1
|
Salgueiro-Toledo VC, Bertol J, Gutierrez C, Serrano-Mestre JL, Ferrer-Luzon N, Vázquez-Iniesta L, Palacios A, Pasquina-Lemonche L, Espaillat A, Lerma L, Weinrick B, Lavin JL, Elortza F, Azkargorta M, Prieto A, Buendía-Nacarino P, Luque-García JL, Neyrolles O, Cava F, Hobbs JK, Sanz J, Prados-Rosales R. Maintenance of cell wall remodeling and vesicle production are connected in Mycobacterium tuberculosis. eLife 2025; 13:RP94982. [PMID: 39960848 PMCID: PMC11832169 DOI: 10.7554/elife.94982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Pathogenic and nonpathogenic mycobacteria secrete extracellular vesicles (EVs) under various conditions. EVs produced by Mycobacterium tuberculosis (Mtb) have raised significant interest for their potential in cell communication, nutrient acquisition, and immune evasion. However, the relevance of vesicle secretion during tuberculosis infection remains unknown due to the limited understanding of mycobacterial vesicle biogenesis. We have previously shown that a transposon mutant in the LCP-related gene virR (virRmut) manifested a strong attenuated phenotype during experimental macrophage and murine infections, concomitant to enhanced vesicle release. In this study, we aimed to understand the role of VirR in the vesicle production process in Mtb. We employ genetic, transcriptional, proteomics, ultrastructural, and biochemical methods to investigate the underlying processes explaining the enhanced vesiculogenesis phenomenon observed in the virRmut. Our results establish that VirR is critical to sustain proper cell permeability via regulation of cell envelope remodeling possibly through the interaction with similar cell envelope proteins, which control the link between peptidoglycan and arabinogalactan. These findings advance our understanding of mycobacterial extracellular vesicle biogenesis and suggest that these set of proteins could be attractive targets for therapeutic intervention.
Collapse
Affiliation(s)
- Vivian C Salgueiro-Toledo
- Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma de MadridMadridSpain
| | - Jorge Bertol
- Institute for Bio-computation and Physics of Complex Systems BIFI, Department of Theoretical Physics, University of ZaragozaZaragozaSpain
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPSToulouseSpain
| | - Jose L Serrano-Mestre
- Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma de MadridMadridSpain
| | - Noelia Ferrer-Luzon
- Institute for Bio-computation and Physics of Complex Systems BIFI, Department of Theoretical Physics, University of ZaragozaZaragozaSpain
| | - Lucia Vázquez-Iniesta
- Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma de MadridMadridSpain
| | - Ainhoa Palacios
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Science and Technology ParkDerioSpain
| | | | - Akbar Espaillat
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå UniversityUmeåSweden
| | - Laura Lerma
- Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma de MadridMadridSpain
| | | | | | - Felix Elortza
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Science and Technology ParkDerioSpain
| | - Mikel Azkargorta
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Science and Technology ParkDerioSpain
| | - Alicia Prieto
- Department of Microbial & Plan Biotechnology, Centro de Investigaciones Biológicas Margarita Salas, Spanish National Research Council (CSIC)MadridSpain
| | | | - Jose L Luque-García
- Department of Analytical Chemistry, Universidad Complutense de MadridMadridSpain
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPSToulouseSpain
| | - Felipe Cava
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå UniversityUmeåSweden
| | - Jamie K Hobbs
- Department of Physics and Astronomy, University of SheffieldSheffieldUnited Kingdom
| | - Joaquín Sanz
- Institute for Bio-computation and Physics of Complex Systems BIFI, Department of Theoretical Physics, University of ZaragozaZaragozaSpain
| | - Rafael Prados-Rosales
- Department of Preventive Medicine and Public Health and Microbiology, Universidad Autónoma de MadridMadridSpain
| |
Collapse
|
2
|
Salgueiro V, Bertol J, Gutierrez C, Serrano-Mestre JL, Ferrer-Luzon N, Vázquez-Iniesta L, Palacios A, Pasquina-Lemonche L, Espaillat A, Lerma L, Weinrick B, Lavin JL, Elortza F, Azkalgorta M, Prieto A, Buendía-Nacarino P, Luque-García JL, Neyrolles O, Cava F, Hobbs JK, Sanz J, Prados-Rosales R. Maintenance of cell wall remodeling and vesicle production are connected in Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.19.567727. [PMID: 38187572 PMCID: PMC10769192 DOI: 10.1101/2023.11.19.567727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Pathogenic and nonpathogenic mycobacteria secrete extracellular vesicles (EVs) under various conditions. EVs produced by Mycobacterium tuberculosis ( Mtb ) have raised significant interest for their potential in cell communication, nutrient acquisition, and immune evasion. However, the relevance of vesicle secretion during tuberculosis infection remains unknown due to the limited understanding of mycobacterial vesicle biogenesis. We have previously shown that a transposon mutant in the LCP-related gene virR ( virR mut ) manifested a strong attenuated phenotype during experimental macrophage and murine infections, concomitant to enhanced vesicle release. In this study, we aimed to understand the role of VirR in the vesicle production process in Mtb . We employ genetic, transcriptional, proteomics, ultrastructural and biochemical methods to investigate the underlying processes explaining the enhanced vesiculogenesis phenomenon observed in the virR mut . Our results establish that VirR is critical to sustain proper cell permeability via regulation of cell envelope remodeling possibly through the interaction with similar cell envelope proteins, which control the link between peptidoglycan and arabinogalactan. These findings advance our understanding of mycobacterial extracellular vesicle biogenesis and suggest that these set of proteins could be attractive targets for therapeutic intervention.
Collapse
|
3
|
Yang J, Ma Y, Yu J, Liu Y, Xia J, Kong X, Jin X, Li J, Lin S, Ruan Y, Yang F, Pi J. Advancing Roles and Therapeutic Potentials of Pyroptosis in Host Immune Defenses against Tuberculosis. Biomolecules 2024; 14:1255. [PMID: 39456188 PMCID: PMC11505957 DOI: 10.3390/biom14101255] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (Mtb) infection, remains a deadly global public health burden. The use of recommended drug combinations in clinic has seen an increasing prevalence of drug-resistant TB, adding to the impediments to global control of TB. Therefore, control of TB and drug-resistant TB has become one of the most pressing issues in global public health, which urges the exploration of potential therapeutic targets in TB and drug-resistant TB. Pyroptosis, a form of programmed cell death characterized by cell swelling and rupture, release of cellular contents and inflammatory responses, has been found to promote pathogen clearance and adopt crucial roles in the control of bacterial infections. It has been demonstrated that Mtb can cause host cell pyroptosis, and these host cells, which are infected by Mtb, can kill Mtb accompanied by pyroptosis, while, at the same time, pyroptosis can also release intracellular Mtb, which may potentially worsen the infection by exacerbating the inflammation. Here, we describe the main pathways of pyroptosis during Mtb infection and summarize the identified effectors of Mtb that regulate pyroptosis to achieve immune evasion. Moreover, we also discuss the potentials of pyroptosis to serve as an anti-TB therapeutic target, with the aim of providing new ideas for the development of TB treatments.
Collapse
Affiliation(s)
- Jiayi Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yuhe Ma
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaqi Yu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yilin Liu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China;
| | - Xinen Kong
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Xiaoying Jin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaxiang Li
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Siqi Lin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yongdui Ruan
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Fen Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiang Pi
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| |
Collapse
|
4
|
Veerapandian R, Gadad SS, Jagannath C, Dhandayuthapani S. Live Attenuated Vaccines against Tuberculosis: Targeting the Disruption of Genes Encoding the Secretory Proteins of Mycobacteria. Vaccines (Basel) 2024; 12:530. [PMID: 38793781 PMCID: PMC11126151 DOI: 10.3390/vaccines12050530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis (TB), a chronic infectious disease affecting humans, causes over 1.3 million deaths per year throughout the world. The current preventive vaccine BCG provides protection against childhood TB, but it fails to protect against pulmonary TB. Multiple candidates have been evaluated to either replace or boost the efficacy of the BCG vaccine, including subunit protein, DNA, virus vector-based vaccines, etc., most of which provide only short-term immunity. Several live attenuated vaccines derived from Mycobacterium tuberculosis (Mtb) and BCG have also been developed to induce long-term immunity. Since Mtb mediates its virulence through multiple secreted proteins, these proteins have been targeted to produce attenuated but immunogenic vaccines. In this review, we discuss the characteristics and prospects of live attenuated vaccines generated by targeting the disruption of the genes encoding secretory mycobacterial proteins.
Collapse
Affiliation(s)
- Raja Veerapandian
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Shrikanth S. Gadad
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX 77030, USA
| | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
5
|
Salgueiro VC, Passemar C, Vázquez-Iniesta L, Lerma L, Floto A, Prados-Rosales R. Extracellular vesicles in mycobacteria: new findings in biogenesis, host-pathogen interactions, and diagnostics. mBio 2024; 15:e0255223. [PMID: 38567992 PMCID: PMC11077946 DOI: 10.1128/mbio.02552-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024] Open
Abstract
Since the discovery of extracellular vesicles (EVs) in mycobacterial species 15 years back, we have learned that this phenomenon is conserved in the Mycobacterium genus and has critical roles in bacterial physiology and host-pathogen interactions. Mycobacterium tuberculosis (Mtb), the tuberculosis (TB) causative agent, produces EVs both in vitro and in vivo including a diverse set of biomolecules with demonstrated immunomodulatory effects. Moreover, Mtb EVs (MEVs) have been shown to possess vaccine properties and carry biomarkers with diagnostic capacity. Although information on MEV biogenesis relative to other bacterial species is scarce, recent studies have shed light on how MEVs originate and are released to the extracellular space. In this minireview, we discuss past and new information about the vesiculogenesis phenomenon in Mtb, including biogenesis, MEV cargo, aspects in the context of host-pathogen interactions, and applications that could help to develop effective tools to tackle the disease.
Collapse
Affiliation(s)
- Vivian C. Salgueiro
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Charlotte Passemar
- Cambridge Center for Lung Infection, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | - Lucía Vázquez-Iniesta
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Lerma
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Andrés Floto
- Cambridge Center for Lung Infection, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | - Rafael Prados-Rosales
- Department of Preventive Medicine, Public Health, and Microbiology. School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
6
|
Comín J, Campos E, Gonzalo-Asensio J, Samper S. Transcriptomic profile of the most successful Mycobacterium tuberculosis strain in Aragon, the MtZ strain, during exponential and stationary growth phases. Microbiol Spectr 2023; 11:e0468522. [PMID: 37882511 PMCID: PMC10714837 DOI: 10.1128/spectrum.04685-22] [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: 11/16/2022] [Accepted: 09/21/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE Aragon Community suffered, during the first years of the beginning of this century, a large outbreak caused by the MtZ strain, producing more than 240 cases to date. MtZ strain and the outbreak have been previously studied from an epidemiological and molecular point of view. In this work, we analyzed the transcriptomic profile of the strain for better understanding of its success among our population. We have discovered that MtZ has some upregulated virulence pathways, such as the ESX-1 system, the cholesterol degradation pathway or the peptidoglycan biosynthesis. Interestingly, MtZ has downregulated the uptake of iron. Another special feature of MtZ strain is the interruption of desA3 gene, essential for producing oleic acid. Although the strain takes a long time to grow in the initial culture media, eventually it is able to reach normal optical densities, suggestive of the presence of another route for obtaining oleic acid in Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Jessica Comín
- Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
| | | | - Jesús Gonzalo-Asensio
- Universidad de Zaragoza, Zaragoza, Spain
- Fundación IIS Aragón, Zaragoza, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Sofía Samper
- Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
- Fundación IIS Aragón, Zaragoza, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| |
Collapse
|
7
|
Stylianou E, Pinpathomrat N, Sampson O, Richard A, Korompis M, McShane H. A five-antigen Esx-5a fusion delivered as a prime-boost regimen protects against M.tb challenge. Front Immunol 2023; 14:1263457. [PMID: 37869008 PMCID: PMC10585038 DOI: 10.3389/fimmu.2023.1263457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/14/2023] [Indexed: 10/24/2023] Open
Abstract
The development of tuberculosis (TB) vaccines has been hindered by the complex nature of Mycobacterium tuberculosis (M.tb) and the absence of clearly defined immune markers of protection. While Bacillus Calmette-Guerin (BCG) is currently the only licensed TB vaccine, its effectiveness diminishes in adulthood. In our previous research, we identified that boosting BCG with an intranasally administered chimpanzee adenovirus expressing the PPE15 antigen of M.tb (ChAdOx1.PPE15) improved its protection. To enhance the vaccine's efficacy, we combined PPE15 with the other three members of the Esx-5a secretion system and Ag85A into a multi-antigen construct (5Ag). Leveraging the mucosal administration safety of ChAdOx1, we targeted the site of M.tb infection to induce localized mucosal responses, while employing modified vaccinia virus (MVA) to boost systemic immune responses. The combination of these antigens resulted in enhanced BCG protection in both the lungs and spleens of vaccinated mice. These findings provide support for advancing ChAdOx1.5Ag and MVA.5Ag to the next stages of vaccine development.
Collapse
Affiliation(s)
- Elena Stylianou
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
8
|
Malaga W, Payros D, Meunier E, Frigui W, Sayes F, Pawlik A, Orgeur M, Berrone C, Moreau F, Mazères S, Gonzalo-Asensio J, Rengel D, Martin C, Astarie-Dequeker C, Mourey L, Brosch R, Guilhot C. Natural mutations in the sensor kinase of the PhoPR two-component regulatory system modulate virulence of ancestor-like tuberculosis bacilli. PLoS Pathog 2023; 19:e1011437. [PMID: 37450466 PMCID: PMC10348564 DOI: 10.1371/journal.ppat.1011437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/22/2023] [Indexed: 07/18/2023] Open
Abstract
The molecular factors and genetic adaptations that contributed to the emergence of Mycobacterium tuberculosis (MTB) from an environmental Mycobacterium canettii-like ancestor, remain poorly investigated. In MTB, the PhoPR two-component regulatory system controls production and secretion of proteins and lipid virulence effectors. Here, we describe that several mutations, present in phoR of M. canettii relative to MTB, impact the expression of the PhoP regulon and the pathogenicity of the strains. First, we establish a molecular model of PhoR and show that some substitutions found in PhoR of M. canettii are likely to impact the structure and activity of this protein. Second, we show that STB-K, the most attenuated available M. canettii strain, displays lower expression of PhoP-induced genes than MTB. Third, we demonstrate that genetic swapping of the phoPR allele from STB-K with the ortholog from MTB H37Rv enhances expression of PhoP-controlled functions and the capacities of the recombinant strain to colonize human macrophages, the MTB target cells, as well as to cause disease in several mouse infection models. Fourth, we extended these observations to other M. canettii strains and confirm that PhoP-controlled functions are expressed at lower levels in most M. canettii strains than in M. tuberculosis. Our findings suggest that distinct PhoR variants have been selected during the evolution of tuberculosis bacilli, contributing to higher pathogenicity and persistence of MTB in the mammalian host.
Collapse
Affiliation(s)
- Wladimir Malaga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Delphine Payros
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Eva Meunier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Wafa Frigui
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Fadel Sayes
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Alexandre Pawlik
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Mickael Orgeur
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Céline Berrone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Flavie Moreau
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Jesus Gonzalo-Asensio
- Grupo de Genética de Micobacterias, Facultad de Medicina, Departamento de Microbiologia, Pediatria, Radiologica y Salud Pùblica, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Institudo de Salud Carlos III, Madrid, Spain
| | - David Rengel
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Carlos Martin
- Grupo de Genética de Micobacterias, Facultad de Medicina, Departamento de Microbiologia, Pediatria, Radiologica y Salud Pùblica, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Institudo de Salud Carlos III, Madrid, Spain
- Servicio de Microbiologia, Hospital Universitario Miguel Servet, ISS Aragon, Zaragoza, Spain
| | - Catherine Astarie-Dequeker
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Lionel Mourey
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
9
|
Villar M, Rajbhandari RM, Artigas-Jerónimo S, Contreras M, Sadaula A, Karmacharya D, Alves PC, Gortázar C, de la Fuente J. Differentially Represented Proteins in Response to Infection with Mycobacterium tuberculosis Identified by Quantitative Serum Proteomics in Asian Elephants. Pathogens 2022; 11:pathogens11091010. [PMID: 36145440 PMCID: PMC9505326 DOI: 10.3390/pathogens11091010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/27/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Tuberculosis is a major global concern. Tuberculosis in wildlife is a risk for zoonotic transmission and becoming one of the challenges for conservation globally. In elephants, the number of cases is likely rising. The aim of this study was to identify proteins related to tuberculosis infection in elephants, which could then be used for the development of diagnostic tools and/or vaccines. A serum proteomics approach was used to characterize differentially represented proteins in response to Mycobacterium tuberculosis in Asian elephants (Elaphas maximus). Blood samples were collected from eight elephants, four of which were antibody positive for tuberculosis and four were antibody negative. Proteomics analysis identified 26 significantly dysregulated proteins in response to tuberculosis. Of these, 10 (38%) were identified as immunoglobulin and 16 (62%) as non-immunoglobulin proteins. The results provided new information on the antibody response to mycobacterial infection and biomarkers associated with tuberculosis and protective response to mycobacteria in Asian elephants. Protective mechanisms included defense against infection (Alpha-1-B glycoprotein A1BG, Serpin family A member 1 SERPINA1, Transthyretin TTR), neuroprotection (TTR), and reduced risks of inflammation, infections, and cancer (SERPINA1, Keratin 10 KRT10). Using a translational biotechnology approach, the results provided information for the identification of candidate diagnostic, prognostic, and protective antigens for monitoring and control of tuberculosis in Asian elephants.
Collapse
Affiliation(s)
- Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Biochemistry Section, Faculty of Science and Chemical Technologies, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Rajesh Man Rajbhandari
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Center for Molecular Dynamics Nepal (CMDN), Thapathali Road 11, Kathmandu 44600, Nepal
- Dep. de Biologia, Faculdade de Ciencias da Universidade do Porto, Rua do Campo Alegre, S/N, Edificio FC4, 4169-007 Porto, Portugal
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado—BIOPOLIS Program in Genomics, Biodiversity and Land Planning, Campus de Vairão, Universidade do Porto, 4485-661 Vairão, Portugal
| | - Sara Artigas-Jerónimo
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
| | - Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
| | - Amir Sadaula
- National Trust for Nature Conservation, Biodiversity Conservation Center, Sauraha, Chitwan 44204, Nepal
| | - Dibesh Karmacharya
- Center for Molecular Dynamics Nepal (CMDN), Thapathali Road 11, Kathmandu 44600, Nepal
| | - Paulo Célio Alves
- Dep. de Biologia, Faculdade de Ciencias da Universidade do Porto, Rua do Campo Alegre, S/N, Edificio FC4, 4169-007 Porto, Portugal
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado—BIOPOLIS Program in Genomics, Biodiversity and Land Planning, Campus de Vairão, Universidade do Porto, 4485-661 Vairão, Portugal
- Estação Biológica de Mértola EBM, Praça Luís de Camões, Mértola, 7750-329 Mértola, Portugal
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Correspondence: (C.G.); or (J.d.l.F.)
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Correspondence: (C.G.); or (J.d.l.F.)
| |
Collapse
|
10
|
Lopez J, Anna F, Authié P, Pawlik A, Ku MW, Blanc C, Souque P, Moncoq F, Noirat A, Hardy D, Sougakoff W, Brosch R, Guinet F, Charneau P, Majlessi L. A lentiviral vector encoding fusion of light invariant chain and mycobacterial antigens induces protective CD4 + T cell immunity. Cell Rep 2022; 40:111142. [PMID: 35905717 DOI: 10.1016/j.celrep.2022.111142] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 12/21/2022] Open
Abstract
Lentiviral vectors (LVs) are highly efficient at inducing CD8+ T cell responses. However, LV-encoded antigens are processed inside the cytosol of antigen-presenting cells, which does not directly communicate with the endosomal major histocompatibility complex class II (MHC-II) presentation pathway. LVs are thus poor at inducing CD4+ T cell response. To overcome this limitation, we devised a strategy whereby LV-encoded antigens are extended at their N-terminal end with the MHC-II-associated light invariant chain (li), which contains an endosome-targeting signal sequence. When evaluated with an LV-encoded polyantigen composed of CD4+ T cell targets from Mycobacterium tuberculosis, intranasal vaccination in mice triggers pulmonary polyfunctional CD4+ and CD8+ T cell responses. Adjuvantation of these LVs extends the mucosal immunity to Th17 and Tc17 responses. A systemic prime and an intranasal boost with one of these LV induces protection against M. tuberculosis. This strategy improves the protective power of LVs against infections and cancers, where CD4+ T cell immunity plays an important role.
Collapse
Affiliation(s)
- Jodie Lopez
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - François Anna
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Alexandre Pawlik
- Institut Pasteur, Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Min-Wen Ku
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Catherine Blanc
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Philippe Souque
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fanny Moncoq
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Amandine Noirat
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - David Hardy
- Institut Pasteur, Histopathology Platform, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Wladimir Sougakoff
- Sorbonne Universités, UPMC Université Paris 06, CIMI-Paris, AP-HP, Hôpital Pitié-Salpêtrière, CNR-MyRMA, 75013 Paris, France
| | - Roland Brosch
- Institut Pasteur, Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Françoise Guinet
- Institut Pasteur, Lymphocytes and Immunity Unit, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
11
|
Mehaffy C, Ryan JM, Kruh-Garcia NA, Dobos KM. Extracellular Vesicles in Mycobacteria and Tuberculosis. Front Cell Infect Microbiol 2022; 12:912831. [PMID: 35719351 PMCID: PMC9204639 DOI: 10.3389/fcimb.2022.912831] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB) remains a public health issue causing millions of infections every year. Of these, about 15% ultimately result in death. Efforts to control TB include development of new and more effective vaccines, novel and more effective drug treatments, and new diagnostics that test for both latent TB Infection and TB disease. All of these areas of research benefit from a good understanding of the physiology of Mycobacterium tuberculosis (Mtb), the primary causative agent of TB. Mtb secreted protein antigens have been the focus of vaccine and diagnosis research for the past century. Recently, the discovery of extracellular vesicles (EVs) as an important source of secreted antigens in Mtb has gained attention. Similarly, the discovery that host EVs can carry Mtb products during in vitro and in vivo infection has spiked interest because of its potential use in blood-based diagnostics. Despite advances in understanding the content of Mtb and Mtb-infected host extracellular vesicles, our understanding on the biogenesis and role of Mtb and host extracellular vesicles during Mtb infection is still nascent. Here, we explore the current literature on extracellular vesicles regarding Mtb, discuss the host and Mtb extracellular vesicles as distinct entities, and discuss current gaps in the field.
Collapse
|
12
|
Dwivedi M, Bajpai K. The chamber of secretome in Mycobacterium tuberculosis as a potential therapeutic target. Biotechnol Genet Eng Rev 2022; 39:1-44. [PMID: 35613080 DOI: 10.1080/02648725.2022.2076031] [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: 11/02/2022]
Abstract
Mycobacterium tuberculosis (MTB) causes one of the ancient diseases, Tuberculosis, affects people around the globe and its severity can be understood by its classification as a second infectious disease after COVID-19 and the 13th leading cause of death according to a WHO report. Despite having advanced diagnostic approaches and therapeutic strategies, unfortunately, TB is still spreading across the population due to the emergence of drug-resistance MTB and Latent TB infection (LTBI). We are seeking for effective approaches to overcome these hindrances and efficient treatment for this perilous disease. Therefore, there is an urgent need to develop drugs based on operative targeting of the bacterial system that could result in both efficient treatment and lesser emergence of MDR-TB. One such promising target could be the secretory systems and especially the Type 7 secretory system (T7SS-ESX) of Mycobacterium tuberculosis, which is crucial for the secretion of effector proteins as well as in establishing host-pathogen interactions of the tubercle bacilli. The five paralogous ESX systems (ESX-1 to EXS-5) have been observed by in silico genome analysis of MTB, among which ESX-1 and ESX-5 are substantial for virulence and mediating host cellular inflammasome. The bacterium growth and virulence can be modulated by targeting the T7SS. In the present review, we demonstrate the current status of therapeutics against MTB and focus on the function and cruciality of T7SS along with other secretory systems as a promising therapeutic target against Tuberculosis.
Collapse
Affiliation(s)
- Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Kriti Bajpai
- Department of Biotechnology, Himachal Pradesh University, Shimla, India
| |
Collapse
|
13
|
Rastogi S, Briken V. Interaction of Mycobacteria With Host Cell Inflammasomes. Front Immunol 2022; 13:791136. [PMID: 35237260 PMCID: PMC8882646 DOI: 10.3389/fimmu.2022.791136] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
The inflammasome complex is important for host defense against intracellular bacterial infections. Mycobacterium tuberculosis (Mtb) is a facultative intracellular bacterium which is able to survive in infected macrophages. Here we discuss how the host cell inflammasomes sense Mtb and other related mycobacterial species. Furthermore, we describe the molecular mechanisms of NLRP3 inflammasome sensing of Mtb which involve the type VII secretion system ESX-1, cell surface lipids (TDM/TDB), secreted effector proteins (LpqH, PPE13, EST12, EsxA) and double-stranded RNA acting on the priming and/or activation steps of inflammasome activation. In contrast, Mtb also mediates inhibition of the NLRP3 inflammasome by limiting exposure of cell surface ligands via its hydrolase, Hip1, by inhibiting the host cell cathepsin G protease via the secreted Mtb effector Rv3364c and finally, by limiting intracellular triggers (K+ and Cl- efflux and cytosolic reactive oxygen species production) via its serine/threonine kinase PknF. In addition, Mtb inhibits the AIM2 inflammasome activation via an unknown mechanism. Overall, there is good evidence for a tug-of-war between Mtb trying to limit inflammasome activation and the host cell trying to sense Mtb and activate the inflammasome. The detailed molecular mechanisms and the importance of inflammasome activation for virulence of Mtb or host susceptibility have not been fully investigated.
Collapse
Affiliation(s)
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| |
Collapse
|
14
|
The Mycobacterium tuberculosis PE_PGRS Protein Family Acts as an Immunological Decoy to Subvert Host Immune Response. Int J Mol Sci 2022; 23:ijms23010525. [PMID: 35008950 PMCID: PMC8745494 DOI: 10.3390/ijms23010525] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 02/04/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) is a successful pathogen that can reside within the alveolar macrophages of the host and can survive in a latent stage. The pathogen has evolved and developed multiple strategies to resist the host immune responses. M.tb escapes from host macrophage through evasion or subversion of immune effector functions. M.tb genome codes for PE/PPE/PE_PGRS proteins, which are intrinsically disordered, redundant and antigenic in nature. These proteins perform multiple functions that intensify the virulence competence of M.tb majorly by modulating immune responses, thereby affecting immune mediated clearance of the pathogen. The highly repetitive, redundant and antigenic nature of PE/PPE/PE_PGRS proteins provide a critical edge over other M.tb proteins in terms of imparting a higher level of virulence and also as a decoy molecule that masks the effect of effector molecules, thereby modulating immuno-surveillance. An understanding of how these proteins subvert the host immunological machinery may add to the current knowledge about M.tb virulence and pathogenesis. This can help in redirecting our strategies for tackling M.tb infections.
Collapse
|
15
|
Anna F, Lopez J, Moncoq F, Blanc C, Authié P, Noirat A, Fert I, Souque P, Nevo F, Pawlik A, Hardy D, Goyard S, Hudrisier D, Brosch R, Guinet F, Neyrolles O, Charneau P, Majlessi L. A lentiviral vector expressing a dendritic cell-targeting multimer induces mucosal anti-mycobacterial CD4 + T-cell immunity. Mucosal Immunol 2022; 15:1389-1404. [PMID: 36104497 PMCID: PMC9473479 DOI: 10.1038/s41385-022-00566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
Most viral vectors, including the potently immunogenic lentiviral vectors (LVs), only poorly direct antigens to the MHC-II endosomal pathway and elicit CD4+ T cells. We developed a new generation of LVs encoding antigen-bearing monomers of collectins substituted at their C-terminal domain with the CD40 ligand ectodomain to target and activate antigen-presenting cells. Host cells transduced with such optimized LVs secreted soluble collectin-antigen polymers with the potential to be endocytosed in vivo and reach the MHC-II pathway. In the murine tuberculosis model, such LVs induced efficient MHC-II antigenic presentation and triggered both CD8+ and CD4+ T cells at the systemic and mucosal levels. They also conferred a significant booster effect, consistent with the importance of CD4+ T cells for protection against Mycobacterium tuberculosis. Given the pivotal role of CD4+ T cells in orchestrating innate and adaptive immunity, this strategy could have a broad range of applications in the vaccinology field.
Collapse
Affiliation(s)
- François Anna
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Jodie Lopez
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Fanny Moncoq
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Catherine Blanc
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Pierre Authié
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Amandine Noirat
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Ingrid Fert
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Philippe Souque
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Fabien Nevo
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Alexandre Pawlik
- grid.428999.70000 0001 2353 6535Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - David Hardy
- grid.428999.70000 0001 2353 6535Histopathology Platform, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Sophie Goyard
- grid.428999.70000 0001 2353 6535Platform for Innovation and Development of Diagnostic Tests, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Denis Hudrisier
- grid.508721.9Institut de Pharmacologie et de Biologie Structurale, IPBS, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Roland Brosch
- grid.428999.70000 0001 2353 6535Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - Françoise Guinet
- grid.428999.70000 0001 2353 6535Lymphocytes and Immunity Unit, INSERM U1223, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - Olivier Neyrolles
- grid.508721.9Institut de Pharmacologie et de Biologie Structurale, IPBS, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Pierre Charneau
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Laleh Majlessi
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| |
Collapse
|
16
|
Das S, Jain S, Ilyas M, Anand A, Kumar S, Sharma N, Singh K, Mahlawat R, Sharma TK, Atmakuri K. Development of DNA Aptamers to Visualize Release of Mycobacterial Membrane-Derived Extracellular Vesicles in Infected Macrophages. Pharmaceuticals (Basel) 2021; 15:ph15010045. [PMID: 35056102 PMCID: PMC8779091 DOI: 10.3390/ph15010045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged into a novel vaccine platform, a biomarker and a nano-carrier for approved drugs. Their accurate detection and visualization are central to their utility in varied biomedical fields. Owing to the limitations of fluorescent dyes and antibodies, here, we describe DNA aptamer as a promising tool for visualizing mycobacterial EVs in vitro. Employing SELEX from a large DNA aptamer library, we identified a best-performing aptamer that is highly specific and binds at nanomolar affinity to EVs derived from three diverse mycobacterial strains (pathogenic, attenuated and avirulent). Confocal microscopy revealed that this aptamer was not only bound to in vitro-enriched mycobacterial EVs but also detected EVs that were internalized by THP-1 macrophages and released by infecting mycobacteria. To the best of our knowledge, this is the first study that detects EVs released by mycobacteria during infection in host macrophages. Within 4 h, most released mycobacterial EVs spread to other parts of the host cell. We predict that this tool will soon hold huge potential in not only delineating mycobacterial EVs-driven pathogenic functions but also in harboring immense propensity to act as a non-invasive diagnostic tool against tuberculosis in general, and extra-pulmonary tuberculosis in particular.
Collapse
Affiliation(s)
- Soonjyoti Das
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
| | - Sapna Jain
- Bacterial Pathogenesis Laboratory, Infection and Immunology Group, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.J.); (M.I.); (S.K.)
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, Delhi, India
| | - Mohd Ilyas
- Bacterial Pathogenesis Laboratory, Infection and Immunology Group, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.J.); (M.I.); (S.K.)
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, Delhi, India
| | - Anjali Anand
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
| | - Saurabh Kumar
- Bacterial Pathogenesis Laboratory, Infection and Immunology Group, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.J.); (M.I.); (S.K.)
| | - Nishant Sharma
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Kuljit Singh
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 18001, Jammu and Kashmir, India
| | - Rahul Mahlawat
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
| | - Tarun Kumar Sharma
- Aptamer Technology and Diagnostics Laboratory (ATDL), Multidisciplinary Clinical and Translational Research Group (MCTR), Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.D.); (A.A.); (N.S.); (K.S.); (R.M.)
- Correspondence: (T.K.S.); (K.A.)
| | - Krishnamohan Atmakuri
- Bacterial Pathogenesis Laboratory, Infection and Immunology Group, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India; (S.J.); (M.I.); (S.K.)
- Correspondence: (T.K.S.); (K.A.)
| |
Collapse
|
17
|
Lagune M, Petit C, Sotomayor FV, Johansen MD, Beckham KSH, Ritter C, Girard-Misguich F, Wilmanns M, Kremer L, Maurer FP, Herrmann JL. Conserved and specialized functions of Type VII secretion systems in non-tuberculous mycobacteria. MICROBIOLOGY-SGM 2021; 167. [PMID: 34224347 DOI: 10.1099/mic.0.001054] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-tuberculous mycobacteria (NTM) are a large group of micro-organisms comprising more than 200 individual species. Most NTM are saprophytic organisms and are found mainly in terrestrial and aquatic environments. In recent years, NTM have been increasingly associated with infections in both immunocompetent and immunocompromised individuals, prompting significant efforts to understand the diverse pathogenic and signalling traits of these emerging pathogens. Since the discovery of Type VII secretion systems (T7SS), there have been significant developments regarding the role of these complex systems in mycobacteria. These specialised systems, also known as Early Antigenic Secretion (ESX) systems, are employed to secrete proteins across the inner membrane. They also play an essential role in virulence, nutrient uptake and conjugation. Our understanding of T7SS in mycobacteria has significantly benefited over the last few years, from the resolution of ESX-3 structure in Mycobacterium smegmatis, to ESX-5 structures in Mycobacterium xenopi and Mycobacterium tuberculosis. In addition, ESX-4, considered until recently as a non-functional system in both pathogenic and non-pathogenic mycobacteria, has been proposed to play an important role in the virulence of Mycobacterium abscessus; an increasingly recognized opportunistic NTM causing severe lung diseases. These major findings have led to important new insights into the functional mechanisms of these biological systems, their implication in virulence, nutrient acquisitions and cell wall shaping, and will be discussed in this review.
Collapse
Affiliation(s)
- Marion Lagune
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, 78180, Montigny-Le-Bretonneux, France
| | - Cecile Petit
- European Molecular Biology Laboratory, Hamburg Unit, Notkestraße 85, 22607 Hamburg, Germany
| | - Flor Vásquez Sotomayor
- National and WHO Supranational Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Matt D Johansen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France.,Present address: Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Kathrine S H Beckham
- European Molecular Biology Laboratory, Hamburg Unit, Notkestraße 85, 22607 Hamburg, Germany
| | - Christina Ritter
- European Molecular Biology Laboratory, Hamburg Unit, Notkestraße 85, 22607 Hamburg, Germany
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, 78180, Montigny-Le-Bretonneux, France
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Notkestraße 85, 22607 Hamburg, Germany.,University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France.,INSERM, IRIM, 34293 Montpellier, France
| | - Florian P Maurer
- National and WHO Supranational Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Germany.,Institute of Medical Microbiology, Virology and Hospital Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, Inserm, Infection et inflammation, 78180, Montigny-Le-Bretonneux, France.,APHP, GHU Paris-Saclay, Hôpital Raymond Poincaré, Service de Microbiologie, Garches, France
| |
Collapse
|
18
|
Li Z, Hu J, Liu P, Cui D, Di H, Wu S. Microarray-based selection of a serum biomarker panel that can discriminate between latent and active pulmonary TB. Sci Rep 2021; 11:14516. [PMID: 34267288 PMCID: PMC8282789 DOI: 10.1038/s41598-021-93893-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Bacterial culture of M. tuberculosis (MTB), the causative agent of tuberculosis (TB), from clinical specimens is the gold standard for laboratory diagnosis of TB, but is slow and culture-negative TB cases are common. Alternative immune-based and molecular approaches have been developed, but cannot discriminate between active TB (ATB) and latent TB (LTBI). Here, to identify biomarkers that can discriminate between ATB and LTBI/healthy individuals (HC), we profiled 116 serum samples (HC, LTBI and ATB) using a protein microarray containing 257 MTB secreted proteins, identifying 23 antibodies against MTB antigens that were present at significantly higher levels in patients with ATB than in those with LTBI and HC (Fold change > 1.2; p < 0.05). A 4-protein biomarker panel (Rv0934, Rv3881c, Rv1860 and Rv1827), optimized using SAM and ROC analysis, had a sensitivity of 67.3% and specificity of 91.2% for distinguishing ATB from LTBI, and 71.2% sensitivity and 96.3% specificity for distinguishing ATB from HC. Validation of the four candidate biomarkers in ELISA assays using 440 serum samples gave consistent results. The promising sensitivity and specificity of this biomarker panel suggest it merits further investigation for its potential as a diagnostic for discriminating between latent and active TB.
Collapse
Affiliation(s)
- Zhihui Li
- Hebei Chest Hospital, Shijiazhuang, 050041, China
| | - Jianjun Hu
- Hebei Chest Hospital, Shijiazhuang, 050041, China
| | | | - Dan Cui
- Hebei Chest Hospital, Shijiazhuang, 050041, China
| | - Hongqin Di
- Hebei Chest Hospital, Shijiazhuang, 050041, China
| | - Shucai Wu
- Hebei Chest Hospital, Shijiazhuang, 050041, China.
| |
Collapse
|
19
|
Abstract
Current models of horizontal gene transfer (HGT) in mycobacteria are based on “distributive conjugal transfer” (DCT), an HGT type described in the fast-growing, saprophytic model organism Mycobacterium smegmatis, which creates genome mosaicism in resulting strains and depends on an ESX-1 type VII secretion system. In contrast, only few data on interstrain DNA transfer are available for tuberculosis-causing mycobacteria, for which chromosomal DNA transfer between two Mycobacterium canettii strains was reported, a process which, however, was not observed for Mycobacterium tuberculosis strains. Here, we have studied a wide range of human- and animal-adapted members of the Mycobacterium tuberculosis complex (MTBC) using an optimized filter-based mating assay together with three selected strains of M. canettii that acted as DNA recipients. Unlike in previous approaches, we obtained a high yield of thousands of recombinants containing transferred chromosomal DNA fragments from various MTBC donor strains, as confirmed by whole-genome sequence analysis of 38 randomly selected clones. While the genome organizations of the obtained recombinants showed mosaicisms of donor DNA fragments randomly integrated into a recipient genome backbone, reminiscent of those described as being the result of ESX-1-mediated DCT in M. smegmatis, we observed similar transfer efficiencies when ESX-1-deficient donor and/or recipient mutants were used, arguing that in tubercle bacilli, HGT is an ESX-1-independent process. These findings provide new insights into the genetic events driving the pathoevolution of M. tuberculosis and radically change our perception of HGT in mycobacteria, particularly for those species that show recombinogenic population structures despite the natural absence of ESX-1 secretion systems.
Collapse
|
20
|
Fevereiro J, Fraga AG, Pedrosa J. Genetics in the Host-Mycobacterium ulcerans interaction. Immunol Rev 2021; 301:222-241. [PMID: 33682158 DOI: 10.1111/imr.12958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 11/30/2022]
Abstract
Buruli ulcer is an emerging infectious disease associated with high morbidity and unpredictable outbreaks. It is caused by Mycobacterium ulcerans, a slow-growing pathogen evolutionarily shaped by the acquisition of a plasmid involved in the production of a potent macrolide-like cytotoxin and by genome rearrangements and downsizing. These events culminated in an uncommon infection pattern, whereby M. ulcerans is both able to induce the initiation of the inflammatory cascade and the cell death of its proponents, as well as to survive within the phagosome and in the extracellular milieu. In such extreme conditions, the host is sentenced to rely on a highly orchestrated genetic landscape to be able to control the infection. We here revisit the dynamics of M. ulcerans infection, drawing parallels from other mycobacterioses and integrating the most recent knowledge on its evolution and pathogenicity in its interaction with the host immune response.
Collapse
Affiliation(s)
- João Fevereiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra G Fraga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
21
|
Pathogenic Determinants of the Mycobacterium kansasii Complex: An Unsuspected Role for Distributive Conjugal Transfer. Microorganisms 2021; 9:microorganisms9020348. [PMID: 33578772 PMCID: PMC7916490 DOI: 10.3390/microorganisms9020348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
The Mycobacterium kansasii species comprises six subtypes that were recently classified into six closely related species; Mycobacterium kansasii (formerly M. kansasii subtype 1), Mycobacterium persicum (subtype 2), Mycobacterium pseudokansasii (subtype 3), Mycobacterium ostraviense (subtype 4), Mycobacterium innocens (subtype 5) and Mycobacterium attenuatum (subtype 6). Together with Mycobacterium gastri, they form the M. kansasii complex. M. kansasii is the most frequent and most pathogenic species of the complex. M. persicum is classically associated with diseases in immunosuppressed patients, and the other species are mostly colonizers, and are only very rarely reported in ill patients. Comparative genomics was used to assess the genetic determinants leading to the pathogenicity of members of the M. kansasii complex. The genomes of 51 isolates collected from patients with and without disease were sequenced and compared with 24 publicly available genomes. The pathogenicity of each isolate was determined based on the clinical records or public metadata. A comparative genomic analysis showed that all M. persicum, M. ostraviense, M innocens and M. gastri isolates lacked the ESX-1-associated EspACD locus that is thought to play a crucial role in the pathogenicity of M. tuberculosis and other non-tuberculous mycobacteria. Furthermore, M. kansasii was the only species exhibiting a 25-Kb-large genomic island encoding for 17 type-VII secretion system-associated proteins. Finally, a genome-wide association analysis revealed that two consecutive genes encoding a hemerythrin-like protein and a nitroreductase-like protein were significantly associated with pathogenicity. These two genes may be involved in the resistance to reactive oxygen and nitrogen species, a required mechanism for the intracellular survival of bacteria. Three non-pathogenic M. kansasii lacked these genes likely due to two distinct distributive conjugal transfers (DCTs) between M. attenuatum and M. kansasii, and one DCT between M. persicum and M. kansasii. To our knowledge, this is the first study linking DCT to reduced pathogenicity.
Collapse
|
22
|
Qian J, Chen R, Wang H, Zhang X. Role of the PE/PPE Family in Host-Pathogen Interactions and Prospects for Anti-Tuberculosis Vaccine and Diagnostic Tool Design. Front Cell Infect Microbiol 2020; 10:594288. [PMID: 33324577 PMCID: PMC7726347 DOI: 10.3389/fcimb.2020.594288] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
The pe/ppe genes are found in pathogenic, slow-growing Mycobacterium tuberculosis and other M. tuberculosis complex (MTBC) species. These genes are considered key factors in host-pathogen interactions. Although the function of most PE/PPE family proteins remains unclear, accumulating evidence suggests that this family is involved in M. tuberculosis infection. Here, we review the role of PE/PPE proteins, which are believed to be linked to the ESX system function. Further, we highlight the reported functions of PE/PPE proteins, including their roles in host cell interaction, immune response regulation, and cell fate determination during complex host-pathogen processes. Finally, we propose future directions for PE/PPE protein research and consider how the current knowledge might be applied to design more specific diagnostics and effective vaccines for global tuberculosis control.
Collapse
Affiliation(s)
- Jianing Qian
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Run Chen
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Augenstreich J, Briken V. Host Cell Targets of Released Lipid and Secreted Protein Effectors of Mycobacterium tuberculosis. Front Cell Infect Microbiol 2020; 10:595029. [PMID: 33194845 PMCID: PMC7644814 DOI: 10.3389/fcimb.2020.595029] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a very successful pathogen, strictly adapted to humans and the cause of tuberculosis. Its success is associated with its ability to inhibit host cell intrinsic immune responses by using an arsenal of virulence factors of different nature. It has evolved to synthesize a series of complex lipids which form an outer membrane and may also be released to enter host cell membranes. In addition, secreted protein effectors of Mtb are entering the host cell cytosol to interact with host cell proteins. We briefly discuss the current model, involving the ESX-1 type seven secretion system and the Mtb lipid phthiocerol dimycoserosate (PDIM), of how Mtb creates pores in the phagosomal membrane to allow Mtb proteins to access to the host cell cytosol. We provide an exhaustive list of Mtb secreted proteins that have effector functions. They modify (mostly inhibit but sometimes activate) host cell pathways such as: phagosome maturation, cell death, cytokine response, xenophagy, reactive oxygen species (ROS) response via NADPH oxidase 2 (NOX2), nitric oxide (NO) response via NO Synthase 2 (NOS2) and antigen presentation via MHC class I and class II molecules. We discuss the host cell targets for each lipid and protein effector and the importance of the Mtb effector for virulence of the bacterium.
Collapse
Affiliation(s)
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| |
Collapse
|
24
|
Bunduc CM, Bitter W, Houben E. Structure and Function of the Mycobacterial Type VII Secretion Systems. Annu Rev Microbiol 2020; 74:315-335. [DOI: 10.1146/annurev-micro-012420-081657] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bacteria have evolved intricate secretion machineries for the successful delivery of large molecules across their cell envelopes. Such specialized secretion systems allow a variety of bacteria to thrive in specific host environments. In mycobacteria, type VII secretion systems (T7SSs) are dedicated protein transport machineries that fulfill diverse and crucial roles, ranging from metabolite uptake to immune evasion and subversion to conjugation. Since the discovery of mycobacterial T7SSs about 15 y ago, genetic, structural, and functional studies have provided insight into the roles and functioning of these secretion machineries. Here, we focus on recent advances in the elucidation of the structure and mechanism of mycobacterial T7SSs in protein secretion. As many of these systems are essential for mycobacterial growth or virulence, they provide opportunities for the development of novel therapies to combat a number of relevant mycobacterial diseases.
Collapse
Affiliation(s)
- Catalin M. Bunduc
- Section of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - W. Bitter
- Section of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Department of Medical Microbiology and Infection Control, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, 1007 MB Amsterdam, The Netherlands
| | - E.N.G. Houben
- Section of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
25
|
Potential Plasticity of the Mannoprotein Repertoire Associated to Mycobacterium tuberculosis Virulence Unveiled by Mass Spectrometry-Based Glycoproteomics. Molecules 2020; 25:molecules25102348. [PMID: 32443484 PMCID: PMC7287972 DOI: 10.3390/molecules25102348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/04/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
To date, Mycobacterium tuberculosis (Mtb) remains the world’s greatest infectious killer. The rise of multidrug-resistant strains stresses the need to identify new therapeutic targets to fight the epidemic. We previously demonstrated that bacterial protein-O-mannosylation is crucial for Mtb infectiousness, renewing the interest of the bacterial-secreted mannoproteins as potential drug-targetable virulence factors. The difficulty of inventorying the mannoprotein repertoire expressed by Mtb led us to design a stringent multi-step workflow for the reliable identification of glycosylated peptides by large-scale mass spectrometry-based proteomics. Applied to the differential analyses of glycoproteins secreted by the wild-type Mtb strain—and by its derived mutant invalidated for the protein-O-mannosylating enzyme PMTub—this approach led to the identification of not only most already known mannoproteins, but also of yet-unknown mannosylated proteins. In addition, analysis of the glycoproteome expressed by the isogenic recombinant Mtb strain overexpressing the PMTub gene revealed an unexpected mannosylation of proteins, with predicted or demonstrated functions in Mtb growth and interaction with the host cell. Since in parallel, a transient increased expression of the PMTub gene has been observed in the wild-type bacilli when infecting macrophages, our results strongly suggest that the Mtb mannoproteome may undergo adaptive regulation during infection of the host cells. Overall, our results provide deeper insights into the complexity of the repertoire of mannosylated proteins expressed by Mtb, and open the way to novel opportunities to search for still-unexploited potential therapeutic targets.
Collapse
|
26
|
Guan Q, Ummels R, Ben-Rached F, Alzahid Y, Amini MS, Adroub SA, van Ingen J, Bitter W, Abdallah AM, Pain A. Comparative Genomic and Transcriptomic Analyses of Mycobacterium kansasii Subtypes Provide New Insights Into Their Pathogenicity and Taxonomy. Front Cell Infect Microbiol 2020; 10:122. [PMID: 32266172 PMCID: PMC7105574 DOI: 10.3389/fcimb.2020.00122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/04/2020] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium kansasii is an important opportunistic pathogen of humans and has a close phylogenetic relationship with Mycobacterium tuberculosis. Seven subtypes (I-VII) have been identified using molecular biology approaches, of which subtype I is the most frequent causative agent of human disease. To investigate the genotypes and pathogenic components of M. kansasii, we sequenced and compared the complete base-perfect genomes of different M. kansasii subtypes. Our findings support the proposition that M. kansasii "subtypes" I-VI, whose assemblies are currently available, should be considered as different species. Furthermore, we identified the exclusive presence of the espACD operon in M. kansasii subtype I, and we confirmed its role in the pathogenicity of M. kansasii in a cell infection model. The espACD operon is exclusively present in mycobacterial species that induce phagosomal rupture in host phagocytes and is known to be a major determinant of ESX1-mediated virulence in pathogenic mycobacteria. Comparative transcriptome analysis of the M. kansasii I-V strains identified genes potentially associated with virulence. Using a comparative genomics approach, we designed primers for PCR genotyping of M. kansasii subtypes I-V and tested their efficacy using clinically relevant strains of M. kansasii.
Collapse
Affiliation(s)
- Qingtian Guan
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Roy Ummels
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Fathia Ben-Rached
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Alzahid
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Mohammad S. Amini
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sabir A. Adroub
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboud UMC Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Abdallah M. Abdallah
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Arnab Pain
- Pathogen Genomics Laboratory, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Center for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Riaz SM, Bjune GA, Wiker HG, Sviland L, Mustafa T. Mycobacterial antigens accumulation in foamy macrophages in murine pulmonary tuberculosis lesions: Association with necrosis and making of cavities. Scand J Immunol 2020; 91:e12866. [PMID: 31960452 DOI: 10.1111/sji.12866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/06/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022]
Abstract
Understanding mechanisms of cavitation in tuberculosis (TB) is the missing link that could advance the field towards better control of the infection. Descriptions of human TB suggest that postprimary TB begins as lipid pneumonia of foamy macrophages that undergoes caseating necrosis and fragmentation to produce cavities. This study aimed to investigate the various mycobacterial antigens accumulating in foamy macrophages and their relation to tissue destruction and necrosis. Pulmonary tissues from mice with slowly progressive TB were studied for histopathology, acid-fast bacilli (AFB) and presence of mycobacterial antigens. Digital quantification using Aperio ImageScope was done. Until week 12 postinfection, mice were healthy, and lesions were small with scarce AFB and mycobacterial antigens. Colony-forming units (CFUs) increased exponentially. At week 16-33, mice were sick, macrophages attained foamy appearance with an increase in antigens (P < .05), 1.5 log increase in CFUs and an approximately onefold increase in AFB. At week 37-41, mice started dying with a shift in morphology towards necrosis. A >20-fold increase in mycobacterial antigens was observed with only less than one log increase in CFUs and sevenfold increase in AFB. Secreted antigens were significantly (P < .05) higher compared to cell-wall antigens throughout infection. Focal areas of necrosis were associated with an approximately 40-fold increase in antigen MPT46, functionally active thioredoxin, and a significant increase in all secreted antigens. In conclusion, mycobacterial antigens accumulate in the foamy macrophages in TB lesions during slowly progressive murine pulmonary TB. Secreted antigens and MPT46 correlated with necrosis, thereby implying that they might trigger the formation of cavities.
Collapse
Affiliation(s)
- Syeda Mariam Riaz
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Gunnar Aksel Bjune
- Department of Community Medicine, Institute of Health and Society, The Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Harald G Wiker
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lisbet Sviland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Tehmina Mustafa
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
28
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
29
|
Behra PRK, Pettersson BMF, Ramesh M, Dasgupta S, Kirsebom LA. Insight into the biology of Mycobacterium mucogenicum and Mycobacterium neoaurum clade members. Sci Rep 2019; 9:19259. [PMID: 31848383 PMCID: PMC6917791 DOI: 10.1038/s41598-019-55464-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 11/09/2022] Open
Abstract
Nontuberculous mycobacteria, NTM, are of growing concern and among these members of the Mycobacterium mucogenicum (Mmuc) and Mycobacterium neoaurum (Mneo) clades can cause infections in humans and they are resistant to first-line anti-tuberculosis drugs. They can be isolated from different ecological niches such as soil, tap water and ground water. Mycobacteria, such as Mmuc and Mneo, are classified as rapid growing mycobacteria, RGM, while the most familiar, Mycobacterium tuberculosis, belongs to the slow growing mycobacteria, SGM. Modern “omics” approaches have provided new insights into our understanding of the biology and evolution of this group of bacteria. Here we present comparative genomics data for seventeen NTM of which sixteen belong to the Mmuc- and Mneo-clades. Focusing on virulence genes, including genes encoding sigma/anti-sigma factors, serine threonine protein kinases (STPK), type VII (ESX genes) secretion systems and mammalian cell entry (Mce) factors we provide insight into their presence as well as phylogenetic relationship in the case of the sigma/anti-sigma factors and STPKs. Our data further suggest that these NTM lack ESX-5 and Mce2 genes, which are known to affect virulence. In this context, Mmuc- and Mneo-clade members lack several of the genes in the glycopeptidolipid (GLP) locus, which have roles in colony morphotype appearance and virulence. For the M. mucogenicum type strain, MmucT, we provide RNASeq data focusing on mRNA levels for sigma factors, STPK, ESX proteins and Mce proteins. These data are discussed and compared to in particular the SGM and fish pathogen Mycobacterium marinum. Finally, we provide insight into as to why members of the Mmuc- and Mneo-clades show resistance to rifampin and isoniazid, and why MmucT forms a rough colony morphotype.
Collapse
Affiliation(s)
- Phani Rama Krishna Behra
- Department of Cell and Molecular Biology, Box 596, BMC, Uppsala University, SE 751 24, Uppsala, Sweden
| | - B M Fredrik Pettersson
- Department of Cell and Molecular Biology, Box 596, BMC, Uppsala University, SE 751 24, Uppsala, Sweden
| | - Malavika Ramesh
- Department of Cell and Molecular Biology, Box 596, BMC, Uppsala University, SE 751 24, Uppsala, Sweden
| | - Santanu Dasgupta
- Department of Cell and Molecular Biology, Box 596, BMC, Uppsala University, SE 751 24, Uppsala, Sweden
| | - Leif A Kirsebom
- Department of Cell and Molecular Biology, Box 596, BMC, Uppsala University, SE 751 24, Uppsala, Sweden.
| |
Collapse
|
30
|
Kroesen VM, Madacki J, Frigui W, Sayes F, Brosch R. Mycobacterial virulence: impact on immunogenicity and vaccine research. F1000Res 2019; 8. [PMID: 32047597 PMCID: PMC6979476 DOI: 10.12688/f1000research.20572.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
The borderline between virulence and efficacy in live attenuated vaccine strains is often blurred and this is also the case for the Bacillus Calmette–Guérin (BCG), the only currently licensed anti-tuberculosis vaccine used on a large, global scale, which was obtained almost 100 years ago. While BCG is more than 99% identical at the genome level to
Mycobacterium tuberculosis, the causative pathogen of human tuberculosis, some important differences in virulence factors cause naturally irreversible attenuation and safety of this vaccine in the immunocompetent host. Some of these virulence factors are involved in persistence capacities of the vaccine strains and also represent strong immunogens, responsible for inducing different host signaling pathways, which have to be taken into consideration for the development of revised and new vaccine strains. Here we discuss a number of selected mycobacterial features in relation to their biological functions and potential impact on virulence and vaccine efficacy.
Collapse
Affiliation(s)
- Vera M Kroesen
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France.,Faculty VI, University of Oldenburg, Oldenburg, Germany
| | - Jan Madacki
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Wafa Frigui
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Fadel Sayes
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| |
Collapse
|
31
|
Multiplexed Quantitation of Intraphagocyte Mycobacterium tuberculosis Secreted Protein Effectors. Cell Rep 2019; 23:1072-1084. [PMID: 29694886 PMCID: PMC5946722 DOI: 10.1016/j.celrep.2018.03.125] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/16/2018] [Accepted: 03/26/2018] [Indexed: 01/21/2023] Open
Abstract
The pathogenic potential of Mycobacterium tuberculosis largely depends on ESX secretion systems exporting members of the multigenic Esx, Esp, and PE/PPE protein families. To study the secretion and regulation patterns of these proteins while circumventing immune cross-reactions due to their extensive sequence homologies, we developed an approach that relies on the recognition of their MHC class II epitopes by highly discriminative T cell receptors (TCRs) of a panel of T cell hybridomas. The latter were engineered so that each expresses a unique fluorescent reporter linked to specific antigen recognition. The resulting polychromatic and multiplexed imaging assay enabled us to measure the secretion of mycobacterial effectors inside infected host cells. We applied this novel technology to a large panel of mutants, clinical isolates, and host-cell types to explore the host-mycobacteria interplay and its impact on the intracellular bacterial secretome, which also revealed the unexpected capacity of phagocytes from lung granuloma to present mycobacterial antigens via MHC class II. T cell hybridomas detect individual mycobacterial proteins without cross-reactivity Detection of mycobacterial proteins by T cells allows visualization of their cellular topography Measurement of intraphagocyte mycobacterial proteins can be performed with T cells A multiplexed assay of mycobacterial protein quantitation has numerous applications
Collapse
|
32
|
Bonilla-Muro MG, Hernández de la Cruz ON, Gonzalez-Barrios JA, Alcaráz-Estrada SL, Castañón-Arreola M. EsxA mainly contributes to the miR-155 overexpression in human monocyte-derived macrophages and potentially affect the immune mechanism of macrophages through miRNA dysregulation. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 54:185-192. [PMID: 31561988 DOI: 10.1016/j.jmii.2019.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 03/24/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND/PURPOSE Mycobacterium tuberculosis is a successful intracellular pathogen that uses multiple proteins to survive within macrophages, one of the most remarkable is the virulence factor EsxA. In this study, we evaluate the participation of EsxA in the miRNAs expression profile of human monocyte-derived macrophages (hMDM), to mapping out the contribution of this virulence factor in the miRNA profile and how these changes can influence and alter immune-related processes and pathways. METHODS The cytotoxic effect of rEsxA on hMDM was evaluated by the neutral red assay. The evaluation of miRNA expression profile in infected and rEsxA-stimulated hMDM was done using TaqMan Low Density Assays, and in silico analyses was carried on to construct Protein-Protein Interaction network of miRNAs targets. RESULTS miR-155 was the only miRNA upregulated consistently in hMDM infected with M. tuberculosis H37Rv or stimulated with rEsxA. In hMDM stimulated with rEsxA, we found 25 miRNA's dysregulated (8 up-regulated and 17 down-regulated). The most significant were the miR-155 and miR-622 that has been observed in the analysis carried out with two different endogenous controls (U6 snRNA and RNU44) for the normalization of expression analysis. This result suggests that rEsxA induces the deregulation of miRNAs that potentially target genes in key pathways for the infection control, like the MAPK signaling pathway, cytokines, and chemokine signaling pathways, and several connected pathways involved in mycobacterial uptake, vesicular traffic, and endosome maturation. CONCLUSION Higher expression levels of miR-155 suggest potential roles of these miRNA in EsxA-dependent immune subversion.
Collapse
Affiliation(s)
| | | | - Juan Antonio Gonzalez-Barrios
- Coordinación de Capacitación, Desarrollo e Investigación, Hospital Regional 1º de Octubre, ISSSTE, Mexico City, Mexico
| | | | | |
Collapse
|
33
|
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]
|
34
|
The Mycobacterium tuberculosis capsule: a cell structure with key implications in pathogenesis. Biochem J 2019; 476:1995-2016. [PMID: 31320388 PMCID: PMC6698057 DOI: 10.1042/bcj20190324] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/17/2023]
Abstract
Bacterial capsules have evolved to be at the forefront of the cell envelope, making them an essential element of bacterial biology. Efforts to understand the Mycobacterium tuberculosis (Mtb) capsule began more than 60 years ago, but the relatively recent development of mycobacterial genetics combined with improved chemical and immunological tools have revealed a more refined view of capsule molecular composition. A glycogen-like α-glucan is the major constituent of the capsule, with lower amounts of arabinomannan and mannan, proteins and lipids. The major Mtb capsular components mediate interactions with phagocytes that favor bacterial survival. Vaccination approaches targeting the mycobacterial capsule have proven successful in controlling bacterial replication. Although the Mtb capsule is composed of polysaccharides of relatively low complexity, the concept of antigenic variability associated with this structure has been suggested by some studies. Understanding how Mtb shapes its envelope during its life cycle is key to developing anti-infective strategies targeting this structure at the host-pathogen interface.
Collapse
|
35
|
Vaziri F, Brosch R. ESX/Type VII Secretion Systems-An Important Way Out for Mycobacterial Proteins. Microbiol Spectr 2019; 7:10.1128/microbiolspec.psib-0029-2019. [PMID: 31298207 PMCID: PMC10957191 DOI: 10.1128/microbiolspec.psib-0029-2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 11/20/2022] Open
Abstract
The causative agent of human tuberculosis, Mycobacterium tuberculosis, has a complex lipid-rich diderm envelope, which acts as a major barrier protecting the bacterium against the hostile environment inside the host cells. For the transfer of diverse molecules across this complex cell envelope, M. tuberculosis has a series of general and specialized protein secretion systems, characterized by the SecA general secretion pathway, the twin-arginine translocation pathway, and five specific ESX type VII secretion systems. In this review, we focus on the latter systems, known as ESX-1 to ESX-5, which were first discovered almost 20 years ago during the in silico analysis of the genome sequence of M. tuberculosis H37Rv. Since then, these systems have been the subject of highly dynamic research due to their involvement in several key biological processes and host-pathogen interactions of the tubercle bacilli.
Collapse
Affiliation(s)
- Farzam Vaziri
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, UMR3525 CNRS, 75015 Paris, France
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, 13164 Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, 13164 Tehran, Iran
| | - Roland Brosch
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, UMR3525 CNRS, 75015 Paris, France
| |
Collapse
|
36
|
A New ESX-1 Substrate in Mycobacterium marinum That Is Required for Hemolysis but Not Host Cell Lysis. J Bacteriol 2019; 201:JB.00760-18. [PMID: 30833360 DOI: 10.1128/jb.00760-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
The ESX-1 (ESAT-6 system 1) secretion system plays a conserved role in the virulence of diverse mycobacterial pathogens, including the human pathogen Mycobacterium tuberculosis and M. marinum, an environmental mycobacterial species. The ESX-1 system promotes the secretion of protein virulence factors to the extracytoplasmic environment. The secretion of these proteins triggers the host response by lysing the phagosome during macrophage infection. Using proteomic analyses of the M. marinum secretome in the presence and absence of a functional ESX-1 system, we and others have hypothesized that MMAR_2894, a PE family protein, is a potential ESX-1 substrate in M. marinum We used genetic and quantitative proteomic approaches to determine if MMAR_2894 is secreted by the ESX-1 system, and we defined the requirement of MMAR_2894 for ESX-1-mediated secretion and virulence. We show that MMAR_2894 is secreted by the ESX-1 system in M. marinum and is itself required for the optimal secretion of the known ESX-1 substrates in M. marinum Moreover, we found that MMAR_2894 was differentially required for hemolysis and cytolysis of macrophages, two lytic activities ascribed to the M. marinum ESX-1 system.IMPORTANCE Both Mycobacterium tuberculosis, the cause of human tuberculosis (TB), and Mycobacterium marinum, a pathogen of ectotherms, use the ESX-1 secretion system to cause disease. There are many established similarities between the ESX-1 systems in M. tuberculosis and in M. marinum Yet the two bacteria infect different hosts, hinting at species-specific functions of the ESX-1 system. Our findings demonstrate that MMAR_2894 is a PE protein secreted by the ESX-1 system of M. marinum We show that MMAR_2894 is required for the optimal secretion of mycobacterial proteins required for disease. Because the MMAR_2894 gene is not conserved in M. tuberculosis, our findings demonstrate that MMAR_2894 may contribute to a species-specific function of the ESX-1 system in M. marinum, providing new insight into how the M. marinum and M. tuberculosis systems differ.
Collapse
|
37
|
Guo Q, Bi J, Li M, Ge W, Xu Y, Fan W, Wang H, Zhang X. ESX Secretion-Associated Protein C From Mycobacterium tuberculosis Induces Macrophage Activation Through the Toll-Like Receptor-4/Mitogen-Activated Protein Kinase Signaling Pathway. Front Cell Infect Microbiol 2019; 9:158. [PMID: 31134163 PMCID: PMC6523024 DOI: 10.3389/fcimb.2019.00158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/26/2019] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis, as a facultative intracellular pathogen, can interact with host macrophages and modulate macrophage function to influence innate and adaptive immunity. Proteins secreted by the ESX-1 secretion system are involved in this relationship. Although the importance of ESX-1 in host-pathogen interactions and virulence is well-known, the primary role is ascribed to EsxA (EAST-6) in mycobacterial pathogenesis and the functions of individual components in the interactions between pathogens and macrophages are still unclear. Here, we investigated the effects of EspC on macrophage activation. The EspC protein is encoded by an espA/C/D cluster, which is not linked to the esx-1 locus, but is essential for the secretion of the major virulence factors of ESX-1, EsxA and EsxB. Our results showed that both EspC protein and EspC overexpression in M. smegmatis induced pro-inflammatory cytokines and enhanced surface marker expression. This mechanism was dependent on Toll-like receptor 4 (TLR4), as demonstrated using EspC-treated macrophages from TLR4-/- mice, leading to decreased pro-inflammatory cytokine secretion and surface marker expression compared with those from wild-type mice. Immunoprecipitation and immunofluorescence assays showed that EspC interacted with TLR4 directly. Moreover, EspC could activate macrophages and promote antigen presentation by inducing mitogen-activated protein kinase (MAPK) phosphorylation and nuclear factor-κB activation. The EspC-induced cytokine expression, surface marker upregulation, and MAPK signaling activation were inhibited when macrophages were blocked with anti-TLR4 antibodies or pretreated with MAPK inhibitors. Furthermore, our results showed that EspC overexpression enhanced the survival of M. smegmatis within macrophages and under stress conditions. Taken together, our results indicated that EspC may be another ESX-1 virulence factor that not only modulates the host innate immune response by activating macrophages through TLR4-dependent MAPK signaling but also plays an important role in the survival of pathogenic mycobacteria in host cells.
Collapse
Affiliation(s)
- Qinglong Guo
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Jing Bi
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ming Li
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Wenxue Ge
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Ying Xu
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Weixing Fan
- Laboratory of Zoonosis, China Animal Health and Epidemiology Center, Qingdao, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Li J, Zhao J, Shen J, Wu C, Liu J. Intranasal immunization with Mycobacterium tuberculosis Rv3615c induces sustained adaptive CD4 + T-cell and antibody responses in the respiratory tract. J Cell Mol Med 2018; 23:596-609. [PMID: 30353641 PMCID: PMC6307849 DOI: 10.1111/jcmm.13965] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/20/2018] [Accepted: 09/23/2018] [Indexed: 02/04/2023] Open
Abstract
Sustained adaptive immunity to pathogens provides effective protection against infections, and effector cells located at the site of infection ensure rapid response to the challenge. Both are essential for the success of vaccine development. To explore new vaccination approach against Mycobacterium tuberculosis (M.tb) infection, we have shown that Rv3615c, identified as ESX-1 substrate protein C of M.tb but not expressed in BCG, induced a dominant Th1-type response of CD4+ T cells from patients with tuberculosis pleurisy, which suggests a potential candidate for vaccine development. But subcutaneous immunization with Rv3615c induced modest T-cell responses systemically, and showed suboptimal protection against virulent M.tb challenge at the site of infection. Here, we use a mouse model to demonstrate that intranasal immunization with Rv3615c induces sustained capability of adaptive CD4+ T- and B-cell responses in lung parenchyma and airway. Rv3615c contains a dominant epitope of mouse CD4+ T cells, Rv3615c41-50 , and elicits CD4+ T-cell response with an effector-memory phenotype and multi-Th1-type cytokine coexpressions. Since T cells resident at mucosal tissue are potent at control of infection at early stage, our data show that intranasal immunization with Rv3615c promotes a sustained regional immunity to M.tb, and suggests a potency in control of M.tb infection. Our study warranties a further investigation of Rv3615c as a candidate for development of effective vaccination against M.tb infection.
Collapse
Affiliation(s)
- Jiangping Li
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Laboratory of Infectious Diseases and Vaccine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Zhao
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Juan Shen
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Changyou Wu
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jie Liu
- Laboratory of Infectious Diseases and Vaccine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Thompson CR, Champion MM, Champion PA. Quantitative N-Terminal Footprinting of Pathogenic Mycobacteria Reveals Differential Protein Acetylation. J Proteome Res 2018; 17:3246-3258. [PMID: 30080413 DOI: 10.1021/acs.jproteome.8b00373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
N-terminal acetylation (NTA) is a post-transcriptional modification of proteins that is conserved from bacteria to humans. In bacteria, the enzymes that mediate protein NTA also promote antimicrobial resistance. In pathogenic mycobacteria, which cause human tuberculosis and other chronic infections, NTA has been linked to pathogenesis and stress response, yet the fundamental biology underlying NTA of mycobacterial proteins remains unclear. We enriched, defined, and quantified the NT-acetylated populations of both cell-associated and secreted proteins from both the human pathogen, Mycobacterium tuberculosis, and the nontuberculous opportunistic pathogen, Mycobacterium marinum. We used a parallel N-terminal enrichment strategy from proteolytic digests coupled to charge-based selection and stable isotope ratio mass spectrometry. We show that NTA of the mycobacterial proteome is abundant, diverse, and primarily on Thr residues, which is unique compared with other bacteria. We isolated both the acetylated and unacetylated forms of 256 proteins, indicating that NTA of mycobacterial proteins is homeostatic. We identified 16 mycobacterial proteins with differential levels of NTA on the cytoplasmic and secreted forms, linking protein modification and localization. Our findings reveal novel biology underlying the NTA of mycobacterial proteins, which may provide a basis to understand NTA in mycobacterial physiology, pathogenesis, and antimicrobial resistance.
Collapse
|
40
|
Mycobacterium tuberculosis Pst/SenX3-RegX3 Regulates Membrane Vesicle Production Independently of ESX-5 Activity. mBio 2018; 9:mBio.00778-18. [PMID: 29895636 PMCID: PMC6016242 DOI: 10.1128/mbio.00778-18] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mycobacterium tuberculosis releases membrane vesicles (MV) that modulate host immune responses and aid in iron acquisition, although they may have additional unappreciated functions. MV production appears to be a regulated process, but virR remains the only characterized genetic regulator of vesiculogenesis. Here, we present data supporting a role for the M. tuberculosis Pst/SenX3-RegX3 signal transduction system in regulating MV production. Deletion of pstA1, which encodes a transmembrane component of the phosphate-specific transport (Pst) system, causes constitutive activation of the SenX3-RegX3 two-component system, leading to increased protein secretion via the specialized ESX-5 type VII secretion system. Using proteomic mass spectrometry, we identified several additional proteins hyper-secreted by the ΔpstA1 mutant, including LpqH, an MV-associated lipoprotein. Nanoparticle tracking analysis revealed a 15-fold increase in MV production by the ΔpstA1 mutant. Both hyper-secretion of LpqH and increased MV release required RegX3 but were independent of VirR, suggesting that Pst/SenX3-RegX3 controls MV release by a novel mechanism. Prior proteomic analysis identified ESX-5 substrates associated with MV. We therefore hypothesized that MV release requires ESX-5 activity. We constructed strains that conditionally express eccD5, which encodes the predicted ESX-5 transmembrane channel. Upon EccD5 depletion, we observed reduced secretion of the ESX-5 substrates EsxN and PPE41, but MV release was unaffected. Our data suggest that ESX-5 does not affect vesicle production and imply that further characterization of the Pst/SenX3-RegX3 regulon might reveal novel mechanisms of M. tuberculosis vesicle biogenesis. In Gram-negative bacteria, MV derived from the outer membrane have diverse functions in bacterial physiology and pathogenesis, and several factors regulating their production have been identified. Though Gram-positive bacteria and mycobacteria that lack an outer membrane also produce vesicles with described roles in pathogenesis, the mechanisms of MV biogenesis in these organisms remain poorly characterized. Defining mechanisms of MV biogenesis might yield significant insights into the importance of MV production during infection. In M. tuberculosis, only a single genetic element, virR, is known to regulate MV production. Our work reveals that the Pst/SenX3-RegX3 signal transduction system is a novel regulator of MV biogenesis that controls MV production by a mechanism that is independent of both VirR and activation of the specialized ESX-5 protein secretion system. Understanding which genes in the RegX3 regulon cause increased MV production might reveal novel molecular mechanisms of MV release.
Collapse
|
41
|
Gcebe N, Michel AL, Hlokwe TM. Non-tuberculous Mycobacterium species causing mycobacteriosis in farmed aquatic animals of South Africa. BMC Microbiol 2018; 18:32. [PMID: 29653505 PMCID: PMC5899368 DOI: 10.1186/s12866-018-1177-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 04/05/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mycobacteriosis caused by non-tuberculous mycobacteria (NTM), is among the most chronic diseases of aquatic animals. In addition, fish mycobacteriosis has substantial economic consequences especially in the aquaculture and fisheries industry as infections may significantly decrease production and trade. Some fish NTM pathogens are highly virulent and zoonotic; as such, infection of aquaria with these pathogens is a public health concern. In this study, we report isolation of nine different NTM species from sixteen aquatic animals including different fish species, frogs and a crocodile. Given the clinical significance of Mycobacterium marinum and its close relation to Mycobacterium tuberculosis, as well as the significance of ESAT 6 and CFP-10 secretion in mycobacterial virulence, we analysed the esxA and esxB nucleotide sequences of M. marinum isolates identified in this study as well as other mycobacteria in the public databases. RESULTS Mycobacterium shimoidei, Mycobacterium marinum, Mycobacterium chelonae, Mycobacterium septicum /M. peregrinum and Mycobacterium porcinum were isolated from gold fish, Guppy, exotic fish species in South Africa, koi and undefined fish, Knysna seahorse, as well Natal ghost frogs respectively, presenting tuberculosis like granuloma. Other NTM species were isolated from the studied aquatic animals without any visible lesions, and these include Mycobacterium sp. N845 T, Mycobacterium fortuitum, a member of the Mycobacterium avium complex, and Mycobacterium szulgai. Phylogenetic analysis of mycobacteria, based on esxA and esxB genes, separated slow growing from rapidly growing mycobacteria as well as pathogenic from non-pathogenic mycobacteria in some cases. CONCLUSIONS Isolation of the different NTM species from samples presenting granuloma suggests the significance of these NTM species in causing mycobacteriosis in these aquatic animals. The study also revealed the potential of esxA and esxB sequences as markers for phylogenetic classification of mycobacteria. Observations regarding use of esxA and esxB sequences for prediction of potential pathogenicity of mycobacteria warrants further investigation of these two genes in a study employing NTM species with well-defined pathogenicity.
Collapse
Affiliation(s)
- Nomakorinte Gcebe
- Tuberculosis Laboratory, Agricultural Research Council - Onderstepoort Veterinary Research, Onderstepoort, South Africa.
| | - Anita L Michel
- Department of Veterinary Tropical Diseases, Bovine Tuberculosis and Brucellosis Research Programme, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - Tiny Motlatso Hlokwe
- Tuberculosis Laboratory, Agricultural Research Council - Onderstepoort Veterinary Research, Onderstepoort, South Africa
| |
Collapse
|
42
|
Donovan ML, Schultz TE, Duke TJ, Blumenthal A. Type I Interferons in the Pathogenesis of Tuberculosis: Molecular Drivers and Immunological Consequences. Front Immunol 2017; 8:1633. [PMID: 29230217 PMCID: PMC5711827 DOI: 10.3389/fimmu.2017.01633] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/09/2017] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) remains a major global health threat. Urgent needs in the fight against TB include improved and innovative treatment options for drug-sensitive and -resistant TB as well as reliable biological indicators that discriminate active from latent disease and enable monitoring of treatment success or failure. Prominent interferon (IFN) inducible gene signatures in TB patients and animal models of Mycobacterium tuberculosis infection have drawn significant attention to the roles of type I IFNs in the host response to mycobacterial infections. Here, we review recent developments in the understanding of the innate immune pathways that drive type I IFN responses in mycobacteria-infected host cells and the functional consequences for the host defense against M. tuberculosis, with a view that such insights might be exploited for the development of targeted host-directed immunotherapies and development of reliable biomarkers.
Collapse
Affiliation(s)
- Meg L Donovan
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Thomas E Schultz
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Taylor J Duke
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
43
|
Recombinant BCG Expressing ESX-1 of Mycobacterium marinum Combines Low Virulence with Cytosolic Immune Signaling and Improved TB Protection. Cell Rep 2017; 18:2752-2765. [PMID: 28297677 DOI: 10.1016/j.celrep.2017.02.057] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/18/2017] [Accepted: 02/16/2017] [Indexed: 12/30/2022] Open
Abstract
Recent insights into the mechanisms by which Mycobacterium tuberculosis, the etiologic agent of human tuberculosis, is recognized by cytosolic nucleotide sensors have opened new avenues for rational vaccine design. The only licensed anti-tuberculosis vaccine, Mycobacterium bovis BCG, provides limited protection. A feature of BCG is the partial deletion of the ESX-1 type VII secretion system, which governs phagosomal rupture and cytosolic pattern recognition, key intracellular phenotypes linked to increased immune signaling. Here, by heterologously expressing the esx-1 region of Mycobacterium marinum in BCG, we engineered a low-virulence, ESX-1-proficient, recombinant BCG (BCG::ESX-1Mmar) that induces the cGas/STING/TBK1/IRF-3/type I interferon axis and enhances AIM2 and NLRP3 inflammasome activity, resulting in both higher proportions of CD8+ T cell effectors against mycobacterial antigens shared with BCG and polyfunctional CD4+ Th1 cells specific to ESX-1 antigens. Importantly, independent mouse vaccination models show that BCG::ESX-1Mmar confers superior protection relative to parental BCG against challenges with highly virulent M. tuberculosis.
Collapse
|
44
|
Abstract
The tuberculosis agent Mycobacterium tuberculosis has undergone a long and selective evolution toward human infection and represents one of the most widely spread pathogens due to its efficient aerosol-mediated human-to-human transmission. With the availability of more and more genome sequences, the evolutionary trajectory of this obligate pathogen becomes visible, which provides us with new insights into the molecular events governing evolution of the bacterium and its ability to accumulate drug-resistance mutations. In this review, we summarize recent developments in mycobacterial research related to this matter that are important for a better understanding of the current situation and future trends and developments in the global epidemiology of tuberculosis, as well as for possible public health intervention possibilities.
Collapse
|
45
|
Abstract
Mycobacterial 6-kDa early secreted antigenic target (ESAT-6) system (ESX) exporters transport proteins across the cytoplasmic membrane. Many proteins transported by ESX systems are then translocated across the mycobacterial cell envelope and secreted from the cell. Although the mechanism underlying protein transport across the mycolate outer membrane remains elusive, the ESX systems are closely connected with and localize to the cell envelope. Links between ESX-associated proteins, cell wall synthesis, and the maintenance of cell envelope integrity have been reported. Genes encoding the ESX systems and those required for biosynthesis of the mycobacterial envelope are coregulated. Here, we review the interplay between ESX systems and the mycobacterial cell envelope.
Collapse
|
46
|
Mycobacterium tuberculosis Rv3615c is a highly immunodominant antigen and specifically induces potent Th1-type immune responses in tuberculosis pleurisy. Clin Sci (Lond) 2017; 131:1859-1876. [PMID: 28588103 DOI: 10.1042/cs20170205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 02/05/2023]
Abstract
T-cell responses have been demonstrated to be essential for preventing Mycobacterium tuberculosis infection. The Th1-cytokines produced by T cells, such as INF-γ, IL-2, and TNF-α, not only limit the invasion of M. tuberculosis but also eliminate the pathogen at the site of infection. Bacillus Calmette-Guérin (BCG) is known to induce Th1-type responses but the protection is inadequate. Identification of immunogenic components, in addition to those expressed in BCG, and induction of a broad spectrum of Th1-type responses provide options for generating sufficient adaptive immunity. Here, we studied human pulmonary T-cell responses induced by the M. tuberculosis-specific antigen Rv3615c, a protein with a similar size and sequence homology to ESAT-6 and CFP-10, which induced dominant CD4+ T-cell responses in human tuberculosis (TB) models. We characterized T-cell responses including cytokine profiling, kinetics of activation, expansion, differentiation, TCR usage, and signaling of activation induced by Rv3615c compared with other M. tuberculosis-specific antigens. The expanded CD4+ T cells induced by Rv3615c predominately produced Th1, but less Th2 and Th17, cytokines and displayed effector/memory phenotypes (CD45RO+CD27-CD127-CCR7-). The magnitude of expansion and cytokine production was comparable to those induced by well-characterized the 6 kDa early secreted antigenic target (ESAT-6), the 10 kDa culture filtrate protein (CFP-10) and BCG. Rv3615c contained multiple epitopes Rv3615c1-15, Rv3615c6-20, Rv3615c66-80, Rv3615c71-85 and Rv3615c76-90 that activated CD4+ T cells. The Rv3615c-specific CD4+ T cells shared biased of T-cell receptor variable region of β chain (TCR Vβ) 1, 2, 4, 5.1, 7.1, 7.2 and/or 22 chains to promote their differentiation and proliferation respectively, by triggering a signaling cascade. Our data suggest that Rv3615c is a major target of Th1-type responses and can be a highly immunodominant antigen specific for M. tuberculosis infection.
Collapse
|
47
|
Clemmensen HS, Knudsen NPH, Rasmussen EM, Winkler J, Rosenkrands I, Ahmad A, Lillebaek T, Sherman DR, Andersen PL, Aagaard C. An attenuated Mycobacterium tuberculosis clinical strain with a defect in ESX-1 secretion induces minimal host immune responses and pathology. Sci Rep 2017; 7:46666. [PMID: 28436493 PMCID: PMC5402389 DOI: 10.1038/srep46666] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/22/2017] [Indexed: 12/24/2022] Open
Abstract
Although Mycobacterium tuberculosis (M.tb) DK9897 is an attenuated strain, it was isolated from a patient with extrapulmonary tuberculosis and vaccination with a subunit vaccine (H56) induced poor protection against it. Both attenuation and lack of protection are because M.tb DK9897 cannot secrete the EsxA virulence factor nor induce a host response against it. Genome sequencing identified a frameshift mutation in the eccCa1 gene. Since the encoded EccCa1 protein provides energy for ESX-1 secretion, it suggested a defect in the ESX-1 type VII secretion system. Genetic complementation with a plasmid carrying the M.tb H37Rv sequence of eccCa1-eccCb1-pe35 re-established EsxA secretion, host specific EsxA T-cell responses, and increased strain virulence. The ESX-1 secretion defect prevents several virulence factors from being functional during infection and therefore attenuates M.tb. It precludes specific T-cell responses against strong antigens and we found very little in vivo cytokine production, gross pathology or granuloma formation in lungs from M.tb DK9897 infected animals. This coincides with M.tb DK9897 being unable to disrupt the phagosome membrane and make contact to the cytosol.
Collapse
Affiliation(s)
- Helena Strand Clemmensen
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Niels Peter Hell Knudsen
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Erik Michael Rasmussen
- International Reference Laboratory of Mycobacteriology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Jessica Winkler
- Center for Infectious Disease Research, Seattle, Washington, 98109, USA
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Ahmad Ahmad
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Troels Lillebaek
- International Reference Laboratory of Mycobacteriology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - David R Sherman
- Center for Infectious Disease Research, Seattle, Washington, 98109, USA
| | - Peter Lawætz Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Claus Aagaard
- Department of Infectious Disease Immunology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| |
Collapse
|
48
|
Use of whole-genome sequencing to distinguish relapse from reinfection in a completed tuberculosis clinical trial. BMC Med 2017; 15:71. [PMID: 28351427 PMCID: PMC5371199 DOI: 10.1186/s12916-017-0834-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/09/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND RIFAQUIN was a tuberculosis chemotherapy trial in southern Africa including regimens with high-dose rifapentine with moxifloxacin. Here, the application of whole-genome sequencing (WGS) is evaluated within RIFAQUIN for identifying new infections in treated patients as either relapses or reinfections. WGS is further compared with mycobacterial interspersed repetitive units-variable number tandem repeats (MIRU-VNTR) typing. This is the first report of WGS being used to evaluate new infections in a completed clinical trial for which all treatment and epidemiological data are available for analysis. METHODS DNA from 36 paired samples of Mycobacterium tuberculosis cultured from patients before and after treatment was typed using 24-loci MIRU-VNTR, in silico spoligotyping and WGS. Following WGS, the sequences were mapped against the reference strain H37Rv, the single-nucleotide polymorphism (SNP) differences between pairs were identified, and a phylogenetic reconstruction was performed. RESULTS WGS indicated that 32 of the paired samples had a very low number of SNP differences (0-5; likely relapses). One pair had an intermediate number of SNP differences, and was likely the result of a mixed infection with a pre-treatment minor genotype that was highly related to the post-treatment genotype; this was reclassified as a relapse, in contrast to the MIRU-VNTR result. The remaining three pairs had very high SNP differences (>750; likely reinfections). CONCLUSIONS WGS and MIRU-VNTR both similarly differentiated relapses and reinfections, but WGS provided significant extra information. The low proportion of reinfections seen suggests that in standard chemotherapy trials with up to 24 months of follow-up, typing the strains brings little benefit to an analysis of the trial outcome in terms of differentiating relapse and reinfection. However, there is a benefit to using WGS as compared to MIRU-VNTR in terms of the additional genotype information obtained, in particular for defining the presence of mixed infections and the potential to identify known and novel drug-resistance markers.
Collapse
|
49
|
Fedrizzi T, Meehan CJ, Grottola A, Giacobazzi E, Fregni Serpini G, Tagliazucchi S, Fabio A, Bettua C, Bertorelli R, De Sanctis V, Rumpianesi F, Pecorari M, Jousson O, Tortoli E, Segata N. Genomic characterