. Clinical and histopathologic studies suggest the presence of Mycobacterium tuberculosis in retinal pigment epithelium (RPE) cells. Methods: A human retinal pigment epithelium (ARPE-19) cell line was infected with a virulent strain of M. tuberculosis (H37Rv). Electron microscopy and colony forming units (CFU) assay were performed to monitor the M. tuberculosis adherence, invasion, and intracellular replication, whereas confocal microscopy was done to study its intracellular fate in the RPE cells. To understand the pathogenesis, the transcriptional profile of M. tuberculosis in ARPE-19 cells was studied by whole genome microarray. Three upregulated M. tuberculosis transcripts were also examined in human IOTB vitreous samples. Results: Scanning electron micrographs of the infected ARPE-19 cells indicated adherence of bacilli, which were further observed to be internalized as monitored by transmission electron microscopy. The CFU assay showed that 22.7 and 8.4% of the initial inoculum of bacilli adhered and invaded the ARPE-19 cells, respectively, with an increase in fold CFU from 1 dpi (0.84) to 5dpi (6.58). The intracellular bacilli were co-localized with lysosomal-associated membrane protein-1 (LAMP-1) and LAMP-2 in ARPE-19 cells. The transcriptome study of intracellular bacilli showed that most of the upregulated transcripts correspond to the genes encoding the proteins involved in the processes such as adherence (e.g., Rv1759c and Rv1026), invasion (e.g., Rv1971 and Rv0169), virulence (e.g., Rv2844 and Rv0775), and intracellular survival (e.g., Rv1884c and Rv2450c) as well as regulators of various metabolic pathways. Two of the upregulated transcripts (Rv1971, Rv1230c) were also present in the vitreous samples of the IOTB patients. Conclusions:M. tuberculosis is phagocytosed by RPE cells and utilizes these cells for intracellular multiplication with the involvement of late endosomal/lysosomal compartments and alters its transcriptional profile plausibly for its intracellular adaptation and survival. The findings of the present study could be important to understanding the molecular pathogenesis of IOTB with a potential role in the development of diagnostics and therapeutics for IOTB.
Collapse
Affiliation(s)
- Sudhanshu Abhishek
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Uma Nahar Saikia
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amod Gupta
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reema Bansal
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishali Gupta
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nirbhai Singh
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Suman Laal
- Department of Pathology, New York University Langone Medical Center, New York, NY, United States
- Veterans Affairs New York Harbor Healthcare System, New York, NY, United States
| | - Indu Verma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
19
|
Abstract
While cancer cell proliferation depends on access to extracellular nutrients, inadequate tumour perfusion means that glucose, amino acids and lipids are often in short supply. To overcome this obstacle to growth, cancer cells utilize multiple scavenging strategies, obtaining macromolecules from the microenvironment and breaking them down in the lysosome to produce substrates for ATP generation and anabolism. Recent studies have revealed four scavenging pathways that support cancer cell proliferation in low-nutrient environments: scavenging of extracellular matrix proteins via integrins, receptor-mediated albumin uptake and catabolism, macropinocytic consumption of multiple components of the tumour microenvironment and the engulfment and degradation of entire live cells via entosis. New evidence suggests that blocking these pathways alone or in combination could provide substantial benefits to patients with incurable solid tumours. Both US Food and Drug Administration (FDA)-approved drugs and several agents in preclinical or clinical development shut down individual or multiple scavenging pathways. These therapies may increase the extent and durability of tumour growth inhibition and/or prevent the development of resistance when used in combination with existing treatments. This Review summarizes the evidence suggesting that scavenging pathways drive tumour growth, highlights recent advances that define the oncogenic signal transduction pathways that regulate scavenging and considers the benefits and detriments of therapeutic strategies targeting scavenging that are currently under development.
Collapse
Affiliation(s)
- Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Vaishali Jayashankar
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
20
|
Kim S, Seo H, Mahmud HA, Islam MI, Lee BE, Cho ML, Song HY. In vitro activity of collinin isolated from the leaves of Zanthoxylum schinifolium against multidrug- and extensively drug-resistant Mycobacterium tuberculosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 46:104-110. [PMID: 30097109 DOI: 10.1016/j.phymed.2018.04.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/14/2018] [Accepted: 04/15/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Tuberculosis is a very serious infectious disease that threatens humanity, and the emergence of multidrug-resistant (MDR), extensively drug-resistant (XDR) strains resistant to drugs suggests that new drug development is urgent. In order to develop new tuberculosis drug, we have conducted in vitro anti-tubercular tests on thousands of plant-derived substances and finally found collinin extracted from the leaves of Zanthoxylum schinifolium, which has an excellent anti-tuberculosis effect. PURPOSE To isolate an anti-tubercular bioactive compound from the leaves of Z. schinifolium and evaluate whether this agent demonstrates any potential in vitro characteristics suitable for the development of future anti-tubercular drugs to treat MDR and XDR Mycobacterium tuberculosis. METHODS The methanolic extracts of the leaves of Z. schinifolium were subjected to bioassay-guided fractionation against M. tuberculosis using a microbial cell viability assay. In addition, following cell cytotoxicity assay, an intracellular anti-mycobacterial activity of the most active anti-tubercular compound was investigated after it was purified. RESULTS The active compound with anti-tubercular activity isolated from leaves of Z. schinifolium was identified as a collinin. The extracted collinin showed anti-tubercular activity against both drug-susceptible and -resistant strains of M. tuberculosis at 50% minimum inhibitory concentrations (MIC50s) of 3.13-6.25 µg/ml in culture broth and MIC50s of 6.25-12.50 µg/ml inside Raw264.7 and A549 cells. Collinin had no cytotoxicity against human lung pneumocytes up to a concentration of 100 µg/ml (selectivity index > 16-32). CONCLUSIONS Collinin extracted from the leaves of Z. schinifolium significantly inhibits the growth of MDR and XDR M. tuberculosis in the culture broth. In addition, it also inhibits the growth of intracellular drug-susceptible and drug-resistant tuberculosis in Raw264.7 and A549 cells. To our knowledge, this is the first report on the in vitro anti-tubercular activity of collinin, and our data suggest collinin as a potential drug to treat drug-resistant tuberculosis. Further studies are warranted to assess the in vivo efficacy and therapeutic potential of collinin.
Collapse
Affiliation(s)
- Sukyung Kim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, South Korea
| | - Hoonhee Seo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, South Korea
| | - Hafij Al Mahmud
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, South Korea
| | - Md Imtiazul Islam
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, South Korea
| | - Byung-Eui Lee
- Department of Chemistry, School of Life Sciences, Soonchunhyang University, Asan, Chungnam 31538, South Korea
| | - Myoung-Lae Cho
- National Development Institute of Korean Medicine, Gyeongsan, Gyeongnam 38540, South Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, South Korea.
| |
Collapse
|
21
|
Chen Z, Shao XY, Wang C, Hua MH, Wang CN, Wang X, Wang QJ, Yao JY, Fan YH, Qin YW. Mycobacterium marinum Infection in Zebrafish and Microglia Imitates the Early Stage of Tuberculous Meningitis. J Mol Neurosci 2018; 64:321-330. [PMID: 29352446 DOI: 10.1007/s12031-018-1026-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/03/2018] [Indexed: 12/27/2022]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) invading and activating microglia causes the most serious subtypes of tuberculosis called tubercular meningitis. However, the developmental process of tubercular meningitis, especially the early phase, is poorly understood due to lacking well-established and well-accepted visible models in vitro and in vivo. Here, consistent with one recent report, we found Mycobacterium marinum (M. marinum) invade the zebrafish brain and subsequently cause granuloma-like structures. We further showed that M. marinum, which shares similar characteristics with M. tuberculosis, can invade microglia and replicate in microglia, which subsequently promote the secretion of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α. M. marinum infection in microglia can also promote autophagy, which conversely limits the replication of M. marinum. Thus, pharmacological activation of autophagy by rapamycin could prevent M. marinum replication. Our study provides in vivo and in vitro models to study underlying pathogenic mechanisms of tubercular meningitis by using M. marinum. Our results also showed that activation of autophagy could be a meaningful way to prevent tubercular meningitis.
Collapse
Affiliation(s)
- Zhan Chen
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, China
| | - Xiao-Yi Shao
- Department of Immunology, Medical College, Nantong University, Nantong, China
| | - Chao Wang
- Department of Immunology, Medical College, Nantong University, Nantong, China
| | - Min-Hui Hua
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cheng-Niu Wang
- Basic Medical Research Center, Medical College, Nantong University, Nantong, China
| | - Xin Wang
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Qian-Jin Wang
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, China
| | - Jin-Yi Yao
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, China
| | - Yi-Hui Fan
- Department of Immunology, Medical College, Nantong University, Nantong, China.
| | - Yong-Wei Qin
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, China.
| |
Collapse
|
22
|
Song OR, Queval CJ, Iantomasi R, Delorme V, Marion S, Veyron-Churlet R, Werkmeister E, Popoff M, Ricard I, Jouny S, Deboosere N, Lafont F, Baulard A, Yeramian E, Marsollier L, Hoffmann E, Brodin P. ArfGAP1 restricts Mycobacterium tuberculosis entry by controlling the actin cytoskeleton. EMBO Rep 2017; 19:29-42. [PMID: 29141986 DOI: 10.15252/embr.201744371] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/03/2017] [Accepted: 10/23/2017] [Indexed: 11/09/2022] Open
Abstract
The interaction of Mycobacterium tuberculosis (Mtb) with pulmonary epithelial cells is critical for early stages of bacillus colonization and during the progression of tuberculosis. Entry of Mtb into epithelial cells has been shown to depend on F-actin polymerization, though the molecular mechanisms are still unclear. Here, we demonstrate that mycobacterial uptake into epithelial cells requires rearrangements of the actin cytoskeleton, which are regulated by ADP-ribosylation factor 1 (Arf1) and phospholipase D1 (PLD1), and is dependent on the M3 muscarinic receptor (M3R). We show that this pathway is controlled by Arf GTPase-activating protein 1 (ArfGAP1), as its silencing has an impact on actin cytoskeleton reorganization leading to uncontrolled uptake and replication of Mtb. Furthermore, we provide evidence that this pathway is critical for mycobacterial entry, while the cellular infection with other pathogens, such as Shigella flexneri and Yersinia pseudotuberculosis, is not affected. Altogether, these results reveal how cortical actin plays the role of a barrier to prevent mycobacterial entry into epithelial cells and indicate a novel role for ArfGAP1 as a restriction factor of host-pathogen interactions.
Collapse
Affiliation(s)
- Ok-Ryul Song
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,Equipe ATIP AVENIR, CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| | - Christophe J Queval
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Raffaella Iantomasi
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Vincent Delorme
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| | - Sabrina Marion
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Romain Veyron-Churlet
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Elisabeth Werkmeister
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Michka Popoff
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France.,CNRS, UMR8520, Institut d'électronique, de microélectronique et de nanotechnologie, Villeneuve d'Ascq, France
| | - Isabelle Ricard
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Samuel Jouny
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Nathalie Deboosere
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Frank Lafont
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Alain Baulard
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Edouard Yeramian
- Unité de Microbiologie Structurale, CNRS UMR3528, Institut Pasteur, Paris, France
| | - Laurent Marsollier
- Equipe ATIP AVENIR, CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Eik Hoffmann
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | - Priscille Brodin
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, Univ. Lille, Lille, France .,Institute Pasteur Korea, Seongnam-si Gyeonggi-do, South Korea
| |
Collapse
|
23
|
Ribeiro GM, Matsumoto CK, Real F, Teixeira D, Duarte RS, Mortara RA, Leão SC, de Souza Carvalho-Wodarz C. Increased survival and proliferation of the epidemic strain Mycobacterium abscessus subsp. massiliense CRM0019 in alveolar epithelial cells. BMC Microbiol 2017; 17:195. [PMID: 28903728 PMCID: PMC5598063 DOI: 10.1186/s12866-017-1102-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Outbreaks of infections caused by rapidly growing mycobacteria have been reported worldwide generally associated with medical procedures. Mycobacterium abscessus subsp. massiliense CRM0019 was obtained during an epidemic of postsurgical infections and was characterized by increased persistence in vivo. To better understand the successful survival strategies of this microorganism, we evaluated its infectivity and proliferation in macrophages (RAW and BMDM) and alveolar epithelial cells (A549). For that, we assessed the following parameters, for both M. abscessus CRM0019 as well as the reference strain M. abscessus ATCC 19977: internalization, intracellular survival for up 3 days, competence to subvert lysosome fusion and the intracellular survival after cell reinfection. RESULTS CRM0019 and ATCC 19977 strains showed the same internalization rate (approximately 30% after 6 h infection), in both A549 and RAW cells. However, colony forming units data showed that CRM0019 survived better in A549 cells than the ATCC 19977 strain. Phagosomal characteristics of CRM0019 showed the bacteria inside tight phagosomes in A549 cells, contrasting to the loosely phagosomal membrane in macrophages. This observation holds for the ATCC 19977 strain in both cell types. The competence to subvert lysosome fusion was assessed by acidification and acquisition of lysosomal protein. For M. abscessus strains the phagosomes were acidified in all cell lines; nevertheless, the acquisition of lysosomal protein was reduced by CRM0019 compared to the ATCC 19977 strain, in A549 cells. Conversely, in macrophages, both M. abscessus strains were located in mature phagosomes, however without bacterial death. Once recovered from macrophages M. abscessus could establish a new intracellular infection. Nevertheless, only CRM0019 showed a higher growth rate in A549, increasing nearly 10-fold after 48 and 72 h. CONCLUSION M. abscessus CRM0019 creates a protective and replicative niche in alveolar epithelial cells mainly by avoiding phagosome maturation. Once recovered from infected macrophages, CRM0019 remains infective and displays greater intracellular growth in A549 cells compared to the ATCC 19977 strain. This evasion strategy in alveolar epithelial cells may contribute to the long survival of the CRM0019 strain in the host and thus to the inefficacy of in vivo treatment.
Collapse
Affiliation(s)
- Giovanni Monteiro Ribeiro
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Cristianne Kayoko Matsumoto
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fernando Real
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Laboratoire Entrée muqueuse du VIH et Immunité muqueuse, Department Infection, Immunité et Inflammation, Institut Cochin, Paris, France
| | - Daniela Teixeira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rafael Silva Duarte
- Laboratório de Micobactérias, Instituto de Microbiologia Professor Paulo de Góes, Cidade Universitária, Rio de Janeiro, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Sylvia Cardoso Leão
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Cristiane de Souza Carvalho-Wodarz
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil. .,Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany.
| |
Collapse
|
24
|
Carranza-Rosales P, Carranza-Torres IE, Guzmán-Delgado NE, Lozano-Garza G, Villarreal-Treviño L, Molina-Torres C, Villarreal JV, Vera-Cabrera L, Castro-Garza J. Modeling tuberculosis pathogenesis through ex vivo lung tissue infection. Tuberculosis (Edinb) 2017; 107:126-132. [PMID: 29050759 PMCID: PMC7106348 DOI: 10.1016/j.tube.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/06/2017] [Accepted: 09/10/2017] [Indexed: 02/02/2023]
Abstract
Tuberculosis (TB) is one of the top 10 causes of death worldwide. Several in vitro and in vivo experimental models have been used to study TB pathogenesis and induction of immune response during Mycobacterium tuberculosis infection. Precision cut lung tissue slices (PCLTS) is an experimental model, in which all the usual cell types of the organ are found, the tissue architecture and the interactions amongst the different cells are maintained. PCLTS in good physiological conditions, monitored by MTT assay and histology, were infected with either virulent Mycobacterium tuberculosis strain H37Rv or the TB vaccine strain Mycobacterium bovis BCG. Histological analysis showed that bacilli infecting lung tissue slices were observed in the alveolar septa, alveolar light spaces, near to type II pneumocytes, and inside macrophages. Mycobacterial infection of PCLTS induced TNF-α production, which is consistent with previous M. tuberculosis in vitro and in vivo studies. This is the first report of using PCLTS as a system to study M. tuberculosis infection. The PCLTS model provides a useful tool to evaluate the innate immune responses and other aspects during the early stages of mycobacterial infection.
Collapse
Affiliation(s)
- Pilar Carranza-Rosales
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Irma Edith Carranza-Torres
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico; Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Avenida Pedro de Alba y Manuel L, Barragán s/n, Cd. Universitaria, 66450, San Nicolás de los Garza, N.L., Mexico.
| | - Nancy Elena Guzmán-Delgado
- Departamento de Patología, Unidad Médica de Alta Especialidad # 34, Instituto Mexicano del Seguro Social, Monterrey, N.L. 64730, Mexico.
| | - Gerardo Lozano-Garza
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Licet Villarreal-Treviño
- Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Avenida Pedro de Alba y Manuel L, Barragán s/n, Cd. Universitaria, 66450, San Nicolás de los Garza, N.L., Mexico.
| | - Carmen Molina-Torres
- Servicio de Dermatología, Hospital Universitario "José E. González", Universidad Autónoma de Nuevo León, Madero y Gonzalitos, Col. Mitras Centro, Monterrey, N.L., Mexico.
| | - Javier Vargas Villarreal
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Lucio Vera-Cabrera
- Servicio de Dermatología, Hospital Universitario "José E. González", Universidad Autónoma de Nuevo León, Madero y Gonzalitos, Col. Mitras Centro, Monterrey, N.L., Mexico.
| | - Jorge Castro-Garza
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| |
Collapse
|
25
|
Mahmud HA, Seo H, Kim S, Islam MI, Nam KW, Cho HD, Song HY. Thymoquinone (TQ) inhibits the replication of intracellular Mycobacterium tuberculosis in macrophages and modulates nitric oxide production. Altern Ther Health Med 2017; 17:279. [PMID: 28545436 PMCID: PMC5445392 DOI: 10.1186/s12906-017-1786-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/10/2017] [Indexed: 11/23/2022]
Abstract
Background Human tuberculosis, which is caused by the pathogen Mycobacterium tuberculosis, remains a major public health concern. Increasing drug resistance poses a threat of disease resurgence and continues to cause considerable mortality worldwide, which necessitates the development of new drugs with improved efficacy. Thymoquinone (TQ), an essential compound of Nigella sativa, was previously reported as an active anti-tuberculosis agent. Methods In this study, the effects of TQ on intracellular mycobacterial replication are examined in macrophages. In addition, its effect on mycobacteria-induced NO production and pro-inflammatory responses were investigated in Mycobacterium tuberculosis (MTB)-infected Type II human alveolar and human myeloid cell lines. Results TQ at concentrations ranging from 12.5 to 25 μg/mL and 6.25 to 12.5 μg/mL reduced intracellular M. tuberculosis H37Rv and extensively drug-resistant tuberculosis (XDR-TB) 72 h post-infection in RAW 264.7 cells. TQ treatment also produced a concentration-dependent reduction in nitric oxide production in both H37Rv and XDR-TB infected RAW 264.7 cells. Furthermore, TQ reduced the expression of inducible nitric oxide synthase (iNOS) and pro-inflammatory molecules such as tumor necrosis factor-alpha (TNF-α) and interlukin-6 (IL-6) in H37Rv-infected cells and eventually reduced pathogen-derived stress in host cells. Conclusions TQ inhibits intracellular H37Rv and XDR-TB replication and MTB-induced production of NO and pro-inflammatory molecules. Therefore, along with its anti-inflammatory effects, TQ represents a prospective treatment option to combat Mycobacterium tuberculosis infection.
Collapse
|
26
|
Kühn S, Lopez-Montero N, Chang YY, Sartori-Rupp A, Enninga J. Imaging macropinosomes during Shigella infections. Methods 2017; 127:12-22. [PMID: 28522322 DOI: 10.1016/j.ymeth.2017.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022] Open
Abstract
Macropinocytosis is the uptake of extracellular fluid within vesicles of varying size that takes place during numerous cellular processes in a large variety of cells. A growing number of pathogens, including viruses, parasites, and bacteria are known to induce macropinocytosis during their entry into targeted host cells. We have recently discovered that the human enteroinvasive, bacterial pathogen Shigella causes in situ macropinosome formation during its entry into epithelial cells. These infection-associated macropinosomes are not generated to ingest the bacteria, but are instead involved in Shigella's intracellular niche formation. They make contacts with the phagocytosed shigellae to promote vacuolar membrane rupture and their cytosolic release. Here, we provide an overview of the different imaging approaches that are currently used to analyze macropinocytosis during infectious processes with a focus on Shigella entry. We detail the advantages and disadvantages of genetically encoded reporters as well as chemical probes to trace fluid phase uptake. In addition, we report how such reporters can be combined with ultrastructural approaches for correlative light electron microscopy either in thin sections or within large volumes. The combined imaging techniques introduced here provide a detailed characterization of macropinosomes during bacterial entry, which, apart from Shigella, are relevant for numerous other ones, including Salmonella, Brucella or Mycobacteria.
Collapse
Affiliation(s)
- Sonja Kühn
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | | | - Yuen-Yan Chang
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Anna Sartori-Rupp
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Jost Enninga
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France.
| |
Collapse
|
27
|
Mortaz E, Alipoor SD, Movassaghi M, Varahram M, Ghorbani J, Folkerts G, Garssen J, Adcock IM. Water-pipe smoke condensate increases the internalization of Mycobacterium Bovis of type II alveolar epithelial cells (A549). BMC Pulm Med 2017; 17:68. [PMID: 28431548 PMCID: PMC5401461 DOI: 10.1186/s12890-017-0413-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/13/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) is a major global health problem, and there is an association between tobacco smoke and TB. Water pipe smoking has become an increasing problem not only in Middle Eastern countries but also globally because users consider it as safer than cigarettes. The presence of high levels of toxic substances in water-pipe smoke may be a predisposing factor that enhances the incidence of pulmonary disorders. For example, uncontrolled macropinocytosis in alveolar epithelial cells following exposure to water-pipe smoke may predispose subjects to pulmonary infection. Here, we studied the effects of water-pipe condense (WPC) on the internalization of Mycobacterium Bovis BCG by macropinocytosis in the alveolar epithelial cell line A549. METHODS A549 cells were exposed to WPC (4 mg/ml) for 24, 48, 72 and 96 h. Cell viability was studied using the methyl thiazolyldipenyl-tetrazolium bromide (MTT) reduction assay and proliferation by bromodeoxyUridine (BrdU) incorporation. Cells were exposed to FITC-Dextran (1 mg/ml) (as a control) and FITC-BCG (MOI = 10) for 20 min at 37 °C before cells were collected and the uptake of BCG-FITC determined by flow cytometry. Similar experiments were performed at 4 °C as a control. The Rho-associated protein kinase (ROCK) inhibitor Y-27632 (1 μM) was used to assess the mechanism by which WPC enhanced BCG uptake. RESULTS WPC (4 mg/ml) increased the uptake of BCG-FITC after 72 (1.3 ± 0.1 fold, p < 0.05) and 96 (1.4 ± 0.05 fold, p < 0.05) hours. No effect on BCG-FITC uptake was observed at 24 or 48 h. WPC also significantly increased the uptake of FITC-Dextran (2.9 ± 0.3 fold, p < 0.05) after 24 h. WPC significantly decreased cell viability after 24 (84 ± 2%, p < 0.05), 48 (78±, 3%, p < 0.05), 72 (64 ± 2%, p < 0.05) and 96 h (45 ± 2%, p < 0.05). Y-27632 completely attenuated the increased uptake of BCG by WPC. Cell proliferation showed a decreasing trend in a time-dependent manner with WPC exposure. CONCLUSION WPC exposure increased epithelial cell endocytosis activity and death as well as enhancing their capacity for macropinocytosis. Our in vitro data indicates possible harmful effects of WPC on the ability of lung epithelial cells to phagocytose mycobacterium.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shamila D Alipoor
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Medical Biotechnology, Molecular Medicine Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Movassaghi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Mohammad Varahram
- Mycobacteriology Research Center (MRC) National Research Institute of Tuberculosis and lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jahangir Ghorbani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Nutricia Research Centre for Specialized Nutrition, Utrecht, The Netherlands
| | - Ian M Adcock
- Cell and Molecular Biology Group, Airways Disease Section, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, UK
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
28
|
Alaridah N, Lutay N, Tenland E, Rönnholm A, Hallgren O, Puthia M, Westergren-Thorsson G, Godaly G. Mycobacteria Manipulate G-Protein-Coupled Receptors to Increase Mucosal Rac1 Expression in the Lungs. J Innate Immun 2016; 9:318-329. [PMID: 28013312 DOI: 10.1159/000453454] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/15/2016] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium bovis bacille Calmette-Guérin (BCG) is currently the only approved vaccine against tuberculosis (TB). BCG mimics M. tuberculosis (Mtb) in its persistence in the body and is used as a benchmark to compare new vaccine candidates. BCG was originally designed for mucosal vaccination, but comprehensive knowledge about its interaction with epithelium is currently lacking. We used primary airway epithelial cells (AECs) and a murine model to investigate the initial events of mucosal BCG interactions. Furthermore, we analysed the impact of the G-protein-coupled receptors (GPCRs), CXCR1 and CXCR2, in this process, as these receptors were previously shown to be important during TB infection. BCG infection of AECs induced GPCR-dependent Rac1 up-regulation, resulting in actin redistribution. The altered distribution of the actin cytoskeleton involved the MAPK signalling pathway. Blocking of the CXCR1 or CXCR2 prior to infection decreased Rac1 expression, and increased epithelial transcriptional activity and epithelial cytokine production. BCG infection did not result in epithelial cell death as measured by p53 phosphorylation and annexin. This study demonstrated that BCG infection of AECs manipulated the GPCRs to suppress epithelial signalling pathways. Future vaccine strategies could thus be improved by targeting GPCRs.
Collapse
Affiliation(s)
- Nader Alaridah
- Division of Laboratory Medicine, Department of MIG, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Shree S, Singh AK, Saxena R, Kumar H, Agarwal A, Sharma VK, Srivastava K, Srivastava KK, Sanyal S, Ramachandran R. The M. tuberculosis HAD phosphatase (Rv3042c) interacts with host proteins and is inhibited by Clofazimine. Cell Mol Life Sci 2016; 73:3401-17. [PMID: 26984196 PMCID: PMC11108430 DOI: 10.1007/s00018-016-2177-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/18/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023]
Abstract
Mycobacterium tuberculosis codes for a HAD-phosphatase, Rv3042c (MtSerB2), that has earlier been characterized as a metabolic enzyme. Here we demonstrate that MtSerB2 is secreted into the cytosol of infected macrophages and is found in bronchoalveolar lavage samples of tuberculosis patients. MtSerB2 induces significant cytoskeleton rearrangements through cofilin activation and affects the expression of genes that regulate actin dynamics. It specifically interacts with HSP90, HSP70 and HSP27 that block apoptotic pathways and not with other HSPs. It actively dephosphorylates MAPK-p38 and NF-kappa B p65 that play crucial roles in inflammatory and immune responses. This in turn leads to down-regulation of Interleukin 8, a chemotactic and inflammatory cytokine. Finally, during evaluation of inhibitors against MtSerB2 we found that Clofazimine, a drug being evaluated for XDR and MDR tuberculosis, inhibits MtSerB2 phosphatase activity and reverses the above effects and interactions with host proteins. Overall, the study identifies that MtSerB2 has new functions that might help the pathogen to evade the host's immune response.
Collapse
Affiliation(s)
- Sonal Shree
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Abhishek Kumar Singh
- Biochemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Richa Saxena
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Harish Kumar
- Biochemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Aparna Agarwal
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Vijay Kumar Sharma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Kanchan Srivastava
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Kishore Kumar Srivastava
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Sabyasachi Sanyal
- Biochemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Ravishankar Ramachandran
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
30
|
Zhu Q, Zhang M, Shi M, Liu Y, Zhao Q, Wang W, Zhang G, Yang L, Zhi J, Zhang L, Hu G, Chen P, Yang Y, Dai W, Liu T, He Y, Feng G, Zhao G. Human B cells have an active phagocytic capability and undergo immune activation upon phagocytosis of Mycobacterium tuberculosis. Immunobiology 2016; 221:558-67. [DOI: 10.1016/j.imbio.2015.12.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 12/06/2015] [Accepted: 12/06/2015] [Indexed: 02/06/2023]
|
31
|
Zerin T, Lee M, Jang WS, Nam KW, Song HY. Anti-inflammatory potential of ursolic acid in Mycobacterium tuberculosis-sensitized and concanavalin A-stimulated cells. Mol Med Rep 2016; 13:2736-44. [PMID: 26847129 DOI: 10.3892/mmr.2016.4840] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/19/2015] [Indexed: 11/06/2022] Open
Abstract
Ursolic acid (3-β-3-hydroxy-urs-12-ene-28-oic-acid; UA) is a triterpenoid carboxylic acid with various pharmaceutical properties. It is commonly found in apples, basil, berries, rosemary, peppermint, lavender, oregano, thyme, hawthorn and prunes. In the present study, the activities of UA against the Mycobacterium tuberculosis H37Rv‑induced release of a panel of inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 from RAW 264.7 murine macrophages, A549 alveolar epithelial cells and in concanavalin A (Con A)-stimulated rat splenocytes were investigated. In addition, the present study examined the ability of UA to reduce the expression levels of the inflammatory mediators, cyclooxygenase‑2 (COX‑2) and inducible nitric oxide synthase (iNOS) in the stimulated cells. The reduction of nitric oxide (NO) release by UA was also examined in the stimulated cells. UA significantly inhibited the mRNA expression levels of TNF‑α, IL‑1β and IL‑6 in the stimulated cells. The expression levels of COX‑2 and iNOS were also suppressed by UA, as was the release of NO at a significant level. The data indicated the potency of UA on different cell types, which may assist in the development of anti‑inflammatory drugs. In the case of adjunct host‑directed immune therapy for tuberculosis, UA may be used, in addition to established antibiotic therapies, to improve treatment efficacy and outcome due to their anti‑inflammatory potential. Further detailed investigations are required to establish its use as an anti-inflammatory.
Collapse
Affiliation(s)
- Tamanna Zerin
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 330‑090, Republic of Korea
| | - Minjung Lee
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 330‑090, Republic of Korea
| | - Woong Sik Jang
- Regional Innovation Center, Soonchunhyang University, Asan, Chungnam 336‑745, Republic of Korea
| | - Kung-Woo Nam
- Department of Life Science and Biotechnology, College of Natural Science, Soonchunhyang University, Asan, Chungnam 336‑745, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan, Chungnam 330‑090, Republic of Korea
| |
Collapse
|
32
|
Abstract
The emerging field of proteomics has contributed greatly to improving our understanding of the human pathogen Mycobacterium tuberculosis over the last two decades. In this chapter we provide a comprehensive overview of mycobacterial proteome research and highlight key findings. First, studies employing a combination of two-dimensional gel electrophoresis and mass spectrometry (MS) provided insights into the proteomic composition, initially of the whole bacillus and subsequently of subfractions, such as the cell wall, cytosol, and secreted proteins. Comparison of results obtained under various culture conditions, i.e., acidic pH, nutrient starvation, and low oxygen tension, aiming to mimic facets of the intracellular lifestyle of M. tuberculosis, provided initial clues to proteins relevant for intracellular survival and manipulation of the host cell. Further attempts were aimed at identifying the biological functions of the hypothetical M. tuberculosis proteins, which still make up a quarter of the gene products of M. tuberculosis, and at characterizing posttranslational modifications. Recent technological advances in MS have given rise to new methods such as selected reaction monitoring (SRM) and data-independent acquisition (DIA). These targeted, cutting-edge techniques combined with a public database of specific MS assays covering the entire proteome of M. tuberculosis allow the simple and reliable detection of any mycobacterial protein. Most recent studies attempt not only to identify but also to quantify absolute amounts of single proteins in the complex background of host cells without prior sample fractionation or enrichment. Finally, we will discuss the potential of proteomics to advance vaccinology, drug discovery, and biomarker identification to improve intervention and prevention measures for tuberculosis.
Collapse
|
33
|
Mvubu NE, Pillay B, Gamieldien J, Bishai W, Pillay M. Canonical pathways, networks and transcriptional factor regulation by clinical strains of Mycobacterium tuberculosis in pulmonary alveolar epithelial cells. Tuberculosis (Edinb) 2015; 97:73-85. [PMID: 26980499 DOI: 10.1016/j.tube.2015.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/30/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Limited knowledge exists on pathways, networks and transcriptional factors regulated within epithelial cells by diverse Mycobacterium tuberculosis genotypes. This study aimed to elucidate these mechanisms induced in A549 epithelial cells by dominant clinical strains in KwaZulu-Natal, South Africa. RNA for sequencing was extracted from epithelial cells at 48 h post-infection with 5 strains at a multiplicity of infection of approximately 10:1. Bioinformatics analysis performed with the RNA-Seq Tuxedo pipeline identified differentially expressed genes. Changes in pathways, networks and transcriptional factors were identified using Ingenuity Pathway Analysis (IPA). The interferon signalling and hepatic fibrosis/hepatic stellate cell activation pathways were among the top 5 canonical pathways in all strains. Hierarchical clustering for enrichment of cholesterol biosynthesis and immune associated pathways revealed similar patterns for Beijing and Unique; F15/LAM4/KZN and F11; and, F28 and H37Rv strains, respectively. However, the induction of top scoring networks varied among the strains. Among the transcriptional factors, only EHL, IRF7, PML, STAT1, STAT2 and VDR were induced by all clinical strains. Activation of the different pathways, networks and transcriptional factors revealed in the current study may be an underlying mechanism that results in the differential host response by clinical strains of M. tuberculosis.
Collapse
Affiliation(s)
- Nontobeko E Mvubu
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Balakrishna Pillay
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa.
| | - Junaid Gamieldien
- South African National Bioinformatics Institute/MRC Unit for Bioinformatics Capacity Development, University of the Western Cape, Bellville 7530, South Africa.
| | - William Bishai
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, 1550 Orleans St., Baltimore, MD, United State of America.
| | - Manormoney Pillay
- Medical Microbiology and Infection Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 719 Umbilo Road, Private Bag 7, Congella 4013, Durban, South Africa.
| |
Collapse
|
34
|
Hmama Z, Peña-Díaz S, Joseph S, Av-Gay Y. Immunoevasion and immunosuppression of the macrophage by Mycobacterium tuberculosis. Immunol Rev 2015; 264:220-32. [PMID: 25703562 DOI: 10.1111/imr.12268] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
By virtue of their position at the crossroads between the innate and adaptive immune response, macrophages play an essential role in the control of bacterial infections. Paradoxically, macrophages serve as the natural habitat to Mycobacterium tuberculosis (Mtb). Mtb subverts the macrophage's mechanisms of intracellular killing and antigen presentation, leading ultimately to the development of tuberculosis (TB) disease. Here, we describe mechanisms of Mtb uptake by the macrophage and address key macrophage functions that are targeted by Mtb-specific effector molecules enabling this pathogen to circumvent host immune response. The macrophage functions described in this review include fusion between phagosomes and lysosomes, production of reactive oxygen and nitrogen species, antigen presentation and major histocompatibility complex class II expression and trafficking, as well as autophagy and apoptosis. All these are Mtb-targeted key cellular pathways, normally working in concert in the macrophage to recognize, respond, and activate 'proper' immune responses. We further analyze and discuss major molecular interactions between Mtb virulence factors and key macrophage proteins and provide implications for vaccine and drug development.
Collapse
Affiliation(s)
- Zakaria Hmama
- Department of Medicine, Division of Infectious Diseases, Infection and Immunity Research Center, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
35
|
Meenu S, Thiagarajan S, Ramalingam S, Michael A, Ramalingam S. Modulation of host ubiquitin system genes in human endometrial cell line infected with Mycobacterium tuberculosis. Med Microbiol Immunol 2015; 205:163-71. [PMID: 26403675 DOI: 10.1007/s00430-015-0432-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/14/2015] [Indexed: 12/18/2022]
Abstract
Endometrium is one of the most commonly affected sites in genital tuberculosis. The understanding of its interaction with the tubercle bacilli is of paramount importance for studying the pathogenesis of this disease. The main objective of this work was to study the interplay between Mycobacterium tuberculosis and host endometrial epithelial cell lines (Ishikawa cell lines), and to identify the differentially expressed genes upon tuberculosis infection. To study this, suppression subtractive hybridization library was constructed using M. tuberculosis H37Rv-infected Ishikawa cell line harvested 24 h post-infection. The subtracted cDNA library was screened, and 105 differentially expressed genes were identified and grouped based on their functions. Since ubiquitination process has gained importance in targeting M. tuberculosis to xenophagy, ubiquitin system genes obtained in the library were selected, and time course analysis of their gene expression was performed. We observed an upregulation of mkrn1 and cops5 and downregulation of zfp91, ndfip2, ube2f, rnft1, psmb6, and psmd13 at 24 h post-infection. From the results obtained, we surmise that ubiquitination pathway genes may have roles in combating tuberculosis which are yet uncharted.
Collapse
Affiliation(s)
- S Meenu
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, Tamil Nadu, 641 004, India
- PSG College of Arts and Science, Coimbatore, Tamil Nadu, 641 004, India
| | - S Thiagarajan
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, Tamil Nadu, 641 004, India
| | - Sudha Ramalingam
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, Tamil Nadu, 641 004, India
| | - A Michael
- PSG College of Arts and Science, Coimbatore, Tamil Nadu, 641 004, India
| | - Sankaran Ramalingam
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, Tamil Nadu, 641 004, India.
| |
Collapse
|
36
|
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
|
37
|
Zerin T, Lee M, Jang WS, Nam KW, Song HY. Ursolic Acid Reduces Mycobacterium tuberculosis-Induced Nitric Oxide Release in Human Alveolar A549 cells. Mol Cells 2015; 38:610-5. [PMID: 26084752 PMCID: PMC4507026 DOI: 10.14348/molcells.2015.2328] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 05/10/2015] [Accepted: 05/14/2015] [Indexed: 01/28/2023] Open
Abstract
Alveolar epithelial cells have been functionally implicated in Mycobacterium tuberculosis infection. This study investigated the role of ursolic acid (UA)-a triterpenoid carboxylic acid with potent antioxidant, anti-tumor, anti-inflammatory, and anti-tuberculosis properties in mycobacterial infection of alveolar epithelial A549 cells. We observed that M. tuberculosis successfully entered A549 cells. Cytotoxi-city was mediated by nitric oxide (NO). A549 toxicity peaked along with NO generation 72 h after infection. The NO generated by mycobacterial infection in A549 cells was insufficient to kill mycobacteria, as made evident by the mycobacteria growth indicator tube time to detect (MGIT TTD) and viable cell count assays. Treatment of mycobacteria-infected cells with UA reduced the expression of inducible nitric oxide synthase, NO generation, and eventually improved cell viability. Moreover, UA was found to quench the translocation of the transcription factor, nuclear factor kappa B (NF-κB), from the cytosol to the nucleus in mycobacteria-infected cells. This study is the first to demonstrate the cytotoxic role of NO in the eradication of mycobacteria and the role of UA in reducing this cytotoxicity in A549 cells.
Collapse
Affiliation(s)
- Tamanna Zerin
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan 330-090,
Korea
| | - Minjung Lee
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan 330-090,
Korea
| | - Woong Sik Jang
- Regional Innovation Center, Soonchunhyang University, Asan 336-745,
Korea
| | - Kung-Woo Nam
- Department of Life Science and Biotechnology, College of Natural Science, Soonchunhyang University, Asan 336-745,
Korea
| | - Ho-yeon Song
- Department of Microbiology, School of Medicine, Soonchunhyang University, Cheonan 330-090,
Korea
| |
Collapse
|
38
|
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: 30] [Impact Index Per Article: 3.0] [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]
|
39
|
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
|
40
|
Baltierra-Uribe SL, García-Vásquez MDJ, Castrejón-Jiménez NS, Estrella-Piñón MP, Luna-Herrera J, García-Pérez BE. Mycobacteria entry and trafficking into endothelial cells. Can J Microbiol 2014; 60:569-77. [PMID: 25113069 DOI: 10.1139/cjm-2014-0087] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelial cells are susceptible to infection by mycobacteria, but the endocytic mechanisms that mycobacteria exploit to enter host cells and their mechanisms of intracellular transport are completely unknown. Using pharmacological inhibitors, we determined that the internalization of Mycobacterium tuberculosis (MTB), Mycobacterium smegmatis (MSM), and Mycobacterium abscessus (MAB) is dependent on the cytoskeleton and is differentially inhibited by cytochalasin D, nocodazole, cycloheximide, wortmannin, and amiloride. Using confocal microscopy, we investigated their endosomal trafficking by analyzing Rab5, Rab7, LAMP-1, and cathepsin D. Our results suggest that MSM exploits macropinocytosis to enter endothelial cells and that the vacuoles containing these bacteria fuse with lysosomes. Conversely, the entry of MTB seems to depend on more than one endocytic route, and the observation that only a subset of the intracellular bacilli was associated with phagolysosomes suggests that these bacteria are able to inhibit endosomal maturation to persist intracellularly. The route of entry for MAB depends mainly on microtubules, which suggests that MAB uses a different trafficking pathway. However, MAB is also able to inhibit endosomal maturation and can replicate intracellularly. Together, these findings provide the first evidence that mycobacteria modulate proteins of host endothelial cells to enter and persist within these cells.
Collapse
Affiliation(s)
- Shantal Lizbeth Baltierra-Uribe
- Department of Immunology, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, 11340 México, D.F., México
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Mycobacterium tuberculosis infection of the central nervous system is thought to be initiated once the bacilli have breached the blood brain barrier and are phagocytosed, primarily by microglial cells. In this study, the interactions of M. tuberculosis with neurons in vitro and in vivo were investigated. The data obtained demonstrate that neurons can act as host cells for M. tuberculosis. M. tuberculosis bacilli were internalized by murine neuronal cultured cells in a time-dependent manner after exposure, with superior uptake by HT22 cells compared to Neuro-2a cells (17.7% versus 9.8%). Internalization of M. tuberculosis bacilli by human SK-N-SH cultured neurons suggested the clinical relevance of the findings. Moreover, primary murine hippocampus-derived neuronal cultures could similarly internalize M. tuberculosis. Internalized M. tuberculosis bacilli represented a productive infection with retention of bacterial viability and replicative potential, increasing 2- to 4-fold within 48 h. M. tuberculosis bacillus infection of neurons was confirmed in vivo in the brains of C57BL/6 mice after intracerebral challenge. This study, therefore, demonstrates neurons as potential new target cells for M. tuberculosis within the central nervous system.
Collapse
|
42
|
Differential signaling of inducible nitric oxide synthase induction in Mycobacterium tuberculosis infected alveolar epithelial cell line A549 in response to cytokines IFN-γ, TNF-α and IL-1β. Int J Mycobacteriol 2014; 3:17-24. [DOI: 10.1016/j.ijmyco.2014.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/13/2014] [Accepted: 01/16/2014] [Indexed: 11/18/2022] Open
|
43
|
Bradburne CE, Verhoeven AB, Manyam GC, Chaudhry SA, Chang EL, Thach DC, Bailey CL, van Hoek ML. Temporal transcriptional response during infection of type II alveolar epithelial cells with Francisella tularensis live vaccine strain (LVS) supports a general host suppression and bacterial uptake by macropinocytosis. J Biol Chem 2013; 288:10780-91. [PMID: 23322778 DOI: 10.1074/jbc.m112.362178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pneumonic tularemia is caused by inhalation of Francisella tularensis, one of the most infectious microbes known. We wanted to study the kinetics of the initial and early interactions between bacterium and host cells in the lung. To do this, we examined the infection of A549 airway epithelial cells with the live vaccine strain (LVS) of F. tularensis. A549 cells were infected and analyzed for global transcriptional response at multiple time points up to 16 h following infection. At 15 min and 2 h, a strong transcriptional response was observed including cytoskeletal rearrangement, intracellular transport, and interferon signaling. However, at later time points (6 and 16 h), very little differential gene expression was observed, indicating a general suppression of the host response consistent with other reported cell lines and murine tissues. Genes for macropinocytosis and actin/cytoskeleton rearrangement were highly up-regulated and common to the 15 min and 2 h time points, suggesting the use of this method for bacterial entry into cells. We demonstrate macropinocytosis through the uptake of FITC-dextran and amiloride inhibition of Francisella LVS uptake. Our results suggest that macropinocytosis is a potential mechanism of intracellular entry by LVS and that the host cell response is suppressed during the first 2-6 h of infection. These results suggest that the attenuated Francisella LVS induces significant host cell signaling at very early time points after the bacteria's interaction with the cell.
Collapse
Affiliation(s)
- Christopher E Bradburne
- Center for Bio/Molecular Science and Engineering, United States Naval Research Laboratory, Washington, DC 20375, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
García-Pérez BE, De la Cruz-López JJ, Castañeda-Sánchez JI, Muñóz-Duarte AR, Hernández-Pérez AD, Villegas-Castrejón H, García-Latorre E, Caamal-Ley A, Luna-Herrera J. Macropinocytosis is responsible for the uptake of pathogenic and non-pathogenic mycobacteria by B lymphocytes (Raji cells). BMC Microbiol 2012; 12:246. [PMID: 23113903 PMCID: PMC3559283 DOI: 10.1186/1471-2180-12-246] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 10/12/2012] [Indexed: 12/18/2022] Open
Abstract
Background The classical roles of B cells include the production of antibodies and cytokines and the generation of immunological memory, these being key factors in the adaptive immune response. However, their role in innate immunity is currently being recognised. Traditionally, B cells have been considered non-phagocytic cells; therefore, the uptake of bacteria by B cells is not extensively documented. In this study, we analysed some of the features of non-specific bacterial uptake by B lymphocytes from the Raji cell line. In our model, B cells were infected with Mycobacterium tuberculosis (MTB), Mycobacterium smegmatis (MSM), and Salmonella typhimurium (ST). Results Our observations revealed that the Raji B cells were readily infected by the three bacteria that were studied. All of the infections induced changes in the cellular membrane during bacterial internalisation. M. smegmatis and S. typhimurium were able to induce important membrane changes that were characterised by abundant filopodia and lamellipodia formation. These membrane changes were driven by actin cytoskeletal rearrangements. The intracellular growth of these bacteria was also controlled by B cells. M. tuberculosis infection also induced actin rearrangement-driven membrane changes; however, the B cells were not able to control this infection. The phorbol 12-myristate 13-acetate (PMA) treatment of B cells induced filopodia and lamellipodia formation, the production of spacious vacuoles (macropinosomes), and the fluid-phase uptake that is characteristic of macropinocytosis. S. typhimurium infection induced the highest fluid-phase uptake, although both mycobacteria also induced fluid uptake. A macropinocytosis inhibitor such as amiloride was used and abolished the bacterial uptake and the fluid-phase uptake that is triggered during the bacterial infection. Conclusions Raji B cells can internalise S. typhimurium and mycobacteria through an active process, such as macropinocytosis, although the resolution of the infection depends on factors that are inherent in the virulence of each pathogen.
Collapse
Affiliation(s)
- Blanca Estela García-Pérez
- Immunology Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, D,F, México
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mycobacterium tuberculosis infection and tissue factor expression in macrophages. PLoS One 2012; 7:e45700. [PMID: 23029190 PMCID: PMC3454383 DOI: 10.1371/journal.pone.0045700] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 08/20/2012] [Indexed: 12/02/2022] Open
Abstract
A number of earlier studies reported the occurrence of thrombotic complications, particularly disseminated intravascular coagulation and deep vein thrombosis, in tuberculosis (TB) patients. The aberrant expression of tissue factor (TF), the primary activator of coagulation cascade, is known to be responsible for thrombotic disorders in many diseases including bacterial infections. Further, expression of TF by cells of the monocyte/macrophage lineage is also shown to contribute to the development and progression of local and systemic inflammatory reactions. In the present study, we have investigated whether Mycobacterium tuberculosis (Mtb) infection induces TF expression in macrophages, and various host and pathogenic factors responsible for TF expression. We have tested the effect of live virulent Mtb H37Rv, gamma-irradiated Mtb H37Rv (γ-Mtb) and various components derived from Mtb H37Rv on TF expression in macrophages. The data presented in the manuscript show that both live virulent Mtb and γ-Mtb treatments markedly increased TF activity in macrophages, predominantly in the CD14+ macrophages. Detailed studies using γ-Mtb showed that the increased TF activity in macrophages following Mtb treatment is the result of TF transcriptional activation. The signaling pathways of TF induction by Mtb appears to be distinct from that of LPS-induced TF expression. Mtb-mediated TF expression is dependent on cooperation of CD14/TLR2/TLR4 and probably yet another unknown receptor/cofactor. Mtb cell wall core components, mycolyl arabinogalactan peptidoglycan (mAGP), phosphatidylinositol mannoside-6 (PIM6) and lipomannan (LM) were identified as factors responsible for induction of TF in the order of mAGP>PIM6>LM. A direct contact between bacteria and macrophage and not Mtb-released soluble factors is critical for TF induction by Mtb. In summary, our data show that Mtb induces TF expression in macrophages and Mtb signaling pathways that elicit TF induction require cooperation of multiple receptors, co-receptors/co-factors including Toll-like receptors. The importance of TF in granuloma formation and containment of Mtb is discussed.
Collapse
|
46
|
Ocampo M, Aristizabal-Ramirez D, Rodriguez DM, Munoz M, Curtidor H, Vanegas M, Patarroyo MA, Patarroyo ME. The role of Mycobacterium tuberculosis Rv3166c protein-derived high-activity binding peptides in inhibiting invasion of human cell lines. Protein Eng Des Sel 2012; 25:235-42. [DOI: 10.1093/protein/gzs011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
47
|
Abstract
Mycobacterium tuberculosis is an old enemy of the human race, with evidence of infection observed as early as 5000 years ago. Although more host-restricted than Mycobacterium bovis, which can infect all warm-blooded vertebrates, M. tuberculosis can infect, and cause morbidity and mortality in, several veterinary species as well. As M. tuberculosis is one of the earliest described bacterial pathogens, the literature describing this organism is vast and overwhelming. This review strives to distill what is currently known about this bacterium and the disease it causes for the veterinary pathologist.
Collapse
Affiliation(s)
- K Sakamoto
- Department of Pathology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Dr, Athens, GA 30602-7388, USA.
| |
Collapse
|
48
|
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
|
49
|
Bai X, Ovrutsky AR, Kartalija M, Chmura K, Kamali A, Honda JR, Oberley-Deegan RE, Dinarello CA, Crapo JD, Chang LY, Chan ED. IL-32 expression in the airway epithelial cells of patients with Mycobacterium avium complex lung disease. Int Immunol 2011; 23:679-91. [PMID: 22033195 DOI: 10.1093/intimm/dxr075] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Lung disease due to Mycobacterium avium complex (MAC) organisms is increasing. A greater understanding of the host immune response to MAC organisms will provide a foundation to develop novel therapies for these recalcitrant infections. IL-32 is a newly described pro-inflammatory cytokine that enhances host immunity against various microbial pathogens. Cytokines that induce IL-32 such as interferon-gamma, IL-18, IL-12 and tumor necrosis factor-alpha are of considerable importance to mycobacterial immunity. We performed immunohistochemistry and morphometric analysis to quantify IL-32 expression in the lungs of 11 patients with MAC lung disease and 10 controls with normal lung tissues. After normalizing for basement membrane length, there was a profound increase in IL-32 expression in the airway epithelial cells of the MAC-infected lungs compared with controls. Following normalization for alveolar surface area, there was a trend toward increased IL-32 expression in type II alveolar cells and alveolar macrophages in the lungs of MAC patients. Human airway epithelial cells (BEAS-2B) infected with M. avium produced IL-32 by a nuclear factor-kappa B-dependent mechanism. In both BEAS-2B cells and human monocyte-derived macrophages, exogenous IL-32γ significantly reduced the growth of intracellular M. avium. This finding was corroborated by an increase in the number of intracellular M. avium recovered from THP-1 monocytes silenced for endogenous IL-32 expression. The anti-mycobacterial effect of IL-32 may be due, in part, to increased apoptosis of infected cells. These findings indicate that IL-32 facilitates host defense against MAC organisms but may also contribute to the airway inflammation associated with MAC pulmonary disease.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Denver Veterans Affairs Medical Center, Denver, CO 80220, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Stavrum R, Stavrum AK, Valvatne H, Riley LW, Ulvestad E, Jonassen I, Assmus J, Doherty TM, Grewal HMS. Modulation of transcriptional and inflammatory responses in murine macrophages by the Mycobacterium tuberculosis mammalian cell entry (Mce) 1 complex. PLoS One 2011; 6:e26295. [PMID: 22039457 PMCID: PMC3200323 DOI: 10.1371/journal.pone.0026295] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 09/23/2011] [Indexed: 11/22/2022] Open
Abstract
The outcome of many infections depends on the initial interactions between agent and host. Aiming at elucidating the effect of the M. tuberculosis Mce1 protein complex on host transcriptional and immunological responses to infection with M. tuberculosis, RNA from murine macrophages at 15, 30, 60 min, 4 and 10 hrs post-infection with M. tuberculosis H37Rv or Δ-mce1 H37Rv was analyzed by whole-genome microarrays and RT-QPCR. Immunological responses were measured using a 23-plex cytokine assay. Compared to uninfected controls, 524 versus 64 genes were up-regulated by 15 min post H37Rv- and Δ-mce1 H37Rv-infection, respectively. By 15 min post-H37Rv infection, a decline of 17 cytokines combined with up-regulation of Ccl24 (26.5-fold), Clec4a2 (23.2-fold) and Pparγ (10.5-fold) indicated an anti-inflammatory response initiated by IL-13. Down-regulation of Il13ra1 combined with up-regulation of Il12b (30.2-fold), suggested switch to a pro-inflammatory response by 4 hrs post H37Rv-infection. Whereas no significant change in cytokine concentration or transcription was observed during the first hour post Δ-mce1 H37Rv-infection, a significant decline of IL-1b, IL-9, IL-13, Eotaxin and GM-CSF combined with increased transcription of Il12b (25.1-fold) and Inb1 (17.9-fold) by 4 hrs, indicated a pro-inflammatory response. The balance between pro-and anti-inflammatory responses during the early stages of infection may have significant bearing on outcome.
Collapse
Affiliation(s)
- Ruth Stavrum
- Section of Microbiology and Immunology, the Gade Institute, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|