1
|
He J, Gao Y, Wang J, Hameed HMA, Wang S, Fang C, Tian X, Zhang J, Han X, Ju Y, Tan Y, Ma J, Ju J, Hu J, Liu J, Zhang T. EmbB and EmbC regulate the sensitivity of Mycobacterium abscessus to echinomycin. MLIFE 2024; 3:459-470. [PMID: 39359678 PMCID: PMC11442130 DOI: 10.1002/mlf2.12139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/28/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024]
Abstract
Treatment of Mycobacterium abscessus (Mab) infections is very challenging due to its intrinsic resistance to most available drugs. Therefore, it is crucial to discover novel anti-Mab drugs. In this study, we explored an intrinsic resistance mechanism through which Mab resists echinomycin (ECH). ECH showed activity against Mab at a minimum inhibitory concentration (MIC) of 2 µg/ml. A ΔembC strain in which the embC gene was knocked out showed hypersensitivity to ECH (MIC: 0.0078-0.0156 µg/ml). The MICs of ECH-resistant strains screened with reference to ΔembC ranged from 0.25 to 1 µg/ml. Mutations in EmbB, including D306A, D306N, R350G, V555I, and G581S, increased the Mab's resistance to ECH when overexpressed in ΔembC individually (MIC: 0.25-0.5 µg/ml). These EmbB mutants, edited using the CRISPR/Cpf1 system, showed heightened resistance to ECH (MIC: 0.25-0.5 µg/ml). The permeability of these Mab strains with edited genes and overexpression was reduced, as evidenced by an ethidium bromide accumulation assay, but it remained significantly higher than that of the parent Mab. In summary, our study demonstrates that ECH exerts potent anti-Mab activity and confirms that EmbB and EmbC are implicated in Mab's sensitivity to ECH. Mutation in EmbB may partially compensate for a loss of EmbC function.
Collapse
Affiliation(s)
- Jing He
- Institute of Physical Science and Information Technology Anhui University Hefei China
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
| | - Yamin Gao
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Jingyun Wang
- School of Pharmacy, Institute of Marine Drug Guangxi University of Traditional Chinese Medicine Nanning China
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology RNAM Center for Marine Microbiology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences Guangzhou China
| | - H M Adnan Hameed
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Shuai Wang
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Cuiting Fang
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Xirong Tian
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Jingran Zhang
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- School of Life Sciences University of Science and Technology of China Hefei China
| | - Xingli Han
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
| | - Yanan Ju
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- School of Life Sciences University of Science and Technology of China Hefei China
| | - Yaoju Tan
- State Key Laboratory of Respiratory Disease Guangzhou Chest Hospital Guangzhou China
| | - Junying Ma
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology RNAM Center for Marine Microbiology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences Guangzhou China
| | - Jianhua Ju
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology RNAM Center for Marine Microbiology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences Guangzhou China
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease Guangzhou Chest Hospital Guangzhou China
| | - Jianxiong Liu
- State Key Laboratory of Respiratory Disease Guangzhou Chest Hospital Guangzhou China
| | - Tianyu Zhang
- Institute of Physical Science and Information Technology Anhui University Hefei China
- State Key Laboratory of Respiratory Disease Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- China-New Zealand Joint Laboratory on Biomedicine and Health Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences Guangzhou China
- University of Chinese Academy of Sciences Beijing China
- School of Life Sciences University of Science and Technology of China Hefei China
- State Key Laboratory of Respiratory Disease Guangzhou Chest Hospital Guangzhou China
| |
Collapse
|
2
|
Italia A, Shaik MM, Peri F. Emerging Extracellular Molecular Targets for Innovative Pharmacological Approaches to Resistant Mtb Infection. Biomolecules 2023; 13:999. [PMID: 37371579 PMCID: PMC10296423 DOI: 10.3390/biom13060999] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Emerging pharmacological strategies that target major virulence factors of antibiotic-resistant Mycobacterium tuberculosis (Mtb) are presented and discussed. This review is divided into three parts corresponding to structures and functions important for Mtb pathogenicity: the cell wall, the lipoarabinomannan, and the secretory proteins. Within the cell wall, we further focus on three biopolymeric sub-components: mycolic acids, arabinogalactan, and peptidoglycan. We present a comprehensive overview of drugs and drug candidates that target cell walls, envelopes, and secretory systems. An understanding at a molecular level of Mtb pathogenesis is provided, and potential future directions in therapeutic strategies are suggested to access new drugs to combat the growing global threat of antibiotic-resistant Mtb infection.
Collapse
Affiliation(s)
| | | | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy; (A.I.); (M.M.S.)
| |
Collapse
|
3
|
Gong Y, Wang J, Li F, Zhu B. Polysaccharides and glycolipids of Mycobacterium tuberculosis and their induced immune responses. Scand J Immunol 2023; 97:e13261. [PMID: 39008002 DOI: 10.1111/sji.13261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 07/16/2024]
Abstract
Tuberculosis (TB) is a chronic infectious disease mainly caused by Mycobacterium tuberculosis (M. tuberculosis). The structures of polysaccharides and glycolipids at M. tuberculosis cell wall vary among different strains, which affect the physiology and pathogenesis of mycobacteria by activating or inhibiting innate and acquired immunity. Among them, some components such as lipomannan (LM) and lipoarabinomannan (LAM) activate innate immunity by recognizing some kinds of pattern recognition receptors (PRRs) like Toll-like receptors, while other components like mannose-capped lipoarabinomannan (ManLAM) could prevent innate immune responses by inhibiting the secretion of pro-inflammatory cytokines and maturation of phagosomes. In addition, many glycolipids can activate natural killer T (NKT) cells and CD1-restricted T cells to produce interferon-γ (IFN-γ). Furthermore, humoral immunity against cell wall components, such as antibodies against LAM, plays a role in immunity against M. tuberculosis infection. Cell wall polysaccharides and glycolipids of M. tuberculosis have potential applications as antigens and adjuvants for novel TB subunit vaccines.
Collapse
Affiliation(s)
- Yang Gong
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Juan Wang
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fei Li
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Mosquera-Restrepo SF, Zuberogoïtia S, Gouxette L, Layre E, Gilleron M, Stella A, Rengel D, Burlet-Schiltz O, Caro AC, Garcia LF, Segura C, Peláez Jaramillo CA, Rojas M, Nigou J. A Mycobacterium tuberculosis fingerprint in human breath allows tuberculosis detection. Nat Commun 2022; 13:7751. [PMID: 36517492 PMCID: PMC9751131 DOI: 10.1038/s41467-022-35453-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
An estimated one-third of tuberculosis (TB) cases go undiagnosed or unreported. Sputum samples, widely used for TB diagnosis, are inefficient at detecting infection in children and paucibacillary patients. Indeed, developing point-of-care biomarker-based diagnostics that are not sputum-based is a major priority for the WHO. Here, in a proof-of-concept study, we tested whether pulmonary TB can be detected by analyzing patient exhaled breath condensate (EBC) samples. We find that the presence of Mycobacterium tuberculosis (Mtb)-specific lipids, lipoarabinomannan lipoglycan, and proteins in EBCs can efficiently differentiate baseline TB patients from controls. We used EBCs to track the longitudinal effects of antibiotic treatment in pediatric TB patients. In addition, Mtb lipoarabinomannan and lipids were structurally distinct in EBCs compared to ex vivo cultured bacteria, revealing specific metabolic and biochemical states of Mtb in the human lung. This provides essential information for the rational development or improvement of diagnostic antibodies, vaccines and therapeutic drugs. Our data collectively indicate that EBC analysis can potentially facilitate clinical diagnosis of TB across patient populations and monitor treatment efficacy. This affordable, rapid and non-invasive approach seems superior to sputum assays and has the potential to be implemented at point-of-care.
Collapse
Affiliation(s)
- Sergio Fabián Mosquera-Restrepo
- Cellular Immunology and Immunogenetics Group (GICIG), Institute of Medical Research, Faculty of Medicine, University Research Headquarters (SIU), University of Antioquia (UdeA), Medellin, Colombia
| | - Sophie Zuberogoïtia
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Lucie Gouxette
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Emilie Layre
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Martine Gilleron
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Alexandre Stella
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - David Rengel
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Odile Burlet-Schiltz
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| | - Ana Cecilia Caro
- Interdisciplinary Group for Molecular Studies (GIEM), Institute of Chemistry, Faculty of Exact and Natural Sciences. University of Antioquia (UdeA), Medellin, Colombia
| | - Luis F Garcia
- Cellular Immunology and Immunogenetics Group (GICIG), Institute of Medical Research, Faculty of Medicine, University Research Headquarters (SIU), University of Antioquia (UdeA), Medellin, Colombia
| | - César Segura
- Malaria Group, University Research Headquarters, University of Antioquia (UdeA), Medellín, Colombia
| | - Carlos Alberto Peláez Jaramillo
- Interdisciplinary Group for Molecular Studies (GIEM), Institute of Chemistry, Faculty of Exact and Natural Sciences. University of Antioquia (UdeA), Medellin, Colombia
| | - Mauricio Rojas
- Cellular Immunology and Immunogenetics Group (GICIG), Institute of Medical Research, Faculty of Medicine, University Research Headquarters (SIU), University of Antioquia (UdeA), Medellin, Colombia.
- Flow Cytometry Core, University Research Headquarters (SIU), University of Antioquia, UdeA, Medellín, Colombia.
| | - Jérôme Nigou
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France.
| |
Collapse
|
5
|
Proteome Profile Changes Induced by Heterologous Overexpression of Mycobacterium tuberculosis-Derived Antigens PstS-1 (Rv0934) and Ag85B (Rv1886c) in Mycobacterium microti. Biomolecules 2022; 12:biom12121836. [PMID: 36551264 PMCID: PMC9775975 DOI: 10.3390/biom12121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/01/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022] Open
Abstract
The development of new tuberculosis vaccines remains a global priority, and recombinant vaccines are a frequently investigated option. These vaccines follow a molecular strategy that may enhance protective efficacy. However, their functional differences, particularly with respect to glycosylation, remain unknown. Recent studies have shown that glycosylation plays a key role in the host-pathogen interactions during immune recognition. The aim of this study was to determine the differences in the glycosylation profiles of two recombinant strains of Mycobacterium microti, overexpressing Ag85B (Rv1886c) and PstS-1 (Rv0934) antigens of M. tuberculosis. For each strain, the glycosylation profile was determined by Western blotting with lectins. The results showed the presence of mannosylated proteins and evidence of linked sialic acid proteins. Interestingly, different proteome and glycoproteome profiles were observed between the two recombinant strains and the wild-type strain. We have shown here that the construction of the recombinant strains of M. microti has altered the proteome and glycosylation profiles of these strains, leading us to ask what impact these changes might have on the immune response.
