1
|
Kireev FD, Lopatnikova JA, Alshevskaya AA, Sennikov SV. Role of Tumor Necrosis Factor in Tuberculosis. Biomolecules 2025; 15:709. [PMID: 40427602 DOI: 10.3390/biom15050709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Tumor necrosis factor (TNF) is a key immunoregulatory cytokine with a dual role in the host response to Mycobacterium tuberculosis. While essential for granuloma formation, macrophage activation, and containment of latent infection, TNF can also contribute to tissue damage and immune pathology. This review systematically analyzes over 300 peer-reviewed studies published between 1980 and 2024, highlighting the molecular and cellular mechanisms of TNF action in tuberculosis (TB). Particular attention is given to TNF receptor signaling pathways, the balance between protective and pathological immune responses, and the modulation of TNF activity during anti-TNF therapy in patients with autoimmune diseases. We discuss how different TNF inhibitors vary in their capacity to interfere with host defense mechanisms, with monoclonal antibodies carrying a higher reactivation risk than receptor-based agents. To enhance conceptual clarity, we provide newly developed schematic representations that integrate current knowledge on TNF-driven immune dynamics, including its interaction with other cytokines, effects on granuloma stability, and role in intracellular bacterial control. Understanding the pleiotropic functions of TNF in tuberculosis pathogenesis is crucial for developing safe immunomodulatory strategies and optimizing the clinical management of patients at risk of latent TB reactivation.
Collapse
Affiliation(s)
- Fedor D Kireev
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099 Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education "I.M. Sechenov First Moscow State Medical University" under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia
| | - Julia A Lopatnikova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099 Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education "I.M. Sechenov First Moscow State Medical University" under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia
| | - Alina A Alshevskaya
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education "I.M. Sechenov First Moscow State Medical University" under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia
| | - Sergey V Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099 Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education "I.M. Sechenov First Moscow State Medical University" under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia
| |
Collapse
|
2
|
Geng J, Long J, Hu Q, Liu M, Ge A, Du Y, Zhang T, Jin Y, Yang H, Chen S, Duan G. Current status of cyclopropane fatty acids on bacterial cell membranes characteristics and physiological functions. Microb Pathog 2025; 200:107295. [PMID: 39805345 DOI: 10.1016/j.micpath.2025.107295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Wide-ranging sophisticated physiological activities of cell membranes are associated with changes in fatty acid structure and composition. The cfa gene is a core regulator of cell membrane fatty acid cyclopropanation reaction. Its encoded cyclopropane fatty acid synthase (CFA synthase) catalyzes the binding of unsaturated fatty acid (UFA) to methylene groups, which undergoes cyclopropanation modification to produce cyclopropane fatty acids (CFAs). Compelling evidence suggests a large role for the cfa gene and CFAs in bacterial adaptive responses. This review provides an overview of the relationship of CFAs with bacterial cell membrane properties and physiological functions, including the roles of cell membrane fluidity, stability, and permeability to protons, bacterial growth, acid resistance, and especially in bacterial antibiotic resistance and pathogenicity. The dysregulation and inhibition of the cfa gene may serve as potential therapeutic targets against bacterial drug resistance and pathogenicity. Therefore, elucidating the biological function of CFAs during the stationary growth phase therefore provides invaluable insights into the bacterial pathogenesis and the development of novel antimicrobial agents.
Collapse
Affiliation(s)
- Juan Geng
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jinzhao Long
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Quanman Hu
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Mengyue Liu
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Anmin Ge
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China; Penglai Center for Disease Control and Prevention, Yantai, China
| | - Yazhe Du
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Teng Zhang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuefei Jin
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Haiyan Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shuaiyin Chen
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China.
| | - Guangcai Duan
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Vadankula GR, Nilkanth VV, Rizvi A, Yandrapally S, Agarwal A, Chirra H, Biswas R, Arifuddin M, Nema V, Mallika A, Mande SC, Banerjee S. Confronting Tuberculosis: A Synthetic Quinoline-Isonicotinic Acid Hydrazide Hybrid Compound as a Potent Lead Molecule Against Mycobacterium tuberculosis. ACS Infect Dis 2024; 10:2288-2302. [PMID: 38717380 DOI: 10.1021/acsinfecdis.4c00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The current tuberculosis (TB) treatment is challenged by a complex first-line treatment for drug-sensitive (DS) TB. Additionally, the prevalence of multidrug (MDR)- and extensively drug (XDR)-resistant TB necessitates the search for new drug prototypes. We synthesized and screened 30 hybrid compounds containing aminopyridine and 2-chloro-3-formyl quinoline to arrive at a compound with potent antimycobacterial activity, UH-NIP-16. Subsequently, antimycobacterial activity against DS and MDR Mycobacterium tuberculosis (M.tb) strains were performed. It demonstrated an MIC50 value of 1.86 ± 0.21 μM for laboratory pathogenic M.tb strain H37Rv and 3.045 ± 0.813 μM for a clinical M.tb strain CDC1551. UH-NIP-16 also decreased the MIC50 values of streptomycin, isoniazid, ethambutol, and bedaquiline to about 45, 55, 68, and 76%, respectively, when used in combination, potentiating their activities. The molecule was active against a clinical MDR M.tb strain. Cytotoxicity on PBMCs from healthy donors and on human cell lines was found to be negligible. Further, blind docking of UH-NIP-16 using Auto Dock Vina and MGL tools onto diverse M.tb proteins showed high binding affinities with multiple M.tb proteins, the top five targets being metabolically critical proteins CelA1, DevS, MmaA4, lysine acetyltransferase, and immunity factor for tuberculosis necrotizing toxin. These bindings were confirmed by fluorescence spectroscopy using a representative protein, MmaA4. Envisaging that a pathogen will have a lower probability of developing resistance to a hybrid molecule with multiple targets, we propose that UH-NIP-16 can be further developed as a lead molecule with the bacteriostatic potential against M.tb, both alone and in combination with first-line drugs.
Collapse
Affiliation(s)
- Govinda Raju Vadankula
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Vipul V Nilkanth
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India
| | - Arshad Rizvi
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Sriram Yandrapally
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Anushka Agarwal
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| | - Hepshibha Chirra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rashmita Biswas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mohammed Arifuddin
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Vijay Nema
- Molecular Biology Division, ICMR-National Institute for Translational Virology and AIDS Research, Pune 411026, India
| | - Alvala Mallika
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Shekhar C Mande
- National Centre for Cell Science, Pune 411007, India
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India
| | - Sharmistha Banerjee
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad (UoH), Hyderabad 500046, India
| |
Collapse
|
4
|
Bories P, Rima J, Tranier S, Marcoux J, Grimoire Y, Tomaszczyk M, Launay A, Fata K, Marrakchi H, Burlet‐Schiltz O, Mourey L, Ducoux‐Petit M, Bardou F, Bon C, Quémard A. HadBD dehydratase from Mycobacterium tuberculosis fatty acid synthase type II: A singular structure for a unique function. Protein Sci 2024; 33:e4964. [PMID: 38501584 PMCID: PMC10949391 DOI: 10.1002/pro.4964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Worldwide, tuberculosis is the second leading infectious killer and multidrug resistance severely hampers disease control. Mycolic acids are a unique category of lipids that are essential for viability, virulence, and persistence of the causative agent, Mycobacterium tuberculosis (Mtb). Therefore, enzymes involved in mycolic acid biosynthesis represent an important class of drug targets. We previously showed that the (3R)-hydroxyacyl-ACP dehydratase (HAD) protein HadD is dedicated mainly to the production of ketomycolic acids and plays a determinant role in Mtb biofilm formation and virulence. Here, we discovered that HAD activity requires the formation of a tight heterotetramer between HadD and HadB, a HAD unit encoded by a distinct chromosomal region. Using biochemical, structural, and cell-based analyses, we showed that HadB is the catalytic subunit, whereas HadD is involved in substrate binding. Based on HadBDMtb crystal structure and substrate-bound models, we identified determinants of the ultra-long-chain lipid substrate specificity and revealed details of structure-function relationship. HadBDMtb unique function is partly due to a wider opening and a higher flexibility of the substrate-binding crevice in HadD, as well as the drastically truncated central α-helix of HadD hotdog fold, a feature described for the first time in a HAD enzyme. Taken together, our study shows that HadBDMtb , and not HadD alone, is the biologically relevant functional unit. These results have important implications for designing innovative antivirulence molecules to fight tuberculosis, as they suggest that the target to consider is not an isolated subunit, but the whole HadBD complex.
Collapse
Affiliation(s)
- Pascaline Bories
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Julie Rima
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Samuel Tranier
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Yasmina Grimoire
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Mathilde Tomaszczyk
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Anne Launay
- Service de TP de BiochimieUniversité de Toulouse, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Karine Fata
- Service de TP de BiochimieUniversité de Toulouse, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Hedia Marrakchi
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Odile Burlet‐Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Lionel Mourey
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Manuelle Ducoux‐Petit
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Fabienne Bardou
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Cécile Bon
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| | - Annaïk Quémard
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, Université Toulouse III ‐ Paul Sabatier (UPS)ToulouseFrance
| |
Collapse
|
5
|
Wang H, Liu D, Zhou X. Effect of Mycolic Acids on Host Immunity and Lipid Metabolism. Int J Mol Sci 2023; 25:396. [PMID: 38203570 PMCID: PMC10778799 DOI: 10.3390/ijms25010396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 01/12/2024] Open
Abstract
Mycolic acids constitute pivotal constituents within the cell wall structure of Mycobacterium tuberculosis. Due to their structural diversity, the composition of mycolic acids exhibits substantial variations among different strains, endowing them with the distinctive label of being the 'signature' feature of mycobacterial species. Within Mycobacterium tuberculosis, the primary classes of mycolic acids include α-, keto-, and methoxy-mycolic acids. While these mycolic acids are predominantly esterified to the cell wall components (such as arabinogalactan, alginate, or glucose) of Mycobacterium tuberculosis, a fraction of free mycolic acids are secreted during in vitro growth of the bacterium. Remarkably, different types of mycolic acids possess varying capabilities to induce foamy macro-phages and trigger immune responses. Additionally, mycolic acids play a regulatory role in the lipid metabolism of host cells, thereby exerting influence over the progression of tuberculosis. Consequently, the multifaceted properties of mycolic acids shape the immune evasion strategy employed by Mycobacterium tuberculosis. A comprehensive understanding of mycolic acids is of paramount significance in the pursuit of developing tuberculosis therapeutics and unraveling the intricacies of its pathogenic mechanisms.