Collapse
|
6
|
Correia-Neves M, Nigou J, Mousavian Z, Sundling C, Källenius G. Immunological hyporesponsiveness in tuberculosis: The role of mycobacterial glycolipids. Front Immunol 2022; 13:1035122. [PMID: 36544778 PMCID: PMC9761185 DOI: 10.3389/fimmu.2022.1035122] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 12/09/2022] Open
Abstract
Glycolipids constitute a major part of the cell envelope of Mycobacterium tuberculosis (Mtb). They are potent immunomodulatory molecules recognized by several immune receptors like pattern recognition receptors such as TLR2, DC-SIGN and Dectin-2 on antigen-presenting cells and by T cell receptors on T lymphocytes. The Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic relatives, phosphatidylinositol mannosides (PIMs) and lipomannan (LM), as well as other Mtb glycolipids, such as phenolic glycolipids and sulfoglycolipids have the ability to modulate the immune response, stimulating or inhibiting a pro-inflammatory response. We explore here the downmodulating effect of Mtb glycolipids. A great proportion of the studies used in vitro approaches although in vivo infection with Mtb might also lead to a dampening of myeloid cell and T cell responses to Mtb glycolipids. This dampened response has been explored ex vivo with immune cells from peripheral blood from Mtb-infected individuals and in mouse models of infection. In addition to the dampening of the immune response caused by Mtb glycolipids, we discuss the hyporesponse to Mtb glycolipids caused by prolonged Mtb infection and/or exposure to Mtb antigens. Hyporesponse to LAM has been observed in myeloid cells from individuals with active and latent tuberculosis (TB). For some myeloid subsets, this effect is stronger in latent versus active TB. Since the immune response in individuals with latent TB represents a more protective profile compared to the one in patients with active TB, this suggests that downmodulation of myeloid cell functions by Mtb glycolipids may be beneficial for the host and protect against active TB disease. The mechanisms of this downmodulation, including tolerance through epigenetic modifications, are only partly explored.
Collapse
Affiliation(s)
- Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal,Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B's), Portuguese (PT) Government Associate Laboratory, Braga, Portugal,Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,School of Mathematics, Statistics, and Computer Science, College of Science, University of Tehran, Tehran, Iran,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,*Correspondence: Gunilla Källenius,
| |
Collapse
|
7
|
Enhanced immunogenicity of Mycobacterium bovis BCG through CRISPRi mediated depletion of AftC. Cell Surf 2022; 8:100088. [PMID: 36405350 PMCID: PMC9651938 DOI: 10.1016/j.tcsw.2022.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis causes the disease tuberculosis and affects a third of the world’s population. The recent COVID-19 pandemic exacerbated the situation with a projected 27% increase in tuberculosis related deaths. M. tuberculosis has an elaborate cell wall consisting of peptidoglycan, arabinogalactan and mycolic acids which shield the bacilli from the toxic bactericidal milieu within phagocytes. Amongst, the numerous glycosyltransferase enzymes involved in mycobacterial cell wall biosynthesis, arabinofuranosyltransferase C (aftC) is responsible for the branching of the arabinan domain in both arabinogalactan and lipoarabinomannan. Using Clustered Regularly Interspaced Short Palindromic Repeats interference (CRISPRi) we have generated aftC knockdowns in Mycobacterium bovis BCG and demonstrated the generation of a truncated, immunogenic lipoarabinomannan within its cell envelope. The aftC depleted BCG mutants were unable to form characteristic mycobacterial pellicular biofilms and elicit a potent immunostimulatory phenotype compared to wild type M. bovis BCG in a THP1 cell line. This study paves the way to further explore novel BCG mutants as promising vaccine boosters in preventing pulmonary tuberculosis.
Collapse
|
8
|
Kim H, Shin SJ. Pathological and protective roles of dendritic cells in Mycobacterium tuberculosis infection: Interaction between host immune responses and pathogen evasion. Front Cell Infect Microbiol 2022; 12:891878. [PMID: 35967869 PMCID: PMC9366614 DOI: 10.3389/fcimb.2022.891878] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are principal defense components that play multifactorial roles in translating innate immune responses to adaptive immunity in Mycobacterium tuberculosis (Mtb) infections. The heterogeneous nature of DC subsets follows their altered functions by interacting with other immune cells, Mtb, and its products, enhancing host defense mechanisms or facilitating pathogen evasion. Thus, a better understanding of the immune responses initiated, promoted, and amplified or inhibited by DCs in Mtb infection is an essential step in developing anti-tuberculosis (TB) control measures, such as host-directed adjunctive therapy and anti-TB vaccines. This review summarizes the recent advances in salient DC subsets, including their phenotypic classification, cytokine profiles, functional alterations according to disease stages and environments, and consequent TB outcomes. A comprehensive overview of the role of DCs from various perspectives enables a deeper understanding of TB pathogenesis and could be useful in developing DC-based vaccines and immunotherapies.
Collapse
|
9
|
Pattanaik KP, Sengupta S, Jit BP, Kotak R, Sonawane A. Host-Mycobacteria conflict: Immune responses of the host vs. the mycobacteria TLR2 and TLR4 ligands and concomitant host-directed therapy. Microbiol Res 2022; 264:127153. [DOI: 10.1016/j.micres.2022.127153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 12/15/2022]
|
10
|
SHI M, YIN P, GUO X, LI Q, SUN L, CAO X. Baicalein regulates NEDD4L-mediated TLR2 ubiquitination to relieve Mycobacterium tuberculosis-induced pneumonia in mice. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.54321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Min SHI
- Xi'an Peihua University Medical School, China
| | | | | | - Qian LI
- Xi'an Peihua University Medical School, China
| | - Lin SUN
- Xi'an Peihua University Medical School, China
| | - Xiaohua CAO
- People's Hospital of Yanliang District, China
| |
Collapse
|
11
|
Pouget M, Coussens AK, Ruggiero A, Koch A, Thomas J, Besra GS, Wilkinson RJ, Bhatt A, Pollakis G, Paxton WA. Generation of Liposomes to Study the Effect of Mycobacterium Tuberculosis Lipids on HIV-1 cis- and trans-Infections. Int J Mol Sci 2021; 22:ijms22041945. [PMID: 33669411 PMCID: PMC7920488 DOI: 10.3390/ijms22041945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/01/2021] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death among HIV-1-infected individuals and Mycobacterium tuberculosis (Mtb) co-infection is an early precipitate to AIDS. We aimed to determine whether Mtb strains differentially modulate cellular susceptibility to HIV-1 infection (cis- and trans-infection), via surface receptor interaction by their cell envelope lipids. Total lipids from pathogenic (lineage 4 Mtb H37Rv, CDC1551 and lineage 2 Mtb HN878, EU127) and non-pathogenic (Mycobacterium bovis BCG and Mycobacterium smegmatis) Mycobacterium strains were integrated into liposomes mimicking the lipid distribution and antigen accessibility of the mycobacterial cell wall. The resulting liposomes were tested for modulating in vitro HIV-1 cis- and trans-infection of TZM-bl cells using single-cycle infectious virus particles. Mtb glycolipids did not affect HIV-1 direct infection however, trans-infection of both R5 and X4 tropic HIV-1 strains were impaired in the presence of glycolipids from M. bovis, Mtb H37Rv and Mtb EU127 strains when using Raji-DC-SIGN cells or immature and mature dendritic cells (DCs) to capture virus. SL1, PDIM and TDM lipids were identified to be involved in DC-SIGN recognition and impairment of HIV-1 trans-infection. These findings indicate that variant strains of Mtb have differential effect on HIV-1 trans-infection with the potential to influence HIV-1 disease course in co-infected individuals.
Collapse
Affiliation(s)
- Marion Pouget
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (M.P.); (A.R.); (J.T.)
- UCD Centre for Experimental Pathogen Host Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Anna K. Coussens
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa; (A.K.C.); (A.K.); (R.J.W.)
- Walter and Eliza Hall Institute of Medical Research, Parkville 3279, Australia
| | - Alessandra Ruggiero
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (M.P.); (A.R.); (J.T.)
- Academic Department of Pediatrics (DPUO), IRCCS Ospedale Pediatrico Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Anastasia Koch
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa; (A.K.C.); (A.K.); (R.J.W.)
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (M.P.); (A.R.); (J.T.)
| | - Gurdyal S. Besra
- Institute of Microbiology and Infection and School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (G.S.B.); (A.B.)
| | - Robert J. Wilkinson
- Wellcome Center for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Observatory, Cape Town 7925, South Africa; (A.K.C.); (A.K.); (R.J.W.)
- Department of Infectious Diseases, Imperial College, London W2 1PG, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Apoorva Bhatt
- Institute of Microbiology and Infection and School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (G.S.B.); (A.B.)
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (M.P.); (A.R.); (J.T.)
- Correspondence: (G.P.); (W.A.P.); Tel.: +44-151-795-9681 (G.P.); +44-151-795-9605 (W.A.P.)
| | - William A. Paxton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (M.P.); (A.R.); (J.T.)
- Correspondence: (G.P.); (W.A.P.); Tel.: +44-151-795-9681 (G.P.); +44-151-795-9605 (W.A.P.)
| |
Collapse
|
12
|
Kilinç G, Saris A, Ottenhoff THM, Haks MC. Host-directed therapy to combat mycobacterial infections. Immunol Rev 2021; 301:62-83. [PMID: 33565103 PMCID: PMC8248113 DOI: 10.1111/imr.12951] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/27/2020] [Indexed: 12/27/2022]
Abstract
Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), are recognized by host innate immune cells, triggering a series of intracellular processes that promote mycobacterial killing. Mycobacteria, however, have developed multiple counter‐strategies to persist and survive inside host cells. By manipulating host effector mechanisms, including phagosome maturation, vacuolar escape, autophagy, antigen presentation, and metabolic pathways, pathogenic mycobacteria are able to establish long‐lasting infection. Counteracting these mycobacteria‐induced host modifying mechanisms can be accomplished by host‐directed therapeutic (HDT) strategies. HDTs offer several major advantages compared to conventional antibiotics: (a) HDTs can be effective against both drug‐resistant and drug‐susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host‐pathogen interactions that have been identified for Mtb for which potential HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host‐pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Layre E. Trafficking of Mycobacterium tuberculosis Envelope Components and Release Within Extracellular Vesicles: Host-Pathogen Interactions Beyond the Wall. Front Immunol 2020; 11:1230. [PMID: 32765485 PMCID: PMC7378356 DOI: 10.3389/fimmu.2020.01230] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022] Open
Abstract
Components of Mycobacterium tuberculosis (Mtb) envelope such as lipoproteins, lypoglycans, lipids, and glycolipids act as Pathogen Associated Molecular Patterns and/or antigens, hence contributing in different ways to the bacillus recognition, phagocytosis, and to immune responses modulation. However, Mtb envelope components are not only encountered at the bacillus-host direct contact but can act remotely from the bacillus envelope. Indeed, they are also released from the bacillus envelope and are detected in different compartments such as the infected cells endosomal compartments or in extracellular vesicles produced by the bacillus itself or by infected cells. Characterizing their trafficking is of main importance for our understanding of their role in host-pathogen interactions and consequently for their potential use as vaccine components. This review aims at providing an overview of the current knowledge of the nature of Mtb envelope components shuttled within extracellular vesicles, the interaction of these vesicles with host immune cells and the remaining black holes.
Collapse
Affiliation(s)
- Emilie Layre
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
14
|
Hoffpauir CT, Bell SL, West KO, Jing T, Wagner AR, Torres-Odio S, Cox JS, West AP, Li P, Patrick KL, Watson RO. TRIM14 Is a Key Regulator of the Type I IFN Response during Mycobacterium tuberculosis Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:153-167. [PMID: 32404352 DOI: 10.4049/jimmunol.1901511] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/20/2020] [Indexed: 01/05/2023]
Abstract
Tripartite motif-containing proteins (TRIMs) play a variety of recently described roles in innate immunity. Although many TRIMs regulate type I IFN expression following cytosolic nucleic acid sensing of viruses, their contribution to innate immune signaling and gene expression during bacterial infection remains largely unknown. Because Mycobacterium tuberculosis is an activator of cGAS-dependent cytosolic DNA sensing, we set out to investigate a role for TRIM proteins in regulating macrophage responses to M. tuberculosis In this study, we demonstrate that TRIM14, a noncanonical TRIM that lacks an E3 ubiquitin ligase RING domain, is a critical negative regulator of the type I IFN response in Mus musculus macrophages. We show that TRIM14 interacts with both cGAS and TBK1 and that macrophages lacking TRIM14 dramatically hyperinduce IFN stimulated gene (ISG) expression following M. tuberculosis infection, cytosolic nucleic acid transfection, and IFN-β treatment. Consistent with a defect in resolution of the type I IFN response, Trim14 knockout macrophages have more phospho-Ser754 STAT3 relative to phospho-Ser727 and fail to upregulate the STAT3 target Socs3, which is required to turn off IFNAR signaling. These data support a model whereby TRIM14 acts as a scaffold between TBK1 and STAT3 to promote phosphorylation of STAT3 at Ser727 and resolve ISG expression. Remarkably, Trim14 knockout macrophages hyperinduce expression of antimicrobial genes like Nos2 and are significantly better than control cells at limiting M. tuberculosis replication. Collectively, these data reveal an unappreciated role for TRIM14 in resolving type I IFN responses and controlling M. tuberculosis infection.
Collapse
Affiliation(s)
- Caitlyn T Hoffpauir
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Kelsi O West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Tao Jing
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77807; and
| | - Allison R Wagner
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Jeffery S Cox
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77807; and
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807;
| |
Collapse
|
15
|
Arai Y, Torigoe S, Matsumaru T, Yamasaki S, Fujimoto Y. The key entity of a DCAR agonist, phosphatidylinositol mannoside Ac 1PIM 1: its synthesis and immunomodulatory function. Org Biomol Chem 2020; 18:3659-3663. [PMID: 32356529 DOI: 10.1039/c9ob02724f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ac1PIM1 is a potential biosynthetic intermediate for phosphatidylinositol mannosides (PIMs) from Mycobacterium tuberculosis. We achieved the first synthesis of Ac1PIM1 by utilizing an allyl-type protecting group strategy and regioselective phosphorylation of inositol. A very potent agonist of an innate immune receptor DCAR, which is better than previously known agonists, is demonstrated.
Collapse
Affiliation(s)
- Yohei Arai
- Faculty of Science and Technology, Keio University. Hiyoshi 3-14-1, Yokohama, Kanagawa 223-8522, Japan.
| | | | | | | | | |
Collapse
|
16
|
Zhang L, Zhao Y, Gao Y, Wu L, Gao R, Zhang Q, Wang Y, Wu C, Wu F, Gurcha SS, Veerapen N, Batt SM, Zhao W, Qin L, Yang X, Wang M, Zhu Y, Zhang B, Bi L, Zhang X, Yang H, Guddat LW, Xu W, Wang Q, Li J, Besra GS, Rao Z. Structures of cell wall arabinosyltransferases with the anti-tuberculosis drug ethambutol. Science 2020; 368:1211-1219. [PMID: 32327601 DOI: 10.1126/science.aba9102] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/06/2020] [Accepted: 04/14/2020] [Indexed: 11/02/2022]
Abstract
The arabinosyltransferases EmbA, EmbB, and EmbC are involved in Mycobacterium tuberculosis cell wall synthesis and are recognized as targets for the anti-tuberculosis drug ethambutol. In this study, we determined cryo-electron microscopy and x-ray crystal structures of mycobacterial EmbA-EmbB and EmbC-EmbC complexes in the presence of their glycosyl donor and acceptor substrates and with ethambutol. These structures show how the donor and acceptor substrates bind in the active site and how ethambutol inhibits arabinosyltransferases by binding to the same site as both substrates in EmbB and EmbC. Most drug-resistant mutations are located near the ethambutol binding site. Collectively, our work provides a structural basis for understanding the biochemical function and inhibition of arabinosyltransferases and the development of new anti-tuberculosis agents.
Collapse
Affiliation(s)
- Lu Zhang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Response, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin 300353, China
| | - Yao Zhao
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Gao
- Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China
| | - Lijie Wu
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ruogu Gao
- University of Chinese Academy of Sciences, Beijing 100101, China.,National Laboratory of Biomacromolecules and Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing 100101, China
| | - Qi Zhang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yinan Wang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100101, China
| | - Chengyao Wu
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Fangyu Wu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Response, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin 300353, China
| | - Sudagar S Gurcha
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Natacha Veerapen
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Sarah M Batt
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Response, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin 300353, China
| | - Ling Qin
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiuna Yang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Manfu Wang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yan Zhu
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bing Zhang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lijun Bi
- National Laboratory of Biomacromolecules and Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing 100101, China
| | - Xian'en Zhang
- National Laboratory of Biomacromolecules and Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing 100101, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Luke W Guddat
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Wenqing Xu
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Quan Wang
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China. .,National Laboratory of Biomacromolecules and Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing 100101, China
| | - Jun Li
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Gurdyal S Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Zihe Rao
- Shanghai Institute for Advanced Immunochemical Studies, iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China. .,State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Response, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin 300353, China.,Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China.,National Laboratory of Biomacromolecules and Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, CAS, Beijing 100101, China
| |
Collapse
|
17
|
Zhou KL, Li X, Zhang XL, Pan Q. Mycobacterial mannose-capped lipoarabinomannan: a modulator bridging innate and adaptive immunity. Emerg Microbes Infect 2019; 8:1168-1177. [PMID: 31379262 PMCID: PMC6713153 DOI: 10.1080/22221751.2019.1649097] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mannose-capped lipoarabinomannan (ManLAM) is a high molecular mass amphipathic lipoglycan identified in pathogenic Mycobacterium tuberculosis (M. tb) and M. bovis Bacillus Calmette-Guérin (BCG). ManLAM, serves as both an immunogen and a modulator of the host immune system, and its critical role in mycobacterial survival during infection has been well-characterized. ManLAM can be recognized by various types of receptors on both innate and adaptive immune cells, including macrophages, dendritic cells (DCs), neutrophils, natural killer T (NKT) cells, T cells and B cells. MamLAM has been shown to affect phagocytosis, cytokine production, antigen presentation, T cell activation and polarization, as well as antibody production. Exploring the mechanisms underlying the roles of ManLAM during mycobacterial infection will aid in improving tuberculosis (TB) prevention, diagnosis and treatment interventions. In this review, we highlight the interaction between ManLAM and receptors, intracellular signalling pathways triggered by ManLAM and its roles in both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Kai-Liang Zhou
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China.,b The eighth hospital of Wuhan , Wuhan , People's Republic of China
| | - Xin Li
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| | - Xiao-Lian Zhang
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| | - Qin Pan
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| |
Collapse
|
18
|
Hu W, Yang S, Shimada Y, Münch M, Marín-Juez R, Meijer AH, Spaink HP. Infection and RNA-seq analysis of a zebrafish tlr2 mutant shows a broad function of this toll-like receptor in transcriptional and metabolic control and defense to Mycobacterium marinum infection. BMC Genomics 2019; 20:878. [PMID: 31747871 PMCID: PMC6869251 DOI: 10.1186/s12864-019-6265-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. Results tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/− control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2−/− larvae than in tlr2+/−, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2−/− versus tlr2+/− shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. Conclusion The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.
Collapse
Affiliation(s)
- Wanbin Hu
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Shuxin Yang
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yasuhito Shimada
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Magnus Münch
- Mathematical Institute, Leiden University, Leiden, the Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rubén Marín-Juez
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Annemarie H Meijer
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
19
|
Mycobacterium tuberculosis Lipoprotein and Lipoglycan Binding to Toll-Like Receptor 2 Correlates with Agonist Activity and Functional Outcomes. Infect Immun 2018; 86:IAI.00450-18. [PMID: 30037791 DOI: 10.1128/iai.00450-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/14/2018] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium tuberculosis causes persistent infection due to its ability to evade host immune responses. M. tuberculosis induces Toll-like receptor 2 (TLR2) signaling, which influences immune responses to M. tuberculosis TLR2 agonists expressed by M. tuberculosis include lipoproteins (e.g., LprG), the glycolipid phosphatidylinositol mannoside 6 (PIM6), and the lipoglycan lipomannan (LM). Another M. tuberculosis lipoglycan, mannose-capped lipoarabinomannan (ManLAM), lacks TLR2 agonist activity. In contrast, PILAM, from Mycobacterum smegmatis, does have TLR2 agonist activity. Our understanding of how M. tuberculosis lipoproteins and lipoglycans interact with TLR2 is limited, and binding of these molecules to TLR2 has not been measured directly. Here, we directly measured M. tuberculosis lipoprotein and lipoglycan binding to TLR2 and its partner receptor, TLR1. LprG, LAM, and LM were all found to bind to TLR2 in the absence of TLR1, but not to TLR1 in the absence of TLR2. Trimolecular interactions were revealed by binding of TLR2-LprG or TLR2-PIM6 complexes to TLR1, whereas binding of TLR2 to TLR1 was not detected in the absence of the lipoprotein or glycolipid. ManLAM exhibited low affinity for TLR2 in comparison to PILAM, LM, and LprG, which correlated with reduced ability of ManLAM to induce TLR2-mediated extracellular-signal-regulated kinase (ERK) activation and tumor necrosis factor alpha (TNF-α) secretion in macrophages. We provide the first direct affinity measurement and kinetic analysis of M. tuberculosis lipoprotein and lipoglycan binding to TLR2. Our results demonstrate that binding affinity correlates with the functional ability of agonists to induce TLR2 signaling.
Collapse
|
20
|
Turner J, Torrelles JB. Mannose-capped lipoarabinomannan in Mycobacterium tuberculosis pathogenesis. Pathog Dis 2018; 76:4953419. [PMID: 29722821 PMCID: PMC5930247 DOI: 10.1093/femspd/fty026] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 03/22/2018] [Indexed: 11/14/2022] Open
Abstract
Mannose-capped lipoarabinomannan (ManLAM), present in all members of the Mycobacterium tuberculosis complex and in other pathogenic Mycobacterium spp, is a high molecular mass amphipathic lipoglycan with a defined critical role in mycobacterial survival during infection. In particular, ManLAM is well-characterized for its importance in providing M. tuberculosis a safe portal of entry to phagocytes, regulating the intracellular trafficking network, as well as immune responses of infected host cells. These ManLAM immunological characteristics are thought to be linked to the subtle but unique and well-defined structural characteristics of this molecule, including but not limited to the degree of acylation, the length of the D-mannan and D-arabinan cores, the length of the mannose caps, as well as the presence of other acidic constituents such as succinates, lactates and/or malates, and also the presence of 5-methylthioxylosyl. The impact of all these structural features on ManLAM spatial conformation and biological functions during M. tuberculosis infection is still uncertain. In this review, we dissect the relationship between ManLAM structure and biological function addressing how this relationship determines M. tuberculosis interactions with host cells, and how it aids this exceptional pathogen during the course of infection.
Collapse
MESH Headings
- Acylation
- Carbohydrate Sequence
- Gene Expression Regulation/immunology
- Host-Pathogen Interactions/immunology
- Humans
- Immunity, Innate
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lipopolysaccharides/chemistry
- Lipopolysaccharides/immunology
- Mannose/chemistry
- Mannose/immunology
- Mannose Receptor
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/immunology
- Microbial Viability
- Mycobacterium tuberculosis/chemistry
- Mycobacterium tuberculosis/immunology
- Mycobacterium tuberculosis/pathogenicity
- Nod2 Signaling Adaptor Protein/genetics
- Nod2 Signaling Adaptor Protein/immunology
- Phagocytes/immunology
- Phagocytes/microbiology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Complement/genetics
- Receptors, Complement/immunology
- Toll-Like Receptors/genetics
- Toll-Like Receptors/immunology
- Tuberculosis, Pulmonary/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
Collapse
Affiliation(s)
- Joanne Turner
- Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX 78227-5301, USA
| | - Jordi B Torrelles
- Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX 78227-5301, USA
| |
Collapse
|
21
|
Gharun K, Senges J, Seidl M, Lösslein A, Kolter J, Lohrmann F, Fliegauf M, Elgizouli M, Alber M, Vavra M, Schachtrup K, Illert AL, Gilleron M, Kirschning CJ, Triantafyllopoulou A, Henneke P. Mycobacteria exploit nitric oxide-induced transformation of macrophages into permissive giant cells. EMBO Rep 2017; 18:2144-2159. [PMID: 29097394 DOI: 10.15252/embr.201744121] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 09/23/2017] [Accepted: 10/02/2017] [Indexed: 12/11/2022] Open
Abstract
Immunity to mycobacteria involves the formation of granulomas, characterized by a unique macrophage (MΦ) species, so-called multinucleated giant cells (MGC). It remains unresolved whether MGC are beneficial to the host, that is, by prevention of bacterial spread, or whether they promote mycobacterial persistence. Here, we show that the prototypical antimycobacterial molecule nitric oxide (NO), which is produced by MGC in excessive amounts, is a double-edged sword. Next to its antibacterial capacity, NO propagates the transformation of MΦ into MGC, which are relatively permissive for mycobacterial persistence. The mechanism underlying MGC formation involves NO-induced DNA damage and impairment of p53 function. Moreover, MGC have an unsurpassed potential to engulf mycobacteria-infected apoptotic cells, which adds a further burden to their antimycobacterial capacity. Accordingly, mycobacteria take paradoxical advantage of antimicrobial cellular efforts by driving effector MΦ into a permissive MGC state.
Collapse
Affiliation(s)
- Kourosh Gharun
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Julia Senges
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Seidl
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Lösslein
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Florens Lohrmann
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School for Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manfred Fliegauf
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Magdeldin Elgizouli
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Martina Vavra
- Division of Infectious Diseases, Department of Internal Medicine 2, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna L Illert
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martine Gilleron
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Carsten J Kirschning
- Institute of Medical Microbiology, Medical Center, University Duisburg-Essen, Essen, Germany
| | - Antigoni Triantafyllopoulou
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany .,Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Bazzi S, Modjtahedi H, Mudan S, Achkar M, Akle C, Bahr GM. Immunomodulatory effects of heat-killed Mycobacterium obuense on human blood dendritic cells. Innate Immun 2017; 23:592-605. [PMID: 28853313 DOI: 10.1177/1753425917727838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heat-killed (HK) Mycobacterium obuense is a novel immunomodulator, currently undergoing clinical evaluation as an immunotherapeutic agent in the treatment of cancer. Here, we examined the effect of in vitro exposure to HK M. obuense on the expression of different categories of surface receptors on human blood myeloid (m) and plasmacytoid (p) DCs. Moreover, we have characterized the cytokine and chemokine secretion patterns of purified total blood DCs stimulated with HK M. obuense. HK M. obuense significantly up-regulated the expression of CD11c, CD80, CD83, CD86, CD274 and MHC class II in whole-blood mDCs and CD80, CD123 and MHC class II in whole-blood pDCs. Down-regulation of CD195 expression in both DC subpopulations was also noted. Further analysis showed that HK M. obuense up-regulated the expression of CD80, CD83 and MHC class II on purified blood DC subpopulations. TLR2 and TLR1 were also identified to be engaged in mediating the HK M. obuense-induced up-regulation of surface receptor expression on whole blood mDCs. In addition, our data demonstrated that HK M. obuense augmented the secretion of CCL4, CCL5, CCL22, CXCL8, IL-6, IL-12p40 and TNF-α by purified total blood DCs. Taken together, our data suggest that HK M. obuense exerts potent differential immunomodulatory effects on human DC subpopulations.
Collapse
Affiliation(s)
- Samer Bazzi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK.,2 Biology Department, Faculty of Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| | - Helmout Modjtahedi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK
| | - Satvinder Mudan
- 3 St George's University of London, Imperial College, London and The Royal Marsden Hospital, London, UK
| | - Marcel Achkar
- 4 Clinical Laboratory Department, Nini Hospital, Tripoli, Lebanon
| | | | - Georges M Bahr
- 6 Faculty of Medicine and Medical Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| |
Collapse
|
23
|
Robinson RT, Huppler AR. The Goldilocks model of immune symbiosis with Mycobacteria and Candida colonizers. Cytokine 2017; 97:49-65. [PMID: 28570933 DOI: 10.1016/j.cyto.2017.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/12/2022]
Abstract
Mycobacteria and Candida species include significant human pathogens that can cause localized or disseminated infections. Although these organisms may appear to have little in common, several shared pathways of immune recognition and response are important for both control and infection-related pathology. In this article, we compare and contrast the innate and adaptive components of the immune system that pertain to these infections in humans and animal models. We also explore a relatively new concept in the mycobacterial field: biological commensalism. Similar to the well-established model of Candida infection, Mycobacteria species colonize their human hosts in equilibrium with the immune response. Perturbations in the immune response permit the progression to pathologic disease at the expense of the host. Understanding the immune factors required to maintain commensalism may aid with the development of diagnostic and treatment strategies for both categories of pathogens.
Collapse
Affiliation(s)
- Richard T Robinson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Anna R Huppler
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Children's Hospital and Health System, Children's Research Institute, Milwaukee, WI, USA.
| |
Collapse
|
24
|
Alvarez-Carbonell D, Garcia-Mesa Y, Milne S, Das B, Dobrowolski C, Rojas R, Karn J. Toll-like receptor 3 activation selectively reverses HIV latency in microglial cells. Retrovirology 2017; 14:9. [PMID: 28166799 PMCID: PMC5294768 DOI: 10.1186/s12977-017-0335-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/19/2017] [Indexed: 01/27/2023] Open
Abstract
Background Multiple toll-like receptors (TLRs) are expressed in cells of the monocytic lineage, including microglia, which constitute the major reservoir for human immunodeficiency virus (HIV) infection in the brain. We hypothesized that TLR receptor mediated responses to inflammatory conditions by microglial cells in the central nervous system (CNS) are able to induce latent HIV proviruses, and contribute to the etiology of HIV-associated neurocognitive disorders. Results Newly developed human microglial cell lines (hµglia), obtained by immortalizing human primary microglia with simian virus-40 (SV40) large T antigen and the human telomerase reverse transcriptase, were used to generate latently infected cells using a single-round HIV virus carrying a green fluorescence protein reporter (hµglia/HIV, clones HC01 and HC69). Treatment of these cells with a panel of TLR ligands showed surprisingly that two potent TLR3 agonists, poly (I:C) and bacterial ribosomal RNA potently reactivated HIV in hμglia/HIV cells. LPS (TLR4 agonist), flagellin (TLR5 agonist), and FSL-1 (TLR6 agonist) reactivated HIV to a lesser extent, while Pam3CSK4 (TLR2/1 agonist) and HKLM (TLR2 agonist) only weakly reversed HIV latency in these cells. While agonists for TLR2/1, 4, 5 and 6 reactivated HIV through transient NF-κB induction, poly (I:C), the TLR3 agonist, did not activate NF-κB, and instead induced the virus by a previously unreported mechanism mediated by IRF3. The selective induction of IRF3 by poly (I:C) was confirmed by chromatin immunoprecipitation (ChIP) analysis. In comparison, in latently infected rat-derived microglial cells (hT-CHME-5/HIV, clone HC14), poly (I:C), LPS and flagellin were only partially active. The TLR response profile in human microglial cells is also distinct from that shown by latently infected monocyte cell lines (THP-1/HIV, clone HA3, U937/HIV, clone HUC5, and SC/HIV, clone HSCC4), where TLR2/1, 4, 5, 6 or 8, but not for TLR3, 7 or 9, reactivated HIV. Conclusions TLR signaling, in particular TLR3 activation, can efficiently reactivate HIV transcription in infected microglia, but not in monocytes or T cells. The unique response profile of microglial cells to TLR3 is fundamental to understanding how the virus responds to continuous microbial exposure, especially during inflammatory episodes, that characterizes HIV infection in the CNS. Electronic supplementary material The online version of this article (doi:10.1186/s12977-017-0335-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Alvarez-Carbonell
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Yoelvis Garcia-Mesa
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Stephanie Milne
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Biswajit Das
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Curtis Dobrowolski
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Roxana Rojas
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave., SOM WRT 200, Cleveland, OH, 44106, USA.
| |
Collapse
|
25
|
Jankute M, Alderwick LJ, Noack S, Veerapen N, Nigou J, Besra GS. Disruption of Mycobacterial AftB Results in Complete Loss of Terminal β(1 → 2) Arabinofuranose Residues of Lipoarabinomannan. ACS Chem Biol 2017; 12:183-190. [PMID: 28033704 PMCID: PMC5259755 DOI: 10.1021/acschembio.6b00898] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Lipoarabinomannan
(LAM) and arabinogalactan (AG) are the two major
mycobacterial cell wall (lipo)polysaccharides, which contain a structurally
similar arabinan domain that is highly branched and assembled in a
stepwise fashion by variety of arabinofuranosyltransferases (ArafT). In addition to playing an essential role in mycobacterial
physiology, LAM and its biochemical precursor lipomannan possess potent
immunomodulatory activities that affect the host immune response.
In the search of additional mycobacterial ArafTs
that participate in the synthesis of the arabinan segment of LAM,
we disrupted aftB (MSMEG_6400) in Mycobacterium smegmatis. The deletion of chromosomal aftB locus could only be achieved in the presence of a rescue
plasmid carrying a functional copy of aftB, strongly
suggesting that it is essential for the viability of M. smegmatis. Isolation and detailed structural characterization of a LAM molecule
derived from the conditional mutant deficient in AftB revealed the
absence of terminal β(1 → 2)-linked arabinofuranosyl
residues. Furthermore, we demonstrated that truncated LAM displays
proinflammatory activity, which is due to its ability to activate
Toll-like receptor 2. All together, our results indicate that AftB
is an essential mycobacterial ArafT that plays a
role in the synthesis of the arabinan domain of LAM.
Collapse
Affiliation(s)
- Monika Jankute
- School
of Biosciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, B15 2TT Birmingham, United Kingdom
| | - Luke J. Alderwick
- School
of Biosciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, B15 2TT Birmingham, United Kingdom
| | - Stephan Noack
- Institute
of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich D-52425, Germany
| | - Natacha Veerapen
- School
of Biosciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, B15 2TT Birmingham, United Kingdom
| | - Jérôme Nigou
- Institut
de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Gurdyal S. Besra
- School
of Biosciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, B15 2TT Birmingham, United Kingdom
| |
Collapse
|
26
|
Zhang H. Upregulation of PIM2 by Underexpression of MicroRNA-135-5p Improves Survival Rates of Skin Allografts by Suppressing Apoptosis of Fibroblast Cells. Med Sci Monit 2017; 23:107-113. [PMID: 28064305 PMCID: PMC5240881 DOI: 10.12659/msm.897613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND It has been reported that miR-135-5p is involved with many diseases. In this study, we aimed at define the relationship between miR-135-5p level and burn patient survival after skin transplantation. MATERIAL AND METHODS Expression of miR-135-5p and PIM2 was measured using real-time PCR and Western blot analysis in the skin samples collected from burn patients who received skin graft or in the fibroblast cells transfected with miR-135-5p mimics or inhibitors. The regulatory association between miR-135-5p and PIM2 was verified using bioinformatics analysis and luciferase assay. RESULTS The expression level of miR-135-5p was determined in 60 tissue samples divided into 2 groups based on the presence of rejection (long survival n=30, and short survival n=30). We found that miR-135-5p was substantially downregulated in the long survival group. We then searched the miRNA database online with the "seed sequence" located within the 3'-UTR of the target gene, and then validated PIM2 to be the direct gene via luciferase reporter assay system. We also established the negative regulatory relationship between miR-135-5p and PIM2 via studying the relative luciferase activity. We also conducted real-time PCR and Western blot analysis to study the mRNA and protein expression level of PIM2 among different groups (long survival n=30, short survival n=30) or cells treated with scramble control, miR-135-5p mimics, PIM2 siRNA, and miR-135-5p inhibitors, indicating the negative regulatory relationship between MiR-135-5p and PIM2. We also conducted experiments to investigate the influence of miR-135-5p and PIM2 on viability and apoptosis of cells. The results showed miR-135-5p reduced the viability of cells, while PIM2 negatively interfered with the viability of cells, and miR-135-5p inhibited apoptosis and PIM2 suppressed apoptosis. CONCLUSIONS MiR-135-5p is involved with the prognosis of burn patients after skin transplantation. PIM2 is a virtual target of miR-135-5p, and there is a negative regulatory relationship between miR-135-5p and PIM2. MiR-135-5p and PIM2 interfered with the viability and apoptosis in cells.
Collapse
Affiliation(s)
- Hongtu Zhang
- Department of Burn and Plastic Surgery, Jining Number 1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
27
|
Structural basis of phosphatidyl-myo-inositol mannosides biosynthesis in mycobacteria. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:1355-1367. [PMID: 27826050 DOI: 10.1016/j.bbalip.2016.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/29/2016] [Accepted: 11/02/2016] [Indexed: 11/22/2022]
Abstract
Phosphatidyl-myo-inositol mannosides (PIMs) are glycolipids of unique chemical structure found in the inner and outer membranes of the cell envelope of all Mycobacterium species. The PIM family of glycolipids comprises phosphatidyl-myo-inositol mono-, di-, tri-, tetra-, penta-, and hexamannosides with different degrees of acylation. PIMs are considered not only essential structural components of the cell envelope but also the precursors of lipomannan and lipoarabinomannan, two major lipoglycans implicated in host-pathogen interactions. Since the description of the complete chemical structure of PIMs, major efforts have been committed to defining the molecular bases of its biosynthetic pathway. The structural characterization of the integral membrane phosphatidyl-myo-inositol phosphate synthase (PIPS), and that of three enzymes working at the protein-membrane interface, the phosphatidyl-myo-inositol mannosyltransferases A and B, and the acyltransferase PatA, established the basis of the early steps of the PIM pathway at the molecular level. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
|
28
|
Prakhar P, Holla S, Ghorpade DS, Gilleron M, Puzo G, Udupa V, Balaji KN. Ac2PIM-responsive miR-150 and miR-143 target receptor-interacting protein kinase 2 and transforming growth factor beta-activated kinase 1 to suppress NOD2-induced immunomodulators. J Biol Chem 2015; 290:26576-86. [PMID: 26391398 PMCID: PMC4646315 DOI: 10.1074/jbc.m115.662817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/15/2015] [Indexed: 11/06/2022] Open
Abstract
Specific and coordinated regulation of innate immune receptor-driven signaling networks often determines the net outcome of the immune responses. Here, we investigated the cross-regulation of toll-like receptor (TLR)2 and nucleotide-binding oligomerization domain (NOD)2 pathways mediated by Ac2PIM, a tetra-acylated form of mycobacterial cell wall component and muramyl dipeptide (MDP), a peptidoglycan derivative respectively. While Ac2PIM treatment of macrophages compromised their ability to induce NOD2-dependent immunomodulators like cyclooxygenase (COX)-2, suppressor of cytokine signaling (SOCS)-3, and matrix metalloproteinase (MMP)-9, no change in the NOD2-responsive NO, TNF-α, VEGF-A, and IL-12 levels was observed. Further, genome-wide microRNA expression profiling identified Ac2PIM-responsive miR-150 and miR-143 to target NOD2 signaling adaptors, RIP2 and TAK1, respectively. Interestingly, Ac2PIM was found to activate the SRC-FAK-PYK2-CREB cascade via TLR2 to recruit CBP/P300 at the promoters of miR-150 and miR-143 and epigenetically induce their expression. Loss-of-function studies utilizing specific miRNA inhibitors establish that Ac2PIM, via the miRNAs, abrogate NOD2-induced PI3K-PKCδ-MAPK pathway to suppress β-catenin-mediated expression of COX-2, SOCS-3, and MMP-9. Our investigation has thus underscored the negative regulatory role of Ac2PIM-TLR2 signaling on NOD2 pathway which could broaden our understanding on vaccine potential or adjuvant utilities of Ac2PIM and/or MDP.
Collapse
Affiliation(s)
- Praveen Prakhar
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India and
| | - Sahana Holla
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India and
| | - Devram Sampat Ghorpade
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India and
| | - Martine Gilleron
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS and Université de Toulouse, 31077 Toulouse, France
| | - Germain Puzo
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS and Université de Toulouse, 31077 Toulouse, France
| | - Vibha Udupa
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India and
| | | |
Collapse
|
29
|
Källenius G, Correia-Neves M, Buteme H, Hamasur B, Svenson SB. Lipoarabinomannan, and its related glycolipids, induce divergent and opposing immune responses to Mycobacterium tuberculosis depending on structural diversity and experimental variations. Tuberculosis (Edinb) 2015; 96:120-30. [PMID: 26586646 DOI: 10.1016/j.tube.2015.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/16/2015] [Indexed: 01/04/2023]
Abstract
Exposure to Mycobacterium tuberculosis (Mtb) may lead to active or latent tuberculosis, or clearance of Mtb, depending essentially on the quality of the host's immune response. This response is initiated through the interaction of Mtb cell wall surface components, mostly glycolipids, with cells of the innate immune system, particularly macrophages (Mφs) and dendritic cells (DCs). The way Mφs and DC alter their cytokine secretome, activate or inhibit different microbicidal mechanisms and present antigens and consequently trigger the T cell-mediated immune response impacts the host immune response against Mtb. Lipoarabinomannan (LAM) is one of the major cell wall components of Mtb. Mannosyl-capped LAM (ManLAM), and its related cell wall-associated types of glycolipids/lipoglycans, namely phosphatidylinositol mannosides (PIMs) and lipomannan (LM), exhibit important and distinct immunomodulatory properties. The structure, internal heterogeneity and abundance of these molecules vary between Mtb strains exhibiting distinct degrees of virulence. Thus ManLAM, LM and PIMs may be considered crucial Mtb-associated virulence factors in the pathogenesis of tuberculosis. Of particular relevance for this review, there is controversy about the specific immunomodulatory properties of these distinct glycolipids, particularly when tested as purified molecules in vitro. In addition to the variability in the glycolipid composition conflicting reports may also result from differences in the protocols used for glycolipid isolation and for in vitro experiments including immune cell types and procedures to generate them. Understanding the immunomodulatory properties of these cell wall glycolipids, how they differ between distinct Mtb strains, and how they influence the degree of Mtb virulence, is of utmost relevance to understand how the host mounts a protective or otherwise pathologic immune response. This is essential for the design of preventive strategies against tuberculosis. Thus, since clarifying the controversy on this matter is crucial we here review, summarize and discuss reported data from in vitro stimulation with the three major Mtb complex cell wall glycolipids (ManLAM, PIMs and LM) in an attempt to conciliate the conflicting findings.
Collapse
Affiliation(s)
- Gunilla Källenius
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, 118 83 Stockholm, Sweden.
| | - Margarida Correia-Neves
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, 118 83 Stockholm, Sweden; Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helen Buteme
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, 118 83 Stockholm, Sweden; Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O Box 7072, Kampala, Uganda
| | - Beston Hamasur
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, 118 83 Stockholm, Sweden
| | - Stefan B Svenson
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, 118 83 Stockholm, Sweden
| |
Collapse
|
30
|
The chemoselective O-glycosylation of alcohols in the presence of a phosphate diester and its application to the synthesis of oligomannosylated phosphatidyl inositols. Tetrahedron 2015. [DOI: 10.1016/j.tet.2015.06.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Vergne I, Gilleron M, Nigou J. Manipulation of the endocytic pathway and phagocyte functions by Mycobacterium tuberculosis lipoarabinomannan. Front Cell Infect Microbiol 2015; 4:187. [PMID: 25629008 PMCID: PMC4290680 DOI: 10.3389/fcimb.2014.00187] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/15/2014] [Indexed: 12/11/2022] Open
Abstract
Lipoarabinomannan is a major immunomodulatory lipoglycan found in the cell envelope of Mycobacterium tuberculosis and related human pathogens. It reproduces several salient properties of M. tuberculosis in phagocytic cells, including inhibition of pro-inflammatory cytokine production, inhibition of phagolysosome biogenesis, and inhibition of apoptosis as well as autophagy. In this review, we present our current knowledge on lipoarabinomannan structure and ability to manipulate the endocytic pathway as well as phagocyte functions. A special focus is put on the molecular mechanisms employed and the signaling pathways hijacked. Available information is discussed in the context of M. tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Isabelle Vergne
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique Toulouse, France ; Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III - Paul Sabatier Toulouse, France
| | - Martine Gilleron
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique Toulouse, France ; Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III - Paul Sabatier Toulouse, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique Toulouse, France ; Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III - Paul Sabatier Toulouse, France
| |
Collapse
|
32
|
Angala SK, Belardinelli JM, Huc-Claustre E, Wheat WH, Jackson M. The cell envelope glycoconjugates of Mycobacterium tuberculosis. Crit Rev Biochem Mol Biol 2014; 49:361-99. [PMID: 24915502 PMCID: PMC4436706 DOI: 10.3109/10409238.2014.925420] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Tuberculosis (TB) remains the second most common cause of death due to a single infectious agent. The cell envelope of Mycobacterium tuberculosis (Mtb), the causative agent of the disease in humans, is a source of unique glycoconjugates and the most distinctive feature of the biology of this organism. It is the basis of much of Mtb pathogenesis and one of the major causes of its intrinsic resistance to chemotherapeutic agents. At the same time, the unique structures of Mtb cell envelope glycoconjugates, their antigenicity and essentiality for mycobacterial growth provide opportunities for drug, vaccine, diagnostic and biomarker development, as clearly illustrated by recent advances in all of these translational aspects. This review focuses on our current understanding of the structure and biogenesis of Mtb glycoconjugates with particular emphasis on one of the most intriguing and least understood aspect of the physiology of mycobacteria: the translocation of these complex macromolecules across the different layers of the cell envelope. It further reviews the rather impressive progress made in the last 10 years in the discovery and development of novel inhibitors targeting their biogenesis.
Collapse
Affiliation(s)
- Shiva Kumar Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University , Fort Collins, CO , USA
| | | | | | | | | |
Collapse
|
33
|
Saraav I, Singh S, Sharma S. Outcome of Mycobacterium tuberculosis and Toll-like receptor interaction: immune response or immune evasion? Immunol Cell Biol 2014; 92:741-6. [PMID: 24983458 DOI: 10.1038/icb.2014.52] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 05/22/2014] [Accepted: 05/25/2014] [Indexed: 12/14/2022]
Abstract
Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, is an intracellular bacterium capable of surviving and persisting within host mononuclear cells. The host response against tubercle bacilli is dominated by fine-tuned interaction of innate and adaptive immune responses. Toll-like receptors (TLRs) play a critical role in the formation of this immune response by facilitating in elaboration of protective T helper type 1 (Th1) cytokines and microbicidal molecules, but the intracellular persistence of M. tuberculosis in the phagosome and processing and presentation of TLR ligands by host antigen-presenting cell leads to continuous and chronic TLR2 signaling. The prolonged stimulation of TLR ultimately results in elaboration of immunosuppressive cytokines and downregulation of antigen presentation by major histocompatibility complex (MHC) class II and therefore becomes beneficial for M. tuberculosis, resulting in its continued survival inside macrophages. An understanding of the host-pathogen interaction in tuberculosis is important to delineate the mechanisms that can modulate the immune response toward protection. This review focuses on the role of TLRs in immune response and immune evasion and how M. tuberculosis maintains its dominance over the host during infection. A precise understanding of the TLRs and M. tuberculosis interaction will undoubtedly lead to the development of novel therapies to combat tuberculosis.
Collapse
Affiliation(s)
- Iti Saraav
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, University of Delhi, Delhi, India
| | - Swati Singh
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, University of Delhi, Delhi, India
| | - Sadhna Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|
34
|
Reba SM, Li Q, Onwuzulike S, Ding X, Karim AF, Hernandez Y, Fulton SA, Harding CV, Lancioni CL, Nagy N, Rodriguez ME, Wearsch PA, Rojas RE. TLR2 engagement on CD4(+) T cells enhances effector functions and protective responses to Mycobacterium tuberculosis. Eur J Immunol 2014; 44:1410-21. [PMID: 24497180 PMCID: PMC4112943 DOI: 10.1002/eji.201344100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/19/2013] [Accepted: 01/31/2014] [Indexed: 11/08/2022]
Abstract
We have previously demonstrated that mycobacterial lipoproteins engage TLR2 on human CD4(+) T cells and upregulate TCR-triggered IFN-γ secretion and cell proliferation in vitro. Here we examined the role of CD4(+) T-cell-expressed TLR2 in Mycobacterium tuberculosis (MTB) Ag-specific T-cell priming and in protection against MTB infection in vivo. Like their human counterparts, mouse CD4(+) T cells express TLR2 and respond to TLR2 costimulation in vitro. This Th1-like response was observed in the context of both polyclonal and Ag-specific TCR stimulation. To evaluate the role of T-cell TLR2 in priming of CD4(+) T cells in vivo, naive MTB Ag85B-specific TCR transgenic CD4(+) T cells (P25 TCR-Tg) were adoptively transferred into Tlr2(-/-) recipient C57BL/6 mice that were then immunized with Ag85B and with or without TLR2 ligand Pam3 Cys-SKKKK. TLR2 engagement during priming resulted in increased numbers of IFN-γ-secreting P25 TCR-Tg T cells 1 week after immunization. P25 TCR-Tg T cells stimulated in vitro via TCR and TLR2 conferred more protection than T cells stimulated via TCR alone when adoptively transferred before MTB infection. Our findings indicate that TLR2 engagement on CD4(+) T cells increases MTB Ag-specific responses and may contribute to protection against MTB infection.
Collapse
Affiliation(s)
- Scott M Reba
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Qing Li
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Sophia Onwuzulike
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Xuedong Ding
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Ahmad F Karim
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Yeritza Hernandez
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Scott A Fulton
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Clifford V Harding
- Pathology Department, Case Western Reserve University, University
Hospitals, Cleveland, OH, USA
- Center for AIDS Research (CFAR), Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Christina L Lancioni
- Department of Pediatrics, Oregon Health and Science University,
Portland, OR, USA
| | - Nancy Nagy
- Pathology Department, Case Western Reserve University, University
Hospitals, Cleveland, OH, USA
| | - Myriam E Rodriguez
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
| | - Pamela A Wearsch
- Pathology Department, Case Western Reserve University, University
Hospitals, Cleveland, OH, USA
| | - Roxana E Rojas
- Department of Medicine, Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
- Center for AIDS Research (CFAR), Case Western Reserve University,
University Hospitals, Cleveland, OH, USA
- Department of Molecular Biology and Microbiology, Case Western
Reserve University, University Hospitals, Cleveland, OH, USA
| |
Collapse
|
35
|
Jiménez-Dalmaroni MJ, Radcliffe CM, Harvey DJ, Wormald MR, Verdino P, Ainge GD, Larsen DS, Painter GF, Ulevitch R, Beutler B, Rudd PM, Dwek RA, Wilson IA. Soluble human TLR2 ectodomain binds diacylglycerol from microbial lipopeptides and glycolipids. Innate Immun 2014; 21:175-93. [PMID: 24591200 DOI: 10.1177/1753425914524077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TLRs are key innate immune receptors that recognize conserved features of biological molecules that are found in microbes. In particular, TLR2 has been reported to be activated by different kinds of microbial ligands. To advance our understanding of the interaction of TLR2 with its ligands, the recombinant human TLR2 ectodomain (hTLR2ED) was expressed using a baculovirus/insect cell expression system and its biochemical, as well as ligand binding, properties were investigated. The hTLR2ED binds synthetic bacterial and mycoplasmal lipopeptides, lipoteichoic acid from Staphylococcus aureus, and synthetic lipoarabinomannan precursors from Mycobacterium at extracellular physiological conditions, in the absence of its co-receptors TLR1 and TLR6. We also determined that lipopeptides and glycolipids cannot bind simultaneously to hTLR2ED and that the phosphatidyl inositol mannoside 2 (Pim2) is the minimal lipoarabinomannan structure for binding to hTLR2ED. Binding of hTLR2ED to Pim4, which contains a diacylglycerol group with one of its acyl chains containing 19 carbon atoms, indicates that hTLR2ED can bind ligands with acyl chains longer than 16 carbon atoms. In summary, our data indicate that diacylglycerol is the ligand moiety of microbial glycolipids and lipoproteins that bind to hTLR2ED and that both types of ligands bind to the same binding site of hTLR2ED.
Collapse
Affiliation(s)
- Maximiliano J Jiménez-Dalmaroni
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK Department of Integrative Structural and Computational Biology, and the Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Catherine M Radcliffe
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - David J Harvey
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Mark R Wormald
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Petra Verdino
- Department of Integrative Structural and Computational Biology, and the Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Gary D Ainge
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - David S Larsen
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University Of Wellington, Gracefield Research Centre, Lower Hutt, New Zealand
| | - Richard Ulevitch
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Bruce Beutler
- Department of Genetics, The Scripps Research Institute, La Jolla, CA, USA
| | - Pauline M Rudd
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Raymond A Dwek
- Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, and the Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
36
|
Navigating through the maze of TLR2 mediated signaling network for better mycobacterium infection control. Biochimie 2014; 102:1-8. [PMID: 24594065 DOI: 10.1016/j.biochi.2014.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/21/2014] [Indexed: 12/12/2022]
Abstract
Toll-like receptor 2 (TLR2), a member of pattern recognition receptors (PRRs) abundant on macrophages, dendritic cells (DCs) and respiratory epithelial cells lining the lung, plays critical role in host immune response against Mycobacterium tuberculosis (MTB) infection. TLR2-mediated elimination of MTB involves multiple pathways such as promoting DCs maturation, generating biased Th1, Th2, Th17 type response, regulating the macrophage activation and cytokine secretion. MTB can also hijack the TLR2 signaling to subvert the host immunity by dampening the macrophages response to IFN-γ, suppressing the processing and presentation of antigens. This review summarizes the intricate network of TLR2-mediated signaling and Mycobacteria effectors involved in MTB-host interaction with an aim to find better target for improved tuberculosis control, especially the host-derived therapy targets. TLR2 agonists with potential to be included in novel tuberculosis vaccines are also discussed.
Collapse
|
37
|
Gao J, Sampson NS. A GMC oxidoreductase homologue is required for acetylation of glycopeptidolipid in Mycobacterium smegmatis. Biochemistry 2014; 53:611-3. [PMID: 24444367 PMCID: PMC3985799 DOI: 10.1021/bi4015083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The Mycobacterium tuberculosis Rv3409c gene is
required for modulation of the Toll-like receptor 2 (TLR-2) signaling
response in infected macrophages. Although each is annotated as encoding
a cholesterol oxidase, neither Rv3409c nor its ortholog MSMEG1604 is required for the metabolism of cholesterol
in mycobacteria. Here we report that a unique lipid, L1334, accumulates
in a MSMEG1604 transposon mutant in the Mycobacterium smegmatis cell envelope. L1334 is a polar glycopeptidolipid that is hyperrhamnosylated
and in which the 6-deoxytalose moiety is not acetylated. The alteration
of L1334 acetylation is consistent with a reduced level of interference
with TLR-2 signaling in mutant infected macrophages.
Collapse
Affiliation(s)
- Jin Gao
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States
| | | |
Collapse
|
38
|
Tanaka H, Ohira S, Yamaguchi Y, Takahashi T. Synthesis of a Phosphatidylinositol Dimannoside Using 2-(Azidomethyl)benzoate Mannosyl Donors. HETEROCYCLES 2014. [DOI: 10.3987/com-14-12944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Rodriguez ME, Loyd CM, Ding X, Karim AF, McDonald DJ, Canaday DH, Rojas RE. Mycobacterial phosphatidylinositol mannoside 6 (PIM6) up-regulates TCR-triggered HIV-1 replication in CD4+ T cells. PLoS One 2013; 8:e80938. [PMID: 24282561 PMCID: PMC3839890 DOI: 10.1371/journal.pone.0080938] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 10/17/2013] [Indexed: 11/19/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of mortality among those infected with human immunodeficiency virus (HIV-1) worldwide. HIV-1 load and heterogeneity are increased both locally and systemically in active TB. Mycobacterium tuberculosis (MTB) infection supports HIV-1 replication through dysregulation of host cytokines, chemokines, and their receptors. However the possibility that mycobacterial molecules released from MTB infected macrophages directly interact with CD4(+) T cells triggering HIV-1 replication has not been fully explored. We studied the direct effect of different MTB molecules on HIV-1 replication (R5-tropic strain Bal) in anti-CD3- stimulated CD4(+) T cells from healthy donors in an antigen presenting cell (APC)-free system. PIM6, a major glycolipid of the mycobacterial cell wall, induced significant increases in the percent of HIV-1 infected T cells and the viral production in culture supernatants. In spite of structural relatedness, none of the other three major MTB cell wall glycolipids had significant impact on HIV-1 replication in T cells. Increased levels of IFN-γ in culture supernatants from cells treated with PIM6 indicate that HIV-1 replication is likely dependent on enhanced T cell activation. In HEK293 cells transfected with TLR2, PIM6 was the strongest TLR2 agonist among the cell wall associated glycolipids tested. PIM6 increased the percentage of HIV infected cells and viral particles in the supernatant in a T-cell-based reporter cell line (JLTRg-R5) transfected with TLR1 and TLR2 but not in the cells transfected with the empty vector (which lack TLR2 expression) confirming that PIM6-induced HIV-1 replication depends at least partially on TLR2 signaling.
Collapse
Affiliation(s)
- Myriam E. Rodriguez
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Candace M. Loyd
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Xuedong Ding
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Ahmad F. Karim
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David J. McDonald
- Department of Molecular Biology and Microbiology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - David H. Canaday
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Cleveland VA Medical Center, Cleveland, Ohio, United States of America
| | - Roxana E. Rojas
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Molecular Biology and Microbiology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
Blanc L, Castanier R, Mishra AK, Ray A, Besra GS, Sutcliffe I, Vercellone A, Nigou J. Gram-positive bacterial lipoglycans based on a glycosylated diacylglycerol lipid anchor are microbe-associated molecular patterns recognized by TLR2. PLoS One 2013; 8:e81593. [PMID: 24278450 PMCID: PMC3836763 DOI: 10.1371/journal.pone.0081593] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 10/24/2013] [Indexed: 11/18/2022] Open
Abstract
Innate immune recognition is the first line of host defense against invading microorganisms. It is a based on the detection, by pattern recognition receptors (PRRs), of invariant molecular signatures that are unique to microorganisms. TLR2 is a PRR that plays a major role in the detection of Gram-positive bacteria by recognizing cell envelope lipid-linked polymers, also called macroamphiphiles, such as lipoproteins, lipoteichoic acids and mycobacterial lipoglycans. These microbe-associated molecular patterns (MAMPs) display a structure based on a lipid anchor, being either an acylated cysteine, a glycosylated diacylglycerol or a mannosyl-phosphatidylinositol respectively, and having in common a diacylglyceryl moiety. A fourth class of macroamphiphile, namely lipoglycans, whose lipid anchor is made, as for lipoteichoic acids, of a glycosylated diacylglycerol unit rather than a mannosyl-phosphatidylinositol, is found in Gram-positive bacteria and produced by certain Actinobacteria, including Micrococcus luteus, Stomatococcus mucilaginosus and Corynebacterium glutamicum. We report here that these alternative lipoglycans are also recognized by TLR2 and that they stimulate TLR2-dependant cytokine production, including IL-8, TNF-α and IL-6, and cell surface co-stimulatory molecule CD40 expression by a human macrophage cell line. However, they differ by their co-receptor requirement and the magnitude of the innate immune response they elicit. M. luteus and S. mucilaginosus lipoglycans require TLR1 for recognition by TLR2 and induce stronger responses than C. glutamicum lipoglycan, sensing of which by TLR2 is dependent on TLR6. These results expand the repertoire of MAMPs recognized by TLR2 to lipoglycans based on a glycosylated diacylglycerol lipid anchor and reinforce the paradigm that macroamphiphiles based on such an anchor, including lipoteichoic acids and alternative lipoglycans, induce TLR2-dependant innate immune responses.
Collapse
Affiliation(s)
- Landry Blanc
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Romain Castanier
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Arun K. Mishra
- National Institute for Medical Research, London, United Kingdom
| | - Aurélie Ray
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Gurdyal S. Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Iain Sutcliffe
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Alain Vercellone
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Jérôme Nigou
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
- * E-mail:
| |
Collapse
|
41
|
Lipids derived from virulent Francisella tularensis broadly inhibit pulmonary inflammation via toll-like receptor 2 and peroxisome proliferator-activated receptor α. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1531-40. [PMID: 23925884 DOI: 10.1128/cvi.00319-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Francisella tularensis is a Gram-negative facultative intracellular pathogen that causes an acute lethal respiratory disease in humans. The heightened virulence of the pathogen is linked to its unique ability to inhibit Toll-like receptor (TLR)-mediated inflammatory responses. The bacterial component and mechanism of this inhibition are unknown. Here we show that lipids isolated from virulent but not attenuated strains of F. tularensis are not detected by host cells, inhibit production of proinflammatory cytokines by primary macrophages in response to known TLR ligands, and suppress neutrophil recruitment in vivo. We further show that lipid-mediated inhibition of inflammation is dependent on TLR2, MyD88, and the nuclear hormone and fatty acid receptor peroxisome proliferator-activated receptor α (PPARα). Pathogen lipid-mediated interference with inflammatory responses through the engagement of TLR2 and PPARα represents a novel manipulation of host signaling pathways consistent with the ability of highly virulent F. tularensis to efficiently evade host immune responses.
Collapse
|
42
|
IL12B expression is sustained by a heterogenous population of myeloid lineages during tuberculosis. Tuberculosis (Edinb) 2013; 93:343-56. [PMID: 23491716 DOI: 10.1016/j.tube.2013.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/21/2013] [Accepted: 02/04/2013] [Indexed: 01/17/2023]
Abstract
IL12B is required for resistance to Mycobacterium tuberculosis (Mtb) infection, promoting the initiation and maintenance of Mtb-specific effector responses. While this makes the IL12-pathway an attractive target for experimental tuberculosis (TB) therapies, data regarding what lineages express IL12B after infection is established are limited. This is not obvious in the lung, an organ in which both hematopoietic and non-hematopoietic lineages produce IL12p40 upon pathogen encounter. Here, we use radiation bone marrow chimeras and Yet40 reporter mice to determine what lineages produce IL12p40 during experimental TB. We observed that hematopoietic IL12p40-production was sufficient to control Mtb, with no contribution by non-hematopoietic lineages. Furthermore, rather than being produced by a single subset, IL12p40 was produced by cells that were heterogenous in their size, granularity, autofluorescence and expression of CD11c, CD11b and CD8α. While depending on the timepoint and tissue examined, the surface phenotype of IL12p40-producers most closely resembled macrophages based on previous surveys of lung myeloid lineages. Importantly, depletion of CD11c(hi) cells during infection had no affect on lung IL12p40-concentrations. Collectively, our data demonstrate that IL12p40 production is sustained by a heterogenous population of myeloid lineages during experimental TB, and that redundant mechanisms of IL12p40-production exist when CD11c(hi) lineages are absent.
Collapse
|
43
|
Bacterial cell wall macroamphiphiles: Pathogen-/microbe-associated molecular patterns detected by mammalian innate immune system. Biochimie 2013; 95:33-42. [DOI: 10.1016/j.biochi.2012.06.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 06/06/2012] [Indexed: 02/02/2023]
|
44
|
Mishra AK, Alves JE, Krumbach K, Nigou J, Castro AG, Geurtsen J, Eggeling L, Saraiva M, Besra GS. Differential arabinan capping of lipoarabinomannan modulates innate immune responses and impacts T helper cell differentiation. J Biol Chem 2012; 287:44173-83. [PMID: 23144457 PMCID: PMC3531733 DOI: 10.1074/jbc.m112.402396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Toll-like receptors (TLRs) recognize pathogens by interacting with pathogen-associated molecular patterns, such as the phosphatidylinositol-based lipoglycans, lipomannan (LM) and lipoarabinomannan (LAM). Such structures are present in several pathogens, including Mycobacterium tuberculosis, being important for the initiation of immune responses. It is well established that the interaction of LM and LAM with TLR2 is a process dependent on the structure of the ligands. However, the implications of structural variations on TLR2 ligands for the development of T helper (Th) cell responses or in the context of in vivo responses are less studied. Herein, we used Corynebacterium glutamicum as a source of lipoglycan intermediates for host interaction studies. In this study, we have deleted a putative glycosyltransferase, NCgl2096, from C. glutamicum and found that it encodes for a novel α(1→2)arabinofuranosyltransferase, AftE. Biochemical analysis of the lipoglycans obtained in the presence (wild type) or absence of NCgl2096 showed that AftE is involved in the biosynthesis of singular arabinans of LAM. In its absence, the resulting molecule is a hypermannosylated (hLM) form of LAM. Both LAM and hLM were recognized by dendritic cells, mainly via TLR2, and triggered the production of several cytokines. hLM was a stronger stimulus for in vitro cytokine production and, as a result, a more potent inducer of Th17 responses. In vivo data confirmed hLM as a stronger inducer of cytokine responses and suggested the involvement of pattern recognition receptors other than TLR2 as sensors for lipoglycans.
Collapse
Affiliation(s)
- Arun K Mishra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
In vitro immunomodulation of a whole blood IFN-γ release assay enhances T cell responses in subjects with latent tuberculosis infection. PLoS One 2012; 7:e48027. [PMID: 23144722 PMCID: PMC3483295 DOI: 10.1371/journal.pone.0048027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 09/25/2012] [Indexed: 11/25/2022] Open
Abstract
Background Activation of innate immunity via pathogen recognition receptors (PRR) modulates adaptive immune responses. PRR ligands are being exploited as vaccine adjuvants and as therapeutics, but their utility in diagnostics has not been explored. Interferon-gamma (IFN-γ) release assays (IGRAs) are functional T cell assays used to diagnose latent tuberculosis infection (LTBI); however, novel approaches are needed to improve their sensitivity. Methods In vitro immunomodulation of a whole blood IGRA (QuantiFERON®-TB GOLD In-Tube) with Toll-like receptor agonists poly(I:C), LPS, and imiquimod was performed on blood from subjects with LTBI and negative controls. Results In vitro immunomodulation significantly enhanced the response of T cells stimulated with M. tuberculosis antigens from subjects with LTBI but not from uninfected controls. Immunomodulation of IGRA revealed T cell responses in subjects with LTBI whose T cells otherwise do not respond to in vitro stimulation with antigens alone. Similar to their in vivo functions, addition of poly(I:C) and LPS to whole blood induced secretion of inflammatory cytokines and IFN-α and enhanced the surface expression of antigen presenting and costimulatory molecules on antigen presenting cells. Conclusions In vitro immunomodulation of whole blood IGRA may be an effective strategy for enhancing the sensitivity of T cells for diagnosis of LTBI.
Collapse
|
46
|
Elass-Rochard E, Rombouts Y, Coddeville B, Maes E, Blervaque R, Hot D, Kremer L, Guérardel Y. Structural determination and Toll-like receptor 2-dependent proinflammatory activity of dimycolyl-diarabino-glycerol from Mycobacterium marinum. J Biol Chem 2012; 287:34432-44. [PMID: 22798072 DOI: 10.1074/jbc.m112.378083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although it was identified in the cell wall of several pathogenic mycobacteria, the biological properties of dimycolyl-diarabino-glycerol have not been documented yet. In this study an apolar glycolipid, presumably corresponding to dimycolyl-diarabino-glycerol, was purified from Mycobacterium marinum and subsequently identified as a 5-O-mycolyl-β-Araf-(1→2)-5-O-mycolyl-α-Araf-(1→1')-glycerol (designated Mma_DMAG) using a combination of nuclear magnetic resonance spectroscopy and mass spectrometry analyses. Lipid composition analysis revealed that mycolic acids were dominated by oxygenated mycolates over α-mycolates and devoid of trans-cyclopropane functions. Highly purified Mma_DMAG was used to demonstrate its immunomodulatory activity. Mma_DMAG was found to induce the secretion of proinflammatory cytokines (TNF-α, IL-8, IL-1β) in human macrophage THP-1 cells and to trigger the expression of ICAM-1 and CD40 cell surface antigens. This activation mechanism was dependent on TLR2, but not on TLR4, as demonstrated by (i) the use of neutralizing anti-TLR2 and -TLR4 antibodies and by (ii) the detection of secreted alkaline phosphatase in HEK293 cells co-transfected with the human TLR2 and secreted embryonic alkaline phosphatase reporter genes. In addition, transcriptomic analyses indicated that various genes encoding proinflammatory factors were up-regulated after exposure of THP-1 cells to Mma_DMAG. Importantly, a wealth of other regulated genes related to immune and inflammatory responses, including chemokines/cytokines and their respective receptors, adhesion molecules, and metalloproteinases, were found to be modulated by Mma_DMAG. Overall, this study suggests that DMAG may be an active cell wall glycoconjugate driving host-pathogen interactions and participating in the immunopathogenesis of mycobacterial infections.
Collapse
Affiliation(s)
- Elisabeth Elass-Rochard
- Université Lille Nord de France, Université Lille1, Unité de Glycobiologie Structurale et Fonctionnelle, UGSF, IFR 147, France.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Mukherjee R, Chatterji D. Glycopeptidolipids: immuno-modulators in greasy mycobacterial cell envelope. IUBMB Life 2012; 64:215-25. [PMID: 22252955 DOI: 10.1002/iub.602] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 11/10/2011] [Indexed: 11/12/2022]
Abstract
Species of opportunistic mycobacteria are the major causative agent for disseminating pulmonary infections in immuno-compromised individuals. These naturally resistant strains recruit a unique type of glycolipid known as glycopeptidolipids (GPLs), noncovalently attached to the outer surface of their thick lipid rich cell envelope. Species specific GPLs constitute the chemical determinants of most nontuberculous mycobacterial serotypes, and their absence from the cell surface confers altered colony morphology, hydrophobicity, and inability to grow as biofilms. The objective of this review is to present a comprehensive account and highlight the renewed interest on this much neglected group of pleiotropic molecules with respect to their structural diversity and biosynthesis. In addition, the role of GPLs in mycobacterial survival, both intracellular and in the environment is also discussed. It also explores the possibility of identifying new targets for intervening Mycobacterium avium complex-related infections. These antigenic molecules have been considered to play a pivotal role in immune suppression and can also induce various cytokine mediated innate immune responses, the molecular mechanism of which remains obscure.
Collapse
Affiliation(s)
- Raju Mukherjee
- Swiss Federal Institute of Technology, Lausanne, Switzerland.
| | | |
Collapse
|
48
|
Hutacharoen P, Ruchirawat S, Boonyarattanakalin S. Biological Activities of Synthetic Oligosaccharides and Glycolipids from Mycobacteria. J Carbohydr Chem 2011. [DOI: 10.1080/07328303.2011.621041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Panatpong Hutacharoen
- a School of Bio-Chemical Engineering and Technology, Sirindhorn International Institute of Technology , Thammasat University , Pathum Thani , 12121 , Thailand
- b Program in Chemical Biology, Chulabhorn Graduate Institute and the Center of Excellence on Environmental Health, Toxicology and Management of Chemicals , Vibhavadee-Rangsit Highway , Lak Si , Bangkok , 10210 , Thailand
| | - Somsak Ruchirawat
- b Program in Chemical Biology, Chulabhorn Graduate Institute and the Center of Excellence on Environmental Health, Toxicology and Management of Chemicals , Vibhavadee-Rangsit Highway , Lak Si , Bangkok , 10210 , Thailand
- c Laboratory of Medicinal Chemistry, Chulabhorn Research Institute , Vibhavadee-Rangsit Highway , Lak Si , Bangkok , 10210 , Thailand
| | - Siwarutt Boonyarattanakalin
- a School of Bio-Chemical Engineering and Technology, Sirindhorn International Institute of Technology , Thammasat University , Pathum Thani , 12121 , Thailand
| |
Collapse
|
49
|
Krishna S, Ray A, Dubey SK, Larrouy-Maumus G, Chalut C, Castanier R, Noguera A, Gilleron M, Puzo G, Vercellone A, Nampoothiri KM, Nigou J. Lipoglycans contribute to innate immune detection of mycobacteria. PLoS One 2011; 6:e28476. [PMID: 22164297 PMCID: PMC3229593 DOI: 10.1371/journal.pone.0028476] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 11/09/2011] [Indexed: 12/20/2022] Open
Abstract
Innate immune recognition is based on the detection, by pattern recognition receptors (PRRs), of molecular structures that are unique to microorganisms. Lipoglycans are macromolecules specific to the cell envelope of mycobacteria and related genera. They have been described to be ligands, as purified molecules, of several PRRs, including the C-type lectins Mannose Receptor and DC-SIGN, as well as TLR2. However, whether they are really sensed by these receptors in the context of a bacterium infection remains unclear. To address this question, we used the model organism Mycobacterium smegmatis to generate mutants altered for the production of lipoglycans. Since their biosynthesis cannot be fully abrogated, we manipulated the biosynthesis pathway of GDP-Mannose to obtain some strains with either augmented (∼1.7 fold) or reduced (∼2 fold) production of lipoglycans. Interestingly, infection experiments demonstrated a direct correlation between the amount of lipoglycans in the bacterial cell envelope on one hand and the magnitude of innate immune signaling in TLR2 reporter cells, monocyte/macrophage THP-1 cell line and human dendritic cells, as revealed by NF-κB activation and IL-8 production, on the other hand. These data establish that lipoglycans are bona fide Microbe-Associated Molecular Patterns contributing to innate immune detection of mycobacteria, via TLR2 among other PRRs.
Collapse
Affiliation(s)
- Shyam Krishna
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
- Biotechnology Division, National Institute for Interdisciplinary Science and Technology, CSIR, Thiruvananthapuram, India
| | - Aurélie Ray
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Shiv K. Dubey
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Gérald Larrouy-Maumus
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Christian Chalut
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Romain Castanier
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Audrey Noguera
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Martine Gilleron
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Germain Puzo
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Alain Vercellone
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - K. Madhavan Nampoothiri
- Biotechnology Division, National Institute for Interdisciplinary Science and Technology, CSIR, Thiruvananthapuram, India
- * E-mail: (JN); (KMN)
| | - Jérôme Nigou
- CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
- * E-mail: (JN); (KMN)
| |
Collapse
|
50
|
Court N, Rose S, Bourigault ML, Front S, Martin OR, Dowling JK, Kenny EF, O'Neill L, Erard F, Quesniaux VFJ. Mycobacterial PIMs inhibit host inflammatory responses through CD14-dependent and CD14-independent mechanisms. PLoS One 2011; 6:e24631. [PMID: 21949737 PMCID: PMC3174970 DOI: 10.1371/journal.pone.0024631] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 08/16/2011] [Indexed: 01/08/2023] Open
Abstract
Mycobacteria develop strategies to evade the host immune system. Among them, mycobacterial LAM or PIMs inhibit the expression of pro-inflammatory cytokines by activated macrophages. Here, using synthetic PIM analogues, we analyzed the mode of action of PIM anti-inflammatory effects. Synthetic PIM(1) isomer and PIM(2) mimetic potently inhibit TNF and IL-12 p40 expression induced by TLR2 or TLR4 pathways, but not by TLR9, in murine macrophages. We show inhibition of LPS binding to TLR4/MD2/CD14 expressing HEK cells by PIM(1) and PIM(2) analogues. More specifically, the binding of LPS to CD14 was inhibited by PIM(1) and PIM(2) analogues. CD14 was dispensable for PIM(1) and PIM(2) analogues functional inhibition of TLR2 agonists induced TNF, as shown in CD14-deficient macrophages. The use of rough-LPS, that stimulates TLR4 pathway independently of CD14, allowed to discriminate between CD14-dependent and CD14-independent anti-inflammatory effects of PIMs on LPS-induced macrophage responses. PIM(1) and PIM(2) analogues inhibited LPS-induced TNF release by a CD14-dependent pathway, while IL-12 p40 inhibition was CD14-independent, suggesting that PIMs have multifold inhibitory effects on the TLR4 signalling pathway.
Collapse
Affiliation(s)
- Nathalie Court
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| | - Stéphanie Rose
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| | - Marie-Laure Bourigault
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| | - Sophie Front
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| | - Olivier R. Martin
- University of Orléans Institut de Chimie Organique et Analytique, Orléans, France
- CNRS UMR6005, Orléans, France
| | | | - Elaine F. Kenny
- School of Biochemistry and Immunology, Trinity College Dublin, Ireland
| | - Luke O'Neill
- School of Biochemistry and Immunology, Trinity College Dublin, Ireland
| | - François Erard
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| | - Valerie F. J. Quesniaux
- University of Orléans Molecular Immunology and Embryology, Orléans, France
- CNRS UMR6218, Orléans, France
| |
Collapse
|