Collapse
Affiliation(s)
- Haoran Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| | - Dingpu Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| |
Collapse
|
6
|
Hailu E, Cantillon D, Madrazo C, Rose G, Wheeler PR, Golby P, Adnew B, Gagneux S, Aseffa A, Gordon SV, Comas I, Young DB, Waddell SJ, Larrouy-Maumus G, Berg S. Lack of methoxy-mycolates characterizes the geographically restricted lineage 7 of Mycobacterium tuberculosis complex. Microb Genom 2023; 9:mgen001011. [PMID: 37171244 PMCID: PMC10272862 DOI: 10.1099/mgen.0.001011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/07/2023] [Indexed: 05/13/2023] Open
Abstract
Lineage 7 (L7) emerged in the phylogeny of the Mycobacterium tuberculosis complex (MTBC) subsequent to the branching of 'ancient' lineage 1 and prior to the Eurasian dispersal of 'modern' lineages 2, 3 and 4. In contrast to the major MTBC lineages, the current epidemiology suggests that prevalence of L7 is highly confined to the Ethiopian population, or when identified outside of Ethiopia, it has mainly been in patients of Ethiopian origin. To search for microbiological factors that may contribute to its restricted distribution, we compared the genome of L7 to the genomes of globally dispersed MTBC lineages. The frequency of predicted functional mutations in L7 was similar to that documented in other lineages. These include mutations characteristic of modern lineages - such as constitutive expression of nitrate reductase - as well as mutations in the VirS locus that are commonly found in ancient lineages. We also identified and characterized multiple lineage-specific mutations in L7 in biosynthesis pathways of cell wall lipids, including confirmed deficiency of methoxy-mycolic acids due to a stop-gain mutation in the mmaA3 gene that encodes a methoxy-mycolic acid synthase. We show that the abolished biosynthesis of methoxy-mycolates of L7 alters the cell structure and colony morphology on selected growth media and impacts biofilm formation. The loss of these mycolic acid moieties may change the host-pathogen dynamic for L7 isolates, explaining the limited geographical distribution of L7 and contributing to further understanding the spread of MTBC lineages across the globe.
Collapse
Affiliation(s)
- Elena Hailu
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Daire Cantillon
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Falmer, UK
- Present address: Department of Tropical Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Carlos Madrazo
- Biomedicine Institute of Valencia, Spanish Research Council (IBV-CSIC), Valencia, Spain
| | - Graham Rose
- Francis Crick Institute, London, UK
- Present address: North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children, London, UK
| | | | - Paul Golby
- Animal and Plant Health Agency, Weybridge, UK
| | | | - Sebastien Gagneux
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Stephen V. Gordon
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Iñaki Comas
- Biomedicine Institute of Valencia, Spanish Research Council (IBV-CSIC), Valencia, Spain
| | - Douglas B. Young
- Francis Crick Institute, London, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Simon J. Waddell
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Falmer, UK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Stefan Berg
- Animal and Plant Health Agency, Weybridge, UK
- Present address: Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
7
|
Mouse Models for Mycobacterium tuberculosis Pathogenesis: Show and Do Not Tell. Pathogens 2022; 12:pathogens12010049. [PMID: 36678397 PMCID: PMC9865329 DOI: 10.3390/pathogens12010049] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/29/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022] Open
Abstract
Science has been taking profit from animal models since the first translational experiments back in ancient Greece. From there, and across all history, several remarkable findings have been obtained using animal models. One of the most popular models, especially for research in infectious diseases, is the mouse. Regarding research in tuberculosis, the mouse has provided useful information about host and bacterial traits related to susceptibility to the infection. The effect of aging, sexual dimorphisms, the route of infection, genetic differences between mice lineages and unbalanced immunity scenarios upon Mycobacterium tuberculosis infection and tuberculosis development has helped, helps and will help biomedical researchers in the design of new tools for diagnosis, treatment and prevention of tuberculosis, despite various discrepancies and the lack of deep study in some areas of these traits.
Collapse
|
8
|
Simper JD, Perez E, Schlesinger LS, Azad AK. Resistance and Susceptibility Immune Factors at Play during Mycobacterium tuberculosis Infection of Macrophages. Pathogens 2022; 11:1153. [PMID: 36297211 PMCID: PMC9611686 DOI: 10.3390/pathogens11101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Collapse
Affiliation(s)
- Jan D. Simper
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Esteban Perez
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Translational Sciences Program, UT Health San Antonio Graduate School of Biomedical Sciences, San Antonio, TX 78229, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| | - Abul K. Azad
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| |
Collapse
|
9
|
Rani A, Alam A, Ahmad F, P. M, Saurabh A, Zarin S, Mitra DK, Hasnain SE, Ehtesham NZ. Mycobacterium tuberculosis Methyltransferase Rv1515c Can Suppress Host Defense Mechanisms by Modulating Immune Functions Utilizing a Multipronged Mechanism. Front Mol Biosci 2022; 9:906387. [PMID: 35813825 PMCID: PMC9263924 DOI: 10.3389/fmolb.2022.906387] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb) gene Rv1515c encodes a conserved hypothetical protein exclusively present within organisms of MTB complex and absent in non-pathogenic mycobacteria. In silico analysis revealed that Rv1515c contain S-adenosylmethionine binding site and methyltransferase domain. The DNA binding and DNA methyltransferase activity of Rv1515c was confirmed in vitro. Knock-in of Rv1515c in a model mycobacteria M. smegmatis (M. s_Rv1515c) resulted in remarkable physiological and morphological changes and conferred the recombinant strain with an ability to adapt to various stress conditions, including resistance to TB drugs. M. s_Rv1515c was phagocytosed at a greater rate and displayed extended intra-macrophage survival in vitro. Recombinant M. s_Rv1515c contributed to enhanced virulence by suppressing the host defense mechanisms including RNS and ROS production, and apoptotic clearance. M. s_Rv1515c, while suppressing the phagolysosomal maturation, modulated pro-inflammatory cytokine production and also inhibited antigen presentation by downregulating the expression of MHC-I/MHC-II and co-stimulatory signals CD80 and CD86. Mice infected with M. s_Rv1515c produced more Treg cells than vector control (M. s_Vc) and exhibited reduced effector T cell responses, along-with reduced expression of macrophage activation markers in the chronic phase of infection. M. s_Rv1515c was able to survive in the major organs of mice up to 7 weeks post-infection. These results indicate a crucial role of Rv1515c in M. tb pathogenesis.
Collapse
Affiliation(s)
- Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT-D), New Delhi, India
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Faraz Ahmad
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Manjunath P.
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Abhinav Saurabh
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Sheeba Zarin
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Dipendra Kumar Mitra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Seyed E. Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi (IIT-D), New Delhi, India
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India
- *Correspondence: Seyed E. Hasnain, , , , Nasreen Z. Ehtesham, ,
| | - Nasreen Z. Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
- *Correspondence: Seyed E. Hasnain, , , , Nasreen Z. Ehtesham, ,
| |
Collapse
|
10
|
Dover LG, Thompson AR, Sutcliffe IC, Sangal V. Phylogenomic Reappraisal of Fatty Acid Biosynthesis, Mycolic Acid Biosynthesis and Clinical Relevance Among Members of the Genus Corynebacterium. Front Microbiol 2021; 12:802532. [PMID: 35003033 PMCID: PMC8733736 DOI: 10.3389/fmicb.2021.802532] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
The genus Corynebacterium encompasses many species of biotechnological, medical or veterinary significance. An important characteristic of this genus is the presence of mycolic acids in their cell envelopes, which form the basis of a protective outer membrane (mycomembrane). Mycolic acids in the cell envelope of Mycobacterium tuberculosis have been associated with virulence. In this study, we have analysed the genomes of 140 corynebacterial strains, including representatives of 126 different species. More than 50% of these strains were isolated from clinical material from humans or animals, highlighting the true scale of pathogenic potential within the genus. Phylogenomically, these species are very diverse and have been organised into 19 groups and 30 singleton strains. We find that a substantial number of corynebacteria lack FAS-I, i.e., have no capability for de novo fatty acid biosynthesis and must obtain fatty acids from their habitat; this appears to explain the well-known lipophilic phenotype of some species. In most species, key genes associated with the condensation and maturation of mycolic acids are present, consistent with the reports of mycolic acids in their species descriptions. Conversely, species reported to lack mycolic acids lacked these key genes. Interestingly, Corynebacterium ciconiae, which is reported to lack mycolic acids, appears to possess all genes required for mycolic acid biosynthesis. We suggest that although a mycolic acid-based mycomembrane is widely considered to be the target for interventions by the immune system and chemotherapeutics, the structure is not essential in corynebacteria and is not a prerequisite for pathogenicity or colonisation of animal hosts.
Collapse
|
11
|
Galy R, Ballereau S, Génisson Y, Mourey L, Plaquevent JC, Maveyraud L. Fragment-Based Ligand Discovery Applied to the Mycolic Acid Methyltransferase Hma (MmaA4) from Mycobacterium tuberculosis: A Crystallographic and Molecular Modelling Study. Pharmaceuticals (Basel) 2021; 14:ph14121282. [PMID: 34959681 PMCID: PMC8708032 DOI: 10.3390/ph14121282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 11/16/2022] Open
Abstract
The mycolic acid biosynthetic pathway represents a promising source of pharmacological targets in the fight against tuberculosis. In Mycobacterium tuberculosis, mycolic acids are subject to specific chemical modifications introduced by a set of eight S-adenosylmethionine dependent methyltransferases. Among these, Hma (MmaA4) is responsible for the introduction of oxygenated modifications. Crystallographic screening of a library of fragments allowed the identification of seven ligands of Hma. Two mutually exclusive binding modes were identified, depending on the conformation of residues 147–154. These residues are disordered in apo-Hma but fold upon binding of the S-adenosylmethionine (SAM) cofactor as well as of analogues, resulting in the formation of the short η1-helix. One of the observed conformations would be incompatible with the presence of the cofactor, suggesting that allosteric inhibitors could be designed against Hma. Chimeric compounds were designed by fusing some of the bound fragments, and the relative binding affinities of initial fragments and evolved compounds were investigated using molecular dynamics simulation and generalised Born and Poisson–Boltzmann calculations coupled to the surface area continuum solvation method. Molecular dynamics simulations were also performed on apo-Hma to assess the structural plasticity of the unliganded protein. Our results indicate a significant improvement in the binding properties of the designed compounds, suggesting that they could be further optimised to inhibit Hma activity.
Collapse
Affiliation(s)
- Romain Galy
- Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31077 Toulouse, France; (R.G.); (L.M.)
| | - Stéphanie Ballereau
- Laboratoire de Synthèse et Physico-Chimie de Molécules d’Intérêt Biologique, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31062 Toulouse, France; (S.B.); (Y.G.); (J.-C.P.)
| | - Yves Génisson
- Laboratoire de Synthèse et Physico-Chimie de Molécules d’Intérêt Biologique, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31062 Toulouse, France; (S.B.); (Y.G.); (J.-C.P.)
| | - Lionel Mourey
- Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31077 Toulouse, France; (R.G.); (L.M.)
| | - Jean-Christophe Plaquevent
- Laboratoire de Synthèse et Physico-Chimie de Molécules d’Intérêt Biologique, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31062 Toulouse, France; (S.B.); (Y.G.); (J.-C.P.)
| | - Laurent Maveyraud
- Institut de Pharmacologie et de Biologie Structurale, Université Toulouse III—Paul Sabatier, Centre National de la Recherche Scientifique, 31077 Toulouse, France; (R.G.); (L.M.)
- Correspondence: ; Tel.: +33-561-17-54-35
| |
Collapse
|
12
|
Madacki J, Kopál M, Jackson M, Korduláková J. Mycobacterial Epoxide Hydrolase EphD Is Inhibited by Urea and Thiourea Derivatives. Int J Mol Sci 2021; 22:2884. [PMID: 33809178 PMCID: PMC7998700 DOI: 10.3390/ijms22062884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022] Open
Abstract
The genome of the human intracellular pathogen Mycobacterium tuberculosis encodes an unusually large number of epoxide hydrolases, which are thought to be involved in lipid metabolism and detoxification reactions needed to endure the hostile environment of host macrophages. These enzymes therefore represent suitable targets for compounds such as urea derivatives, which are known inhibitors of soluble epoxide hydrolases. In this work, we studied in vitro the effect of the thiourea drug isoxyl on six epoxide hydrolases of M. tuberculosis using a fatty acid substrate. We show that one of the proteins inhibited by isoxyl is EphD, an enzyme involved in the metabolism of mycolic acids, key components of the mycobacterial cell wall. By analyzing mycolic acid profiles, we demonstrate the inhibition of EphD epoxide hydrolase activity by isoxyl and two other urea-based inhibitors, thiacetazone and AU1235, inside the mycobacterial cell.
Collapse
Affiliation(s)
- Jan Madacki
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská Dolina, Ilkovičova 6, 842 15 Bratislava, Slovakia; (J.M.); (M.K.)
| | - Martin Kopál
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská Dolina, Ilkovičova 6, 842 15 Bratislava, Slovakia; (J.M.); (M.K.)
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA;
| | - Jana Korduláková
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská Dolina, Ilkovičova 6, 842 15 Bratislava, Slovakia; (J.M.); (M.K.)
| |
Collapse
|
13
|
Discovery of a novel dehydratase of the fatty acid synthase type II critical for ketomycolic acid biosynthesis and virulence of Mycobacterium tuberculosis. Sci Rep 2020; 10:2112. [PMID: 32034201 PMCID: PMC7005898 DOI: 10.1038/s41598-020-58967-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
The fatty acid synthase type II (FAS-II) multienzyme system builds the main chain of mycolic acids (MAs), important lipid pathogenicity factors of Mycobacterium tuberculosis (Mtb). Due to their original structure, the identification of the (3 R)-hydroxyacyl-ACP dehydratases, HadAB and HadBC, of Mtb FAS-II complex required in-depth work. Here, we report the discovery of a third dehydratase protein, HadDMtb (Rv0504c), whose gene is non-essential and sits upstream of cmaA2 encoding a cyclopropane synthase dedicated to keto- and methoxy-MAs. HadDMtb deletion triggered a marked change in Mtb keto-MA content and size distribution, deeply impacting the production of full-size molecules. Furthermore, abnormal MAs, likely generated from 3-hydroxylated intermediates, accumulated. These data strongly suggest that HadDMtb catalyzes the 3-hydroxyacyl dehydratation step of late FAS-II elongation cycles during keto-MA biosynthesis. Phenotyping of Mtb hadD deletion mutant revealed the influence of HadDMtb on the planktonic growth, colony morphology and biofilm structuration, as well as on low temperature tolerance. Importantly, HadDMtb has a strong impact on Mtb virulence in the mouse model of infection. The effects of the lack of HadDMtb observed both in vitro and in vivo designate this protein as a bona fide target for the development of novel anti-TB intervention strategies.
Collapse
|
14
|
Guerra-De-Blas PDC, Bobadilla-Del-Valle M, Sada-Ovalle I, Estrada-García I, Torres-González P, López-Saavedra A, Guzmán-Beltrán S, Ponce-de-León A, Sifuentes-Osornio J. Simvastatin Enhances the Immune Response Against Mycobacterium tuberculosis. Front Microbiol 2019; 10:2097. [PMID: 31616387 PMCID: PMC6764081 DOI: 10.3389/fmicb.2019.02097] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/26/2019] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis remains a serious threat worldwide. For this reason, it is necessary to identify agents that shorten the duration of treatment, strengthen the host immune system, and/or decrease the damage caused by the infection. Statins are drugs that reduce plasma cholesterol levels and have immunomodulatory, anti-inflammatory and antimicrobial effects. Although there is evidence that statins may contribute to the containment of Mycobacterium tuberculosis infection, their effects on peripheral blood mononuclear cells (PBMCs) involved in the immune response have not been previously described. Using PBMCs from 10 healthy subjects infected with M. tuberculosis H37Rv, we analyzed the effects of simvastatin on the treatment of the infections in an in vitro experimental model. Direct quantification of M. tuberculosis growth (in CFU/mL) was performed. Phenotypes and cell activation were assessed via multi-color flow cytometry. Culture supernatant cytokine levels were determined via cytokine bead arrays. The induction of apoptosis and autophagy was evaluated via flow cytometry and confocal microscopy. Simvastatin decreased the growth of M. tuberculosis in PBMCs, increased the proportion of NKT cells in culture, increased the expression of co-stimulatory molecules in monocytes, promoted the secretion of the cytokines IL-1β and IL-12p70, and activated apoptosis and autophagy in monocytes, resulting in a significant reduction in bacterial load. We also observed an increase in IL-10 production. We did not observe any direct antimycobacterial activity. This study provides new insight into the mechanism through which simvastatin reduces the mycobacterial load in infected PBMCs. These results demonstrate that simvastatin activates several immune mechanisms that favor the containment of M. tuberculosis infection, providing relevant evidence to consider statins as candidates for host-directed therapy. They also suggest that future studies are needed to define the roles of statin-induced anti-inflammatory mechanisms in tuberculosis treatment.
Collapse
Affiliation(s)
- Paola Del Carmen Guerra-De-Blas
- Laboratorio de Microbiología Clínica, Departamento de Infectología, Dirección de Medicina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miriam Bobadilla-Del-Valle
- Laboratorio de Microbiología Clínica, Departamento de Infectología, Dirección de Medicina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isabel Sada-Ovalle
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Iris Estrada-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Pedro Torres-González
- Laboratorio de Microbiología Clínica, Departamento de Infectología, Dirección de Medicina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alejandro López-Saavedra
- Unidad Biomédica de Investigación en Cáncer, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Silvia Guzmán-Beltrán
- Laboratorio de Inmunología Integrativa, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfredo Ponce-de-León
- Laboratorio de Microbiología Clínica, Departamento de Infectología, Dirección de Medicina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Sifuentes-Osornio
- Laboratorio de Microbiología Clínica, Departamento de Infectología, Dirección de Medicina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
15
|
Madacki J, Mas Fiol G, Brosch R. Update on the virulence factors of the obligate pathogen Mycobacterium tuberculosis and related tuberculosis-causing mycobacteria. INFECTION GENETICS AND EVOLUTION 2019; 72:67-77. [DOI: 10.1016/j.meegid.2018.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/02/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022]
|
16
|
Okay S, Çetin R, Karabulut F, Doğan C, Sürücüoğlu S, Kızıldoğan AK. Immune responses elicited by the recombinant Erp, HspR, LppX, MmaA4, and OmpA proteins from Mycobacterium tuberculosis in mice. Acta Microbiol Immunol Hung 2019; 66:219-234. [PMID: 30484328 DOI: 10.1556/030.65.2018.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunogenic potency of the recombinant Erp, HspR, LppX, MmaA4, and OmpA proteins from Mycobacterium tuberculosis (MTB), formulated with Montanide ISA 720 VG adjuvant, was evaluated in BALB/c mice for the first time in this study. The five vaccine formulations, adjuvant, and BCG vaccine were subcutaneously injected into mice, and the sera were collected at days 0, 15, 30, 41, and 66. The humoral and cellular immune responses against vaccine formulations were determined by measuring serum IgG and serum interferon-gamma (IFN-γ) and interleukin-12 (IL-12) levels, respectively. All formulations significantly increased IgG levels post-vaccination. The highest increase in IFN-γ level was provided by MmaA4 formulation. The Erp, HspR, and LppX formulations were as effective as BCG in enhancement of IFN-γ level. The most efficient vaccine boosting the IL-12 level was HspR formulation, especially at day 66. Erp formulation also increased the IL-12 level more than BCG at days 15 and 30. The IL-12 level boosted by MmaA4 formulation was found to be similar to that by BCG. OmpA formulation was inefficient in enhancement of cellular immune responses. This study showed that MmaA4, HspR, and Erp proteins from MTB are successful in eliciting both humoral and cellular immune responses in mice.
Collapse
Affiliation(s)
- Sezer Okay
- 1 Faculty of Science, Department of Biology, Çankiri Karatekin University, Çankiri, Turkey
| | - Rukiye Çetin
- 1 Faculty of Science, Department of Biology, Çankiri Karatekin University, Çankiri, Turkey
| | - Fatih Karabulut
- 1 Faculty of Science, Department of Biology, Çankiri Karatekin University, Çankiri, Turkey
| | - Cennet Doğan
- 1 Faculty of Science, Department of Biology, Çankiri Karatekin University, Çankiri, Turkey
| | - Süheyla Sürücüoğlu
- 2 Faculty of Medicine, Department of Medical Microbiology, Manisa Celal Bayar University, Manisa, Turkey
| | - Aslıhan Kurt Kızıldoğan
- 3 Faculty of Agriculture, Department of Agricultural Biotechnology, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
17
|
Saelens JW, Viswanathan G, Tobin DM. Mycobacterial Evolution Intersects With Host Tolerance. Front Immunol 2019; 10:528. [PMID: 30967867 PMCID: PMC6438904 DOI: 10.3389/fimmu.2019.00528] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past 200 years, tuberculosis (TB) has caused more deaths than any other infectious disease, likely infecting more people than it has at any other time in human history. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, is an obligate human pathogen that has evolved through the millennia to become an archetypal human-adapted pathogen. This review focuses on the evolutionary framework by which Mtb emerged as a specialized human pathogen and applies this perspective to the emergence of specific lineages that drive global TB burden. We consider how evolutionary pressures, including transmission dynamics, host tolerance, and human population patterns, may have shaped the evolution of diverse mycobacterial genomes.
Collapse
Affiliation(s)
- Joseph W. Saelens
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
18
|
Guerra-De-Blas PDC, Torres-González P, Bobadilla-Del-Valle M, Sada-Ovalle I, Ponce-De-León-Garduño A, Sifuentes-Osornio J. Potential Effect of Statins on Mycobacterium tuberculosis Infection. J Immunol Res 2018; 2018:7617023. [PMID: 30581876 PMCID: PMC6276473 DOI: 10.1155/2018/7617023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/11/2018] [Accepted: 10/23/2018] [Indexed: 02/03/2023] Open
Abstract
Tuberculosis is one of the 10 leading causes of death in the world. The current treatment is based on a combination of antimicrobials administered for six months. It is essential to find therapeutic agents with which the treatment time can be shortened and strengthen the host immune response against Mycobacterium tuberculosis. M. tuberculosis needs cholesterol to infect and survive inside the host, but the progression of the infection depends to a large extent on the capacity of the immune response to contain the infection. Statins inhibit the synthesis of cholesterol and have pleiotropic effects on the immune system, which have been associated with better results in the treatment of several infectious diseases. Recently, it has been reported that cells treated with statins are more resistant to M. tuberculosis infection, and they have even been proposed as adjuvants in the treatment of M. tuberculosis infection. The aim of this review is to summarize the immunopathogenesis of tuberculosis and its mechanisms of evasion and to compile the available scientific information on the effect of statins in the treatment of tuberculosis.
Collapse
Affiliation(s)
- Paola Del Carmen Guerra-De-Blas
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Pedro Torres-González
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miriam Bobadilla-Del-Valle
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isabel Sada-Ovalle
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfredo Ponce-De-León-Garduño
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Sifuentes-Osornio
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
19
|
Baron S, Peleg Y, Grunwald J, Morgenstern D, Elad N, Peretz M, Albeck S, Levin Y, Welch JT, DeWeerd KA, Schwarz A, Burstein Y, Diskin R, Shakked Z, Zimhony O. Expression of a recombinant, 4'-Phosphopantetheinylated, active M. tuberculosis fatty acid synthase I in E. coli. PLoS One 2018; 13:e0204457. [PMID: 30248156 PMCID: PMC6152951 DOI: 10.1371/journal.pone.0204457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/07/2018] [Indexed: 11/18/2022] Open
Abstract
Background Fatty acid synthase 1 (FAS I) from Mycobacterium tuberculosis (Mtb) is an essential protein and a promising drug target. FAS I is a multi-functional, multi-domain protein that is organized as a large (1.9 MDa) homohexameric complex. Acyl intermediates produced during fatty acid elongation are attached covalently to an acyl carrier protein (ACP) domain. This domain is activated by the transfer of a 4'-Phosphopantetheine (4'-PP, also termed P-pant) group from CoA to ACP catalyzed by a 4'-PP transferase, termed acyl carrier protein synthase (AcpS). Methods In order to obtain an activated FAS I in E. coli, we transformed E. coli with tagged Mtb fas1 and acpS genes encoded by a separate plasmid. We induced the expression of Mtb FAS I following induction of AcpS expression. FAS I was purified by Strep-Tactin affinity chromatography. Results Activation of Mtb FAS I was confirmed by the identification of a bound P-pant group on serine at position 1808 by mass spectrometry. The purified FAS I displayed biochemical activity shown by spectrophotometric analysis of NADPH oxidation and by CoA production, using the Ellman reaction. The purified Mtb FAS I forms a hexameric complex shown by negative staining and cryo-EM. Conclusion Purified hexameric and active Mtb FAS I is required for binding and drug inhibition studies and for structure-function analysis of this enzyme. This relatively simple and short procedure for Mtb FAS I production should facilitate studies of this enzyme.
Collapse
Affiliation(s)
- Szilvia Baron
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoav Peleg
- Structural Proteomics Unit (SPU), Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Grunwald
- Structural Proteomics Unit (SPU), Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Elad
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Peretz
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Albeck
- Structural Proteomics Unit (SPU), Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- De Botton Institute for Protein Profiling, Weizmann Institute of Science, Rehovot, Israel
| | - John T. Welch
- Department of Chemistry, College of Arts and Sciences University at Albany, New York, United States of America
| | - Kim A. DeWeerd
- Molecular Core Facility College of Arts and Sciences University at Albany, New York, United States of America
| | - Alon Schwarz
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yigal Burstein
- Department of Organic Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Diskin
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Zippora Shakked
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Zimhony
- Kaplan Medical Center, Rehovot, affiliated to the School of Medicine, Hebrew University and Hadassah, Jerusalem, Israel
- * E-mail: ,
| |
Collapse
|
20
|
Effect of Homocysteine on Biofilm Formation by Mycobacteria. Indian J Microbiol 2018; 58:287-293. [PMID: 30013272 DOI: 10.1007/s12088-018-0739-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/03/2018] [Indexed: 12/29/2022] Open
Abstract
Mycobacteria show peculiar aggregated outgrowth like biofilm on the surface of solid or liquid media. Biofilms harbor antibiotic resistant bacteria in a self-produced extracellular matrix that signifies the bacterial fate to sedentary existence. Despite years of research, very little is known about the mechanisms that contribute to biofilm formation. LuxS has been previously known to play a role in biofilm formation in Autoinducer-2 dependent manner. We here show the effect of LuxS product-homocysteine, on the biofilm forming ability of non-tuberculous mycobacteria, Mycobacterium smegmatis and Mycobacterium bovis BCG showing AI-2 independent phenotypic effect of LuxS. Exogenous supplementation of homocysteine in the culture media leads to aberrant cording, pellicle outgrowth, and biofilm formation. Thus, our study contributes to the better understanding of the mechanism of mycobacterial biofilm formation and sheds light on the role of LuxS product homocysteine. In addition, we highlight the contribution of activated methyl cycle in bacterial quorum sensing.
Collapse
|
21
|
Williams SJ. Sensing Lipids with Mincle: Structure and Function. Front Immunol 2017; 8:1662. [PMID: 29230225 PMCID: PMC5711825 DOI: 10.3389/fimmu.2017.01662] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/13/2017] [Indexed: 01/06/2023] Open
Abstract
Mincle is a C-type lectin receptor that has emerged as an important player in innate immunity through its capacity to recognize a wide range of lipidic species derived from damaged/altered self and foreign microorganisms. Self-ligands include sterols (e.g., cholesterol), and β-glucosylceramides, and the protein SAP130, which is released upon cell death. Foreign lipids comprise those from both microbial pathogens and commensals and include glycerol, glucose and trehalose mycolates, and glycosyl diglycerides. A large effort has focused on structural variation of these ligands to illuminate the structure–activity relationships required for the agonism of signaling though Mincle and has helped identify key differences in ligand recognition between human and rodent Mincle. These studies in turn have helped identify new Mincle ligands, further broadening our understanding of the diversity of organisms and lipidic species recognized by Mincle. Finally, progress toward the development of Mincle agonists as vaccine adjuvants providing humoral and cell-mediated immunity with reduced toxicity is discussed.
Collapse
Affiliation(s)
- Spencer J Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Awuh JA, Flo TH. Molecular basis of mycobacterial survival in macrophages. Cell Mol Life Sci 2017; 74:1625-1648. [PMID: 27866220 PMCID: PMC11107535 DOI: 10.1007/s00018-016-2422-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/06/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Abstract
Macrophages play an essential role in the immune system by ingesting and degrading invading pathogens, initiating an inflammatory response and instructing adaptive immune cells, and resolving inflammation to restore homeostasis. More interesting is the fact that some bacteria have evolved to use macrophages as a natural habitat and tools of spread in the host, e.g., Mycobacterium tuberculosis (Mtb) and some non-tuberculous mycobacteria (NTM). Mtb is considered one of humanity's most successful pathogens and is the causal agent of tuberculosis, while NTMs cause opportunistic infections all of which are of significant public health concern. Here, we describe mechanisms by which intracellular pathogens, with an emphasis on mycobacteria, manipulate macrophage functions to circumvent killing and live inside these cells even under considerable immunological pressure. Such macrophage functions include the selective evasion or engagement of pattern recognition receptors, production of cytokines, reactive oxygen and nitrogen species, phagosome maturation, as well as other killing mechanisms like autophagy and cell death. A clear understanding of host responses elicited by a specific pathogen and strategies employed by the microbe to evade or exploit these is of significant importance for the development of effective vaccines and targeted immunotherapy against persistent intracellular infections like tuberculosis.
Collapse
Affiliation(s)
- Jane Atesoh Awuh
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, PB 8905, 7491, Trondheim, Norway
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, PB 8905, 7491, Trondheim, Norway.
| |
Collapse
|
23
|
Fineran P, Lloyd-Evans E, Lack NA, Platt N, Davis LC, Morgan AJ, Höglinger D, Tatituri RVV, Clark S, Williams IM, Tynan P, Al Eisa N, Nazarova E, Williams A, Galione A, Ory DS, Besra GS, Russell DG, Brenner MB, Sim E, Platt FM. Pathogenic mycobacteria achieve cellular persistence by inhibiting the Niemann-Pick Type C disease cellular pathway. Wellcome Open Res 2016. [PMID: 28008422 DOI: 10.12688/wellcomeopenres.10036.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Tuberculosis remains a major global health concern. The ability to prevent phagosome-lysosome fusion is a key mechanism by which intracellular mycobacteria, including Mycobacterium tuberculosis, achieve long-term persistence within host cells. The mechanisms underpinning this key intracellular pro-survival strategy remain incompletely understood. Host macrophages infected with persistent mycobacteria share phenotypic similarities with cells taken from patients suffering from Niemann-Pick Disease Type C (NPC), a rare lysosomal storage disease in which endocytic trafficking defects and lipid accumulation within the lysosome lead to cell dysfunction and cell death. We investigated whether these shared phenotypes reflected an underlying mechanistic connection between mycobacterial intracellular persistence and the host cell pathway dysfunctional in NPC. METHODS The induction of NPC phenotypes in macrophages from wild-type mice or obtained from healthy human donors was assessed via infection with mycobacteria and subsequent measurement of lipid levels and intracellular calcium homeostasis. The effect of NPC therapeutics on intracellular mycobacterial load was also assessed. RESULTS Macrophages infected with persistent intracellular mycobacteria phenocopied NPC cells, exhibiting accumulation of multiple lipid types, reduced lysosomal Ca2+ levels, and defects in intracellular trafficking. These NPC phenotypes could also be induced using only lipids/glycomycolates from the mycobacterial cell wall. These data suggest that persistent intracellular mycobacteria inhibit the NPC pathway, likely via inhibition of the NPC1 protein, and subsequently induce altered acidic store Ca2+ homeostasis. Reduced lysosomal calcium levels may provide a mechanistic explanation for the reduced levels of phagosome-lysosome fusion in mycobacterial infection. Treatments capable of correcting defects in NPC mutant cells via modulation of host cell calcium were of benefit in promoting clearance of mycobacteria from infected host cells. CONCLUSION These findings provide a novel mechanistic explanation for mycobacterial intracellular persistence, and suggest that targeting interactions between the mycobacteria and host cell pathways may provide a novel avenue for development of anti-TB therapies.
Collapse
Affiliation(s)
- Paul Fineran
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Emyr Lloyd-Evans
- Department of Pharmacology, University of Oxford, Oxford, UK.,School of Biosciences, Cardiff University, Cardiff, UK
| | - Nathan A Lack
- Department of Pharmacology, University of Oxford, Oxford, UK.,School of Medicine, Koç University, Istanbul, Turkey
| | - Nick Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lianne C Davis
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Doris Höglinger
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | | | - Ian M Williams
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Patricia Tynan
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nada Al Eisa
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Evgeniya Nazarova
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | | | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, USA
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Michael B Brenner
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Edith Sim
- Department of Pharmacology, University of Oxford, Oxford, UK.,Faculty of Science Engineering and Computing, Kingston University, Kingston upon Thames, UK
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Tima HG, Al Dulayymi JR, Denis O, Lehebel P, Baols KS, Mohammed MO, L'Homme L, Sahb MM, Potemberg G, Legrand S, Lang R, Beyaert R, Piette J, Baird MS, Huygen K, Romano M. Inflammatory Properties and Adjuvant Potential of Synthetic Glycolipids Homologous to Mycolate Esters of the Cell Wall of Mycobacterium tuberculosis. J Innate Immun 2016; 9:162-180. [PMID: 27855374 DOI: 10.1159/000450955] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 09/21/2016] [Indexed: 12/30/2022] Open
Abstract
The cell wall of mycobacteria is characterised by glycolipids composed of different classes of mycolic acids (MAs; alpha-, keto-, and methoxy-) and sugars (trehalose, glucose, and arabinose). Studies using mutant Mtb strains have shown that the structure of MAs influences the inflammatory potential of these glycolipids. As mutant Mtb strains possess a complex mixture of glycolipids, we analysed the inflammatory potential of single classes of mycolate esters of the Mtb cell wall using 38 different synthetic analogues. Our results show that synthetic trehalose dimycolate (TDM) and trehalose, glucose, and arabinose monomycolates (TMM, GMM, and AraMM) activate bone marrow-derived dendritic cells in terms of the production of pro-inflammatory cytokines (IL-6 and TNF-α) and reactive oxygen species, upregulation of costimulatory molecules, and activation of NLRP3 inflammasome by a mechanism dependent on Mincle. These findings demonstrate that Mincle receptor can also recognise pentose esters and seem to contradict the hypothesis that production of GMM is an escape mechanism used by pathogenic mycobacteria to avoid recognition by the innate immune system. Finally, our experiments indicate that TMM and GMM, as well as TDM, can promote Th1 and Th17 responses in mice in an OVA immunisation model, and that further analysis of their potential as novel adjuvants for subunit vaccines is warranted.
Collapse
Affiliation(s)
- Hermann Giresse Tima
- Scientific Service Immunology, O.D. Communicable and Infectious Diseases, Scientific Institute of Public Health (WIV-ISP), Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fineran P, Lloyd-Evans E, Lack NA, Platt N, Davis LC, Morgan AJ, Höglinger D, Tatituri RVV, Clark S, Williams IM, Tynan P, Al Eisa N, Nazarova E, Williams A, Galione A, Ory DS, Besra GS, Russell DG, Brenner MB, Sim E, Platt FM. Pathogenic mycobacteria achieve cellular persistence by inhibiting the Niemann-Pick Type C disease cellular pathway. Wellcome Open Res 2016; 1:18. [PMID: 28008422 PMCID: PMC5172425 DOI: 10.12688/wellcomeopenres.10036.2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tuberculosis remains a major global health concern. The ability to prevent phagosome-lysosome fusion is a key mechanism by which intracellular mycobacteria, including Mycobacterium tuberculosis, achieve long-term persistence within host cells. The mechanisms underpinning this key intracellular pro-survival strategy remain incompletely understood. Host macrophages infected with persistent mycobacteria share phenotypic similarities with cells taken from patients suffering from Niemann-Pick Disease Type C (NPC), a rare lysosomal storage disease in which endocytic trafficking defects and lipid accumulation within the lysosome lead to cell dysfunction and cell death. We investigated whether these shared phenotypes reflected an underlying mechanistic connection between mycobacterial intracellular persistence and the host cell pathway dysfunctional in NPC. METHODS The induction of NPC phenotypes in macrophages from wild-type mice or obtained from healthy human donors was assessed via infection with mycobacteria and subsequent measurement of lipid levels and intracellular calcium homeostasis. The effect of NPC therapeutics on intracellular mycobacterial load was also assessed. RESULTS Macrophages infected with persistent intracellular mycobacteria phenocopied NPC cells, exhibiting accumulation of multiple lipid types, reduced lysosomal Ca2+ levels, and defects in intracellular trafficking. These NPC phenotypes could also be induced using only lipids/glycomycolates from the mycobacterial cell wall. These data suggest that persistent intracellular mycobacteria inhibit the NPC pathway, likely via inhibition of the NPC1 protein, and subsequently induce altered acidic store Ca2+ homeostasis. Reduced lysosomal calcium levels may provide a mechanistic explanation for the reduced levels of phagosome-lysosome fusion in mycobacterial infection. Treatments capable of correcting defects in NPC mutant cells via modulation of host cell calcium were of benefit in promoting clearance of mycobacteria from infected host cells. CONCLUSION These findings provide a novel mechanistic explanation for mycobacterial intracellular persistence, and suggest that targeting interactions between the mycobacteria and host cell pathways may provide a novel avenue for development of anti-TB therapies.
Collapse
Affiliation(s)
- Paul Fineran
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Emyr Lloyd-Evans
- Department of Pharmacology, University of Oxford, Oxford, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Nathan A. Lack
- Department of Pharmacology, University of Oxford, Oxford, UK
- School of Medicine, Koç University, Istanbul, Turkey
| | - Nick Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lianne C. Davis
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Doris Höglinger
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | | | - Ian M. Williams
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Patricia Tynan
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nada Al Eisa
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Evgeniya Nazarova
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | | | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Daniel S. Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, USA
| | | | - David G. Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Michael B. Brenner
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Edith Sim
- Department of Pharmacology, University of Oxford, Oxford, UK
- Faculty of Science Engineering and Computing, Kingston University, Kingston upon Thames, UK
| | | |
Collapse
|
26
|
Maggioli MF, Palmer MV, Thacker TC, Vordermeier HM, McGill JL, Whelan AO, Larsen MH, Jacobs WR, Waters WR. Increased TNF-α/IFN-γ/IL-2 and Decreased TNF-α/IFN-γ Production by Central Memory T Cells Are Associated with Protective Responses against Bovine Tuberculosis Following BCG Vaccination. Front Immunol 2016; 7:421. [PMID: 27799930 PMCID: PMC5066095 DOI: 10.3389/fimmu.2016.00421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/27/2016] [Indexed: 12/30/2022] Open
Abstract
Central memory T cell (Tcm) and polyfunctional CD4 T cell responses contribute to vaccine-elicited protection with both human and bovine tuberculosis (TB); however, their combined role in protective immunity to TB is unclear. To address this question, we evaluated polyfunctional cytokine responses by CD4 T cell effector/memory populations from bacille Calmette–Guerin (BCG) vaccinated and non-vaccinated calves by flow cytometry prior to and after aerosol challenge with virulent Mycobacterium bovis. Polyfunctional cytokine expression patterns in the response by Tcm, effector memory, and effector T cell subsets were similar between BCG-vaccinated and M. bovis-infected calves, only differing in magnitude (i.e., infected > vaccinated). BCG vaccination, however, did alter the kinetics of the ensuing response to virulent M. bovis infection. Early after challenge (3 weeks post-infection), non-vaccinates had greater antigen-specific interferon-γ (IFN-γ)/tumor necrosis factor-α (TNF-α) and lesser IFN-γ/TNF-α/IL-2 responses by Tcm cells than did vaccinated animals. Importantly, these differences were also associated with mycobacterial burden upon necropsy. Polyfunctional responses to ESAT-6:CFP10 (antigens not synthesized by BCG strains) were detected in memory subsets, as well as in effector cells, as early as 3 weeks after challenge. These findings suggest that cell fate divergence may occur early after antigen priming in the response to bovine TB and that memory and effector T cells may expand concurrently during the initial phase of the immune response. In summary, robust IFN-γ/TNF-α response by Tcm cells is associated with greater mycobacterial burden, while IFN-γ/TNF-α/IL-2 response by Tcm cells are indicative of a protective response to bovine TB.
Collapse
Affiliation(s)
- Mayara F Maggioli
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Ames, IA, USA; Imbio, Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Mitchell V Palmer
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| | - Tyler C Thacker
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| | | | - Jodi L McGill
- Department of Diagnostic Medicine and Pathology, College of Veterinary Medicine, Kansas State University , Manhattan, KS , USA
| | - Adam O Whelan
- Defense Science and Technology Laboratory, Porton Down , Wiltshire , UK
| | - Michelle H Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine , Bronx, NY , USA
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine , Bronx, NY , USA
| | - W Ray Waters
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| |
Collapse
|
27
|
Pedroza-Roldán C, Guapillo C, Barrios-Payán J, Mata-Espinosa D, Aceves-Sánchez MDJ, Marquina-Castillo B, Hernández-Pando R, Flores-Valdez MA. The BCGΔBCG1419c strain, which produces more pellicle in vitro, improves control of chronic tuberculosis in vivo. Vaccine 2016; 34:4763-70. [PMID: 27546876 DOI: 10.1016/j.vaccine.2016.08.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/26/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023]
Abstract
Mycobacterium tuberculosis (Mtb) has been a threat to humans since ancient times, and it is the main causative agent of tuberculosis (TB). Until today, the only licensed vaccine against Mtb is the live attenuated M. bovis Bacillus Calmette-Guérin (BCG), which has variable levels of protection against the pulmonary form of infection. The quest for a new vaccine is a priority given the rise of multidrug-resistant Mtb around the world, as well as the tremendous burden imposed by latent TB. The objective of this study was to evaluate the immunogenicity and capacity of protection of a modified BCG strain (BCGΔBCG1419c) lacking the c-di-GMP phosphodiesterase gene BCG1419c, in diverse mice models. In a previous report, we have shown that BCGΔBCG1419c was capable of increasing biofilm production and after intravenous infection of immunocompetent mice; this strain persisted longer in lungs than parental BCG Pasteur. This led us to hypothesize that BCGΔBCG1419c might therefore possess some advantage as vaccine candidate. Our results in this report indicate that compared to conventional BCG, vaccination with BCGΔBCG1419c induced a better activation of specific T-lymphocytes population, was equally effective in preventing weight loss despite being used at lower dose, reduced tissue damage (pneumonic scores), increased local IFNγ(+) T cells, and diminished bacterial burden in lungs of BALB/c mice infected intratracheally with high dose Mtb H37Rv to induce progressive TB. Moreover, vaccination with BCGΔBCG1419c improved resistance to reactivation after immunosuppression induced by corticosterone in a murine model of chronic infection similar to latent TB. Furthermore, despite showing increased persistence in immunocompetent mice, BCGΔBCG1419c was as attenuated as parental BCG in nude mice. To our knowledge, this is the first demonstration that a modified BCG vaccine candidate with increased pellicle/biofilm production has the capacity to protect against Mtb challenge in chronic and reactivation models of infection.
Collapse
Affiliation(s)
- César Pedroza-Roldán
- Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y diseño del Estado de Jalisco, A.C., Av. Normalistas No. 800, Col. Colinas de la Normal, 44270 Guadalajara, Jalisco, Mexico
| | - Carolina Guapillo
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Tlalpan, México, D.F. 14000, Mexico
| | - Jorge Barrios-Payán
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Tlalpan, México, D.F. 14000, Mexico
| | - Dulce Mata-Espinosa
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Tlalpan, México, D.F. 14000, Mexico
| | - Michel de Jesús Aceves-Sánchez
- Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y diseño del Estado de Jalisco, A.C., Av. Normalistas No. 800, Col. Colinas de la Normal, 44270 Guadalajara, Jalisco, Mexico
| | - Brenda Marquina-Castillo
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Tlalpan, México, D.F. 14000, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Tlalpan, México, D.F. 14000, Mexico.
| | - Mario Alberto Flores-Valdez
- Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y diseño del Estado de Jalisco, A.C., Av. Normalistas No. 800, Col. Colinas de la Normal, 44270 Guadalajara, Jalisco, Mexico.
| |
Collapse
|
28
|
F420H2 Is Required for Phthiocerol Dimycocerosate Synthesis in Mycobacteria. J Bacteriol 2016; 198:2020-8. [PMID: 27185825 DOI: 10.1128/jb.01035-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/06/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Phthiocerol dimycocerosates (PDIM) are a group of cell surface-associated apolar lipids of Mycobacterium tuberculosis and closely related mycobacteria, such as Mycobacterium bovis and Mycobacterium leprae A characteristic methoxy group of these lipids is generated from the methylation of a hydroxyl group of the direct precursors, the phthiotriols. The precursors arise from the reduction of phthiodiolones, the keto intermediates, by a ketoreductase. The putative phthiodiolone ketoreductase (PKR) is encoded by Rv2951c in M. tuberculosis and BCG_2972c in M. bovis BCG, and these open reading frames (ORFs) encode identical amino acid sequences. We investigated the cofactor requirement of the BCG_2972c protein. A comparative analysis based on the crystallographic structures of similar enzymes identified structural elements for binding of coenzyme F420 and hydrophobic phthiodiolones in PKR. Coenzyme F420 is a deazaflavin coenzyme that serves several key functions in pathogenic and nonpathogenic mycobacteria. We found that an M. bovis BCG mutant lacking F420-dependent glucose-6-phosphate dehydrogenase (Fgd), which generates F420H2 (glucose-6-phosphate + F420 → 6-phosphogluconate + F420H2), was devoid of phthiocerols and accumulated phthiodiolones. When the mutant was provided with F420H2, a broken-cell slurry of the mutant converted accumulated phthiodiolones to phthiocerols; F420H2 was generated in situ from F420 and glucose-6-phosphate by the action of Fgd. Thus, the reaction mixture was competent in reducing phthiodiolones to phthiotriols (phthiodiolones + F420H2 → phthiotriols + F420), which were then methylated to phthiocerols. These results established the mycobacterial phthiodiolone ketoreductase as an F420H2-dependent enzyme (fPKR). A phylogenetic analysis of close homologs of fPKR revealed potential F420-dependent lipid-modifying enzymes in a broad range of mycobacteria. IMPORTANCE Mycobacterium tuberculosis is the causative agent of tuberculosis, and phthiocerol dimycocerosates (PDIM) protect this pathogen from the early innate immune response of an infected host. Thus, the PDIM synthesis system is a potential target for the development of effective treatments for tuberculosis. The current study shows that a PDIM synthesis enzyme is dependent on the coenzyme F420 F420 is universally present in mycobacteria and absent in humans. This finding expands the number of experimentally validated F420-dependent enzymes in M. tuberculosis to six, each of which helps the pathogen to evade killing by the host immune system, and one of which activates an antituberculosis drug, PA-824. This work also has relevance to leprosy, since similar waxy lipids are found in Mycobacterium leprae.
Collapse
|
29
|
Induction of Unconventional T Cells by a Mutant Mycobacterium bovis BCG Strain Formulated in Cationic Liposomes Correlates with Protection against Mycobacterium tuberculosis Infections of Immunocompromised Mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:638-47. [PMID: 27226281 DOI: 10.1128/cvi.00232-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/18/2016] [Indexed: 02/08/2023]
Abstract
Earlier studies aimed at defining protective immunity induced by Mycobacterium bovis BCG immunization have largely focused on the induction of antituberculosis CD4(+) and CD8(+) T cell responses. Here we describe a vaccine consisting of a BCGΔmmaA4 deletion mutant formulated in dimethyl dioctadecyl-ammonium bromide (DDA) with d-(+)-trehalose 6,6'-dibehenate (TDB) (DDA/TDB) adjuvant (A4/Adj) that protected TCRδ(-/-) mice depleted of CD4(+), CD8(+), and NK1.1(+) T cells against an aerosol challenge with M. tuberculosis These mice were significantly protected relative to mice immunized with a nonadjuvanted BCGΔmmaA4 (BCG-A4) mutant and nonvaccinated controls at 2 months and 9 months postvaccination. In the absence of all T cells following treatment with anti-Thy1.2 antibody, the immunized mice lost the ability to control the infection. These results indicate that an unconventional T cell population was mediating protection in the absence of CD4(+), CD8(+), NK1.1(+), and TCRγδ T cells and could exhibit memory. Focusing on CD4(-) CD8(-) double-negative (DN) T cells, we found that these cells accumulated in the lungs postchallenge significantly more in A4/Adj-immunized mice and induced significantly greater frequencies of pulmonary gamma interferon (IFN-γ)-producing cells than were seen in the nonvaccinated or nonadjuvanted BCG control groups. Moreover, pulmonary DN T cells from the A4/Adj group exhibited significantly higher IFN-γ integrated median fluorescence intensity (iMFI) values than were seen in the control groups. We also showed that enriched DN T cells from mice immunized with A4/Adj could control mycobacterial growth in vitro significantly better than naive whole-spleen cells. These results suggest that formulating BCG in DDA/TDB adjuvant confers superior protection in immunocompromised mice and likely involves the induction of long-lived memory DN T cells.
Collapse
|
30
|
Targeting Mycobacterium tuberculosis Tumor Necrosis Factor Alpha-Downregulating Genes for the Development of Antituberculous Vaccines. mBio 2016; 7:mBio.01023-15. [PMID: 27247233 PMCID: PMC4895118 DOI: 10.1128/mbio.01023-15] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNF) plays a critical role in the control of Mycobacterium tuberculosis, in part by augmenting T cell responses through promoting macrophage phagolysosomal fusion (thereby optimizing CD4+ T cell immunity by enhancing antigen presentation) and apoptosis (a process that can lead to cross-priming of CD8+ T cells). M. tuberculosis can evade antituberculosis (anti-TB) immunity by inhibiting host cell TNF production via expression of specific mycobacterial components. We hypothesized that M. tuberculosis mutants with an increased capacity to induce host cell TNF production (TNF-enhancing mutants) and thus with enhanced immunogenicity can be useful for vaccine development. To identify mycobacterial genes that regulate host cell TNF production, we used a TNF reporter macrophage clone to screen an H37Rv M. tuberculosis cosmid library constructed in M. smegmatis. The screen has identified a set of TNF-downregulating mycobacterial genes that, when deleted in H37Rv, generate TNF-enhancing mutants. Analysis of mutants disrupted for a subset of TNF-downregulating genes, annotated to code for triacylglycerol synthases and fatty acyl-coenzyme A (acyl-CoA) synthetase, enzymes that concern lipid biosynthesis and metabolism, has revealed that these strains can promote macrophage phagolysosomal fusion and apoptosis better than wild-type (WT) bacilli. Immunization of mice with the TNF-enhancing M. tuberculosis mutants elicits CD4+ and CD8+ T cell responses that are superior to those engendered by WT H37Rv. The results suggest that TNF-upregulating M. tuberculosis genes can be targeted to enhance the immunogenicity of mycobacterial strains that can serve as the substrates for the development of novel anti-TB vaccines. One way to control tuberculosis (TB), which remains a major global public health burden, is by immunization with an effective vaccine. The efficacy of Mycobacterium bovis BCG, the only currently approved TB vaccine, is inconsistent. Tumor necrosis factor alpha (TNF) is a cytokine that plays an important role in controlling TB. M. tuberculosis, the causative agent of TB, can counter this TNF-based defense by decreasing host cell TNF production. This study identified M. tuberculosis genes that can mediate inhibition of TNF production by macrophage (an immune cell critical to the control of TB). We have knocked out a number of these genes to generate M. tuberculosis mutants that can enhance macrophage TNF production. Immunization with these mutants in mice triggered a T cell response stronger than that elicited by the parental bacillus. Since T cell immunity is pivotal in controlling M. tuberculosis, the TNF-enhancing mutants can be used to develop novel TB vaccines.
Collapse
|
31
|
Tima HG, Huygen K, Romano M. Innate signaling by mycobacterial cell wall components and relevance for development of adjuvants for subunit vaccines. Expert Rev Vaccines 2016; 15:1409-1420. [PMID: 27206681 DOI: 10.1080/14760584.2016.1187067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Pathogen recognition receptors (PRRs) recognize pathogen-associated molecular patterns, triggering the induction of inflammatory innate responses and contributing to the development of specific adaptive immune responses. Novel adjuvants have been developed based on agonists of PRRs. Areas covered: Lipid pathogen-associated molecular patterns (PAMPs) present in the cell wall of mycobacteria are revised, with emphasis on agonists of C-type lectin receptors, signaling pathways, and preclinical data supporting their use as novel adjuvants inducing cell-mediated immune responses. Their potential use as lipid antigens in novel tuberculosis subunit vaccines is also discussed. Expert commentary: Few adjuvants are licensed for human use and mainly favour antibody-mediated protective immunity. Use of lipid PAMPs that trigger cell-mediated immune responses could lead to the development of adjuvants for vaccines against intracellular pathogens and cancer.
Collapse
Affiliation(s)
- Hermann Giresse Tima
- a Immunology Service, Communicable and Infectious Diseases Department , Scientific Institute of Public Health (WIV-ISP) , Brussels , Belgium
| | - Kris Huygen
- a Immunology Service, Communicable and Infectious Diseases Department , Scientific Institute of Public Health (WIV-ISP) , Brussels , Belgium
| | - Marta Romano
- a Immunology Service, Communicable and Infectious Diseases Department , Scientific Institute of Public Health (WIV-ISP) , Brussels , Belgium
| |
Collapse
|
32
|
Patin EC, Willcocks S, Orr S, Ward TH, Lang R, Schaible UE. Mincle-mediated anti-inflammatory IL-10 response counter-regulates IL-12 in vitro. Innate Immun 2016; 22:181-5. [PMID: 26939595 PMCID: PMC4826952 DOI: 10.1177/1753425916636671] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/27/2016] [Indexed: 01/22/2023] Open
Abstract
The role of macrophage-inducible C-type lectin (Mincle) in anti-inflammatory responses has not yet been fully characterized. Herein, we show that engagement of Mincle by trehalose-dimycolate or mycobacteria promotes IL-10 production in macrophages, which causes down-regulation of IL-12p40 secretion. Thus, Mincle mediates both pro- as well as anti-inflammatory responses.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Department of Immunology and Infection, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK Priority Area Infections, Research Center Borstel, Borstel, Germany Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Sam Willcocks
- Department of Immunology and Infection, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK
| | - Selinda Orr
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Theresa H Ward
- Department of Immunology and Infection, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrich E Schaible
- Department of Immunology and Infection, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK Priority Area Infections, Research Center Borstel, Borstel, Germany German Center for Infection Research, TTU-TB, Borstel, Germany
| |
Collapse
|
33
|
Abstract
Through thousands of years of reciprocal coevolution, Mycobacterium tuberculosis has become one of humanity's most successful pathogens, acquiring the ability to establish latent or progressive infection and persist even in the presence of a fully functioning immune system. The ability of M. tuberculosis to avoid immune-mediated clearance is likely to reflect a highly evolved and coordinated program of immune evasion strategies that interfere with both innate and adaptive immunity. These include the manipulation of their phagosomal environment within host macrophages, the selective avoidance or engagement of pattern recognition receptors, modulation of host cytokine production, and the manipulation of antigen presentation to prevent or alter the quality of T-cell responses. In this article we review an extensive array of published studies that have begun to unravel the sophisticated program of specific mechanisms that enable M. tuberculosis and other pathogenic mycobacteria to persist and replicate in the face of considerable immunological pressure from their hosts. Unraveling the mechanisms by which M. tuberculosis evades or modulates host immune function is likely to be of major importance for the development of more effective new vaccines and targeted immunotherapy against tuberculosis.
Collapse
|
34
|
Waters WR, Maggioli MF, Palmer MV, Thacker TC, McGill JL, Vordermeier HM, Berney-Meyer L, Jacobs WR, Larsen MH. Interleukin-17A as a Biomarker for Bovine Tuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:168-80. [PMID: 26677202 PMCID: PMC4744917 DOI: 10.1128/cvi.00637-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/10/2015] [Indexed: 11/20/2022]
Abstract
T helper 17 (Th17)-associated cytokines are integral to the immune responses to tuberculosis, initiating both protective and harmful inflammatory responses. The aim of the present study was to evaluate applied aspects of interleukin-17 (IL-17) biology in the context of Mycobacterium bovis infection of cattle. Using transcriptome sequencing (RNA-Seq), numerous Th17-associated cytokine genes (including IL-17A, IL-17F, IL-22, IL-19, and IL-27) were upregulated >9-fold in response to purified protein derivative stimulation of peripheral blood mononuclear cells from experimentally M. bovis-infected cattle. Protective vaccines elicited IL-17A, IL-17F, IL-22, and IL-27 responses. Reduced IL-17A responses by vaccine recipients, compared to nonvaccinated animals, at 2.5 weeks after M. bovis challenge correlated with reduced disease burdens. Additionally, IL-17A and interferon gamma (IFN-γ) responses were highly correlated and exhibited similar diagnostic capacities. The present findings support the use of Th17-associated cytokines as biomarkers of infection and protection in the immune responses to bovine tuberculosis.
Collapse
Affiliation(s)
- W Ray Waters
- National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA
| | - Mayara F Maggioli
- National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA Iowa State University, College of Veterinary Medicine, Ames, Iowa, USA
| | - Mitchell V Palmer
- National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA
| | - Tyler C Thacker
- National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, Iowa, USA
| | - Jodi L McGill
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | | | - Linda Berney-Meyer
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, New York, USA
| | - William R Jacobs
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, New York, USA
| | - Michelle H Larsen
- Albert Einstein College of Medicine, Department of Microbiology and Immunology, Bronx, New York, USA
| |
Collapse
|
35
|
Slama N, Jamet S, Frigui W, Pawlik A, Bottai D, Laval F, Constant P, Lemassu A, Cam K, Daffé M, Brosch R, Eynard N, Quémard A. The changes in mycolic acid structures caused byhadCmutation have a dramatic effect on the virulence ofMycobacterium tuberculosis. Mol Microbiol 2015; 99:794-807. [DOI: 10.1111/mmi.13266] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Nawel Slama
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Stevie Jamet
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Wafa Frigui
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics; Paris France
| | - Alexandre Pawlik
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics; Paris France
| | - Daria Bottai
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics; Paris France
| | - Françoise Laval
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Patricia Constant
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Anne Lemassu
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Kaymeuang Cam
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Mamadou Daffé
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| | - Roland Brosch
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics; Paris France
| | - Nathalie Eynard
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
| | - Annaïk Quémard
- Département Tuberculose & Biologie des Infections; Institut de Pharmacologie et de Biologie Structurale - UMR5089; Centre National de la Recherche Scientifique; Toulouse France
- Institut de Pharmacologie et de Biologie Structurale; Université de Toulouse; Université Paul Sabatier; Toulouse France
| |
Collapse
|
36
|
Ng TW, Saavedra-Ávila NA, Kennedy SC, Carreño LJ, Porcelli SA. Current efforts and future prospects in the development of live mycobacteria as vaccines. Expert Rev Vaccines 2015; 14:1493-507. [PMID: 26366616 DOI: 10.1586/14760584.2015.1089175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of more effective vaccines against Mycobacterium tuberculosis (Mtb) remains a major goal in the effort to reduce the enormous global burden of disease caused by this pathogen. Whole-cell vaccines based on live mycobacteria with attenuated virulence represent an appealing approach, providing broad antigen exposure and intrinsic adjuvant properties to prime durable immune responses. However, designing vaccine strains with an optimal balance between attenuation and immunogenicity has proven to be extremely challenging. Recent basic and clinical research efforts have broadened our understanding of Mtb pathogenesis and created numerous new vaccine candidates that have been designed to overcome different aspects of immune evasion by Mtb. In this review, we provide an overview of the current efforts to create improved vaccines against tuberculosis based on modifications of live attenuated mycobacteria. In addition, we discuss the use of such vaccine strains as vectors for stimulating protective immunity against other infectious diseases and cancers.
Collapse
Affiliation(s)
- Tony W Ng
- a 1 Albert Einstein College of Medicine - Microbiology & Immunology, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Noemí A Saavedra-Ávila
- a 1 Albert Einstein College of Medicine - Microbiology & Immunology, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Steven C Kennedy
- a 1 Albert Einstein College of Medicine - Microbiology & Immunology, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Leandro J Carreño
- a 1 Albert Einstein College of Medicine - Microbiology & Immunology, 1300 Morris Park Avenue, Bronx, NY 10461, USA.,b 2 Millennium Institute on Immunology and Immunotherapy, Programa Disciplinario de Inmunologia, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Steven A Porcelli
- a 1 Albert Einstein College of Medicine - Microbiology & Immunology, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
37
|
Abstract
Developments in the use of genomics to guide natural product discovery and a recent emphasis on understanding the molecular mechanisms of microbiota-host interactions have converged on the discovery of small molecules from the human microbiome. Here, we review what is known about small molecules produced by the human microbiota. Numerous molecules representing each of the major metabolite classes have been found that have a variety of biological activities, including immune modulation and antibiosis. We discuss technologies that will affect how microbiota-derived molecules are discovered in the future and consider the challenges inherent in finding specific molecules that are critical for driving microbe-host and microbe-microbe interactions and understanding their biological relevance.
Collapse
Affiliation(s)
- Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and the California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
38
|
Viswanathan G, Joshi SV, Sridhar A, Dutta S, Raghunand TR. Identifying novel mycobacterial stress associated genes using a random mutagenesis screen in Mycobacterium smegmatis. Gene 2015. [PMID: 26211627 DOI: 10.1016/j.gene.2015.07.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Cell envelope associated components of Mycobacterium tuberculosis (M.tb) have been implicated in stress response, immune modulation and in vivo survival of the pathogen. Although many such factors have been identified, there is a large disparity between the number of genes predicted to be involved in functions linked to the envelope and those described in the literature. To identify and characterise novel stress related factors associated with the mycobacterial cell envelope, we isolated colony morphotype mutants of Mycobacterium smegmatis (M. smegmatis), based on the hypothesis that mutants with unusual colony morphology may have defects in the biosynthesis of cell envelope components. On testing their susceptibility to stress conditions relevant to M.tb physiology, multiple mutants were found to be sensitive to Isoniazid, Diamide and H2O2, indicative of altered permeability due to changes in cell envelope composition. Two mutants showed defects in biofilm formation implying possible roles for the target genes in antibiotic tolerance and/or virulence. These assays identified novel stress associated roles for several mycobacterial genes including sahH, tatB and aceE. Complementation analysis of selected mutants with the M. smegmatis genes and their M.tb homologues showed phenotypic restoration, validating their link to the observed phenotypes. A mutant carrying an insertion in fhaA encoding a forkhead associated domain containing protein, showed reduced survival in THP-1 macrophages, providing in vivo validation to this screen. Taken together, these results suggest that the M.tb homologues of a majority of the identified genes may play significant roles in the pathogenesis of tuberculosis.
Collapse
Affiliation(s)
| | - Shrilaxmi V Joshi
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, India
| | - Aditi Sridhar
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, India
| | - Sayantanee Dutta
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, India
| | | |
Collapse
|
39
|
Mendum TA, Wu H, Kierzek AM, Stewart GR. Lipid metabolism and Type VII secretion systems dominate the genome scale virulence profile of Mycobacterium tuberculosis in human dendritic cells. BMC Genomics 2015; 16:372. [PMID: 25956932 PMCID: PMC4425887 DOI: 10.1186/s12864-015-1569-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/23/2015] [Indexed: 12/20/2022] Open
Abstract
Background Mycobacterium tuberculosis continues to kill more people than any other bacterium. Although its archetypal host cell is the macrophage, it also enters, and survives within, dendritic cells (DCs). By modulating the behaviour of the DC, M. tuberculosis is able to manipulate the host’s immune response and establish an infection. To identify the M. tuberculosis genes required for survival within DCs we infected primary human DCs with an M. tuberculosis transposon library and identified mutations with a reduced ability to survive. Results Parallel sequencing of the transposon inserts of the surviving mutants identified a large number of genes as being required for optimal intracellular fitness in DCs. Loci whose mutation attenuated intracellular survival included those involved in synthesising cell wall lipids, not only the well-established virulence factors, pDIM and cord factor, but also sulfolipids and PGL, which have not previously been identified as having a direct virulence role in cells. Other attenuated loci included the secretion systems ESX-1, ESX-2 and ESX-4, alongside many PPE genes, implicating a role for ESX-5. In contrast the canonical ESAT-6 family of ESX substrates did not have intra-DC fitness costs suggesting an alternative ESX-1 associated virulence mechanism. With the aid of a gene-nutrient interaction model, metabolic processes such as cholesterol side chain catabolism, nitrate reductase and cysteine-methionine metabolism were also identified as important for survival in DCs. Conclusion We conclude that many of the virulence factors required for survival in DC are shared with macrophages, but that survival in DCs also requires several additional functions, such as cysteine-methionine metabolism, PGLs, sulfolipids, ESX systems and PPE genes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1569-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tom A Mendum
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Huihai Wu
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Andrzej M Kierzek
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Graham R Stewart
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
40
|
Zimhony O, Schwarz A, Raitses-Gurevich M, Peleg Y, Dym O, Albeck S, Burstein Y, Shakked Z. AcpM, the meromycolate extension acyl carrier protein of Mycobacterium tuberculosis, is activated by the 4'-phosphopantetheinyl transferase PptT, a potential target of the multistep mycolic acid biosynthesis. Biochemistry 2015; 54:2360-71. [PMID: 25785780 DOI: 10.1021/bi501444e] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Modification of acyl carrier proteins (ACP) or domains by the covalent binding of a 4'-phosphopantetheine (4'-PP) moiety is a fundamental condition for activation of fatty acid synthases (FASes) and polyketide synthases (PKSes). Binding of 4'-PP is mediated by 4' phosphopantetheinyl transfersases (PPTases). Mycobacterium tuberculosis (Mtb) possesses two essential PPTases: acyl carrier protein synthase (Mtb AcpS), which activates the multidomain fatty acid synthase I (FAS I), and Mtb PptT, an Sfp-type broad spectrum PPTase that activates PKSes. To date, it has not been determined which of the two Mtb PPTases, AcpS or PptT, activates the meromycolate extension ACP, Mtb AcpM, en route to the production of mycolic acids, the main components of the mycobacterial cell wall. In this study, we tested the enzymatic activation of a highly purified Mtb apo-AcpM to Mtb holo-AcpM by either Mtb PptT or Mtb AcpS. By using SDS-PAGE band shift assay and mass spectrometry analysis, we found that Mtb PptT is the PPTase that activates Mtb AcpM. We measured the catalytic activity of Mtb PptT toward CoA, using an activation assay of a blue pigment synthase, BpsA (a nonribosomal peptide synthase, NRPS). BpsA activation by Mtb PptT was inhibited by Mtb apo-AcpM through competition for CoA, in accord with Mtb AcpM activation. A structural model of the putative interaction between Mtb PptT and Mtb AcpM suggests that both hydrophobic and electrostatic interactions stabilize this complex. To conclude, activation of Mtb AcpM by Mtb PptT reveals a potential target of the multistep mycolic acid biosynthesis.
Collapse
Affiliation(s)
- Oren Zimhony
- †Kaplan Medical Center, Affiliated to the School of Medicine, Hebrew University of Jerusalem and Hadassah Medical Center, POB1 Rehovot 76100, Israel
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Orme IM, Robinson RT, Cooper AM. The balance between protective and pathogenic immune responses in the TB-infected lung. Nat Immunol 2015; 16:57-63. [PMID: 25521685 DOI: 10.1038/ni.3048] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/05/2014] [Indexed: 01/01/2023]
Abstract
Tuberculosis is a disease of the lung, and efficient transmission is dependent on the generation of a lesion in the lung, which results in a bacterium-laden cough. Mycobacterium tuberculosis (Mtb) is able to manipulate both the innate and acquired immune response of the host. This manipulation results in an effective CD4(+) T cell response that limits disease throughout the body but can also promote the development of progressively destructive lesions in the lung. In this way Mtb infection can result in an ambulatory individual who has a lesion in the lung capable of transmitting Mtb. The inflammatory environment within the lung lesion is manipulated by Mtb throughout infection and can limit the expression of acquired immunity by a variety of pathways.
Collapse
Affiliation(s)
- Ian M Orme
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Richard T Robinson
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
42
|
Welsh KJ, Hunter RL, Actor JK. Trehalose 6,6'-dimycolate--a coat to regulate tuberculosis immunopathogenesis. Tuberculosis (Edinb) 2014; 93 Suppl:S3-9. [PMID: 24388646 DOI: 10.1016/s1472-9792(13)70003-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) remains a significant public health burden worldwide. Treatment of this disease requires a minimum of six months and there is no vaccine available for the most common form of the disease. Increasing evidence suggests that the mycobacterial glycolipid trehalose 6,6' dimycolate (TDM; cord factor) plays a key role in the pathogenesis of TB disease. TDM protects the TB bacilli from macrophage-mediated killing, inhibits effective antigen presentation, and reduces the formation of protective T-cell responses. TDM promotes initiation of granuloma formation and likely plays a role in caseation. Furthermore, TDM may contribute to the development of post primary disease. Receptors for TDM were recently described and are expected to contribute to our knowledge of the molecular pathogenesis of TB disease. In this manner, understanding TDM may prove promising towards development of targeted TB therapeutics to limit clinical pathologies.
Collapse
Affiliation(s)
- Kerry J Welsh
- Department of Pathology, Medical School, University of Texas-Houston Medical School, Houston, Texas, USA
| | - Robert L Hunter
- Department of Pathology, Medical School, University of Texas-Houston Medical School, Houston, Texas, USA
| | - Jeffrey K Actor
- Department of Pathology, Medical School, University of Texas-Houston Medical School, Houston, Texas, USA.
| |
Collapse
|
43
|
|
44
|
Abstract
ABSTRACT
Mycolic acids are major and specific long-chain fatty acids that represent essential components of the
Mycobacterium tuberculosis
cell envelope. They play a crucial role in the cell wall architecture and impermeability, hence the natural resistance of mycobacteria to most antibiotics, and represent key factors in mycobacterial virulence. Biosynthesis of mycolic acid precursors requires two types of fatty acid synthases (FASs), the eukaryotic-like multifunctional enzyme FAS I and the acyl carrier protein (ACP)–dependent FAS II systems, which consists of a series of discrete mono-functional proteins, each catalyzing one reaction in the pathway. Unlike FAS II synthases of other bacteria, the mycobacterial FAS II is incapable of
de novo
fatty acid synthesis from acetyl-coenzyme A, but instead elongates medium-chain-length fatty acids previously synthesized by FAS I, leading to meromycolic acids. In addition, mycolic acid subspecies with defined biological properties can be distinguished according to the chemical modifications decorating the meromycolate. Nearly all the genetic components involved in both elongation and functionalization of the meromycolic acid have been identified and are generally clustered in distinct transcriptional units. A large body of information has been generated on the enzymology of the mycolic acid biosynthetic pathway and on their genetic and biochemical/structural characterization as targets of several antitubercular drugs. This chapter is a comprehensive overview of mycolic acid structure, function, and biosynthesis. Special emphasis is given to recent work addressing the regulation of mycolic acid biosynthesis, adding new insights to our understanding of how pathogenic mycobacteria adapt their cell wall composition in response to environmental changes.
Collapse
|
45
|
Host-pathogen interactions during Mycobacterium tuberculosis infections. Curr Top Microbiol Immunol 2014; 374:211-41. [PMID: 23881288 DOI: 10.1007/82_2013_332] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The intimate and persistent connection between Mycobacterium tuberculosis and its human host suggests that the pathogen has evolved extensive mechanisms to evade eradication by the immune system. In particular, the organism has adapted to replicate within phagocytic cells, especially macrophages, which are specialized to kill microbes. Over the past decade of M. tuberculosis research, the means to manipulate both the organism and the host has ushered in an exciting time that has uncovered some of the mechanisms of the innate macrophage-pathogen interactions that lie at the heart of M. tuberculosis pathogenesis, though many interactions likely still await discovery. In this chapter, we will delve into some of these advances, with an emphasis on the interactions that occur on the cellular level when M. tuberculosis cells encounter macrophages. In particular, we focus on two major aspects of M. tuberculosis biology regarding the proximal physical interface between the bacterium and host, namely the interactions with the phagosomal membrane as well as the distinctive mycobacterial cell wall. Importantly, some of the emerging paradigms in M. tuberculosis pathogenesis and host response represent common themes in bacterial pathogenesis, such as the role of host cell membrane perforation in intracellular survival and host response. However, the array of unique bacterial lipid mediators and their interaction with host cells highlights the unique biology of this persistent pathogen.
Collapse
|
46
|
Purwantini E, Mukhopadhyay B. Rv0132c of Mycobacterium tuberculosis encodes a coenzyme F420-dependent hydroxymycolic acid dehydrogenase. PLoS One 2013; 8:e81985. [PMID: 24349169 PMCID: PMC3859598 DOI: 10.1371/journal.pone.0081985] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/19/2013] [Indexed: 11/19/2022] Open
Abstract
The ability of Mycobacterium tuberculosis to manipulate and evade human immune system is in part due to its extraordinarily complex cell wall. One of the key components of this cell wall is a family of lipids called mycolic acids. Oxygenation of mycolic acids generating methoxy- and ketomycolic acids enhances the pathogenic attributes of M. tuberculosis. Thus, the respective enzymes are of interest in the research on mycobacteria. The generation of methoxy- and ketomycolic acids proceeds through intermediary formation of hydroxymycolic acids. While the methyl transferase that generates methoxymycolic acids from hydroxymycolic acids is known, hydroxymycolic acids dehydrogenase that oxidizes hydroxymycolic acids to ketomycolic acids has been elusive. We found that hydroxymycolic acid dehydrogenase is encoded by the rv0132c gene and the enzyme utilizes F420, a deazaflavin coenzyme, as electron carrier, and accordingly we called it F420-dependent hydroxymycolic acid dehydrogenase. This is the first report on the involvement of F420 in the synthesis of a mycobacterial cell envelope. Also, F420-dependent hydroxymycolic acid dehydrogenase was inhibited by PA-824, and therefore, it is a previously unknown target for this new tuberculosis drug.
Collapse
Affiliation(s)
- Endang Purwantini
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Biswarup Mukhopadhyay
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Departments of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
- Virginia Tech Carilion School of Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Genetic regulation of vesiculogenesis and immunomodulation in Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2013; 110:E4790-7. [PMID: 24248369 DOI: 10.1073/pnas.1320118110] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) restrains immune responses well enough to escape eradication but elicits enough immunopathology to ensure its transmission. Here we provide evidence that this host-pathogen relationship is regulated in part by a cytosolic, membrane-associated protein with a unique structural fold, encoded by the Mtb gene rv0431. The protein acts by regulating the quantity of Mtb-derived membrane vesicles bearing Toll-like receptor 2 ligands, including the lipoproteins LpqH and SodC. We propose that rv0431 be named "vesiculogenesis and immune response regulator."
Collapse
|
48
|
Keto-mycolic acid-dependent pellicle formation confers tolerance to drug-sensitive Mycobacterium tuberculosis. mBio 2013; 4:e00222-13. [PMID: 23653446 PMCID: PMC3663190 DOI: 10.1128/mbio.00222-13] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chronic nature of tuberculosis (TB), its requirement of long duration of treatment, its ability to evade immune intervention, and its propensity to relapse after drug treatment is discontinued are reminiscent of other chronic, biofilm-associated bacterial diseases. Historically, Mycobacterium tuberculosis was grown as a pellicle, a biofilm-like structure, at the liquid-air interface in a variety of synthetic media. Notably, the most widely administered human vaccine, BCG, is grown as a pellicle for vaccine production. However, the molecular requirements for this growth remain ill defined. Here, we demonstrate that keto-mycolic acids (keto-MA) are essential for pellicle growth, and mutants lacking in or depleted of this MA species are unable to form a pellicle. We investigated the role of the pellicle biofilm in the reduction of antibiotic sensitivity known as drug tolerance using the pellicle-defective ΔmmaA4 mutant strain. We discovered that the ΔmmaA4 mutant, which is both pellicle defective and highly sensitive to rifampicin (RIF) under planktonic growth, when incorporated within the wild-type pellicle biofilm, was protected from the bactericidal activity of RIF. The observation that growth within the M. tuberculosis pellicle biofilm can confer drug tolerance to a drug-hypersensitive strain suggests that identifying molecular requirements for pellicle growth could lead to development of novel interventions against mycobacterial infections. Our findings also suggest that a class of drugs that can disrupt M. tuberculosis biofilm formation, when used in conjunction with conventional antibiotics, has the potential to overcome drug tolerance. Two of the most important questions in tuberculosis (TB) research are (i) how does Mycobacterium tuberculosis persist in the human host for decades in the face of an active immune response and (ii) why does it take six months and four drugs to treat uncomplicated TB. Both these aspects of M. tuberculosis biology are reminiscent of infections caused by organisms capable of forming biofilms. M. tuberculosis is capable of growing as a biofilm-like structure called the pellicle. In this study, we demonstrate that a specific cell wall component, keto-mycolic acid, is essential for pellicle growth. We also demonstrate that a strain of M. tuberculosis that is both drug sensitive and pellicle defective exhibits commensal behavior and becomes drug tolerant by becoming part of a heterogeneous pellicle, a characteristic of multispecies biofilms. These observations could have important implications for identifying novel pathways for M. tuberculosis drug tolerance and the design of new modalities to rapidly treat TB.
Collapse
|
49
|
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
|
50
|
Gómez-Velasco A, Bach H, Rana AK, Cox LR, Bhatt A, Besra GS, Av-Gay Y. Disruption of the serine/threonine protein kinase H affects phthiocerol dimycocerosates synthesis in Mycobacterium tuberculosis. MICROBIOLOGY-SGM 2013; 159:726-736. [PMID: 23412844 PMCID: PMC3709824 DOI: 10.1099/mic.0.062067-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mycobacterium tuberculosis possesses a complex cell wall that is unique and essential for interaction of the pathogen with its human host. Emerging evidence suggests that the biosynthesis of complex cell-wall lipids is mediated by serine/threonine protein kinases (STPKs). Herein, we show, using in vivo radiolabelling, MS and immunostaining analyses, that targeted deletion of one of the STPKs, pknH, attenuates the production of phthiocerol dimycocerosates (PDIMs), a major M. tuberculosis virulence lipid. Comparative protein expression analysis revealed that proteins in the PDIM biosynthetic pathway are differentially expressed in a deleted pknH strain. Furthermore, we analysed the composition of the major lipoglycans, lipoarabinomannan (LAM) and lipomannan (LM), and found a twofold higher LAM/LM ratio in the mutant strain. Thus, we provide experimental evidence that PknH contributes to the production and synthesis of M. tuberculosis cell-wall components.
Collapse
Affiliation(s)
- Anaximandro Gómez-Velasco
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Amrita K Rana
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Liam R Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Apoorva Bhatt
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Yossef Av-Gay
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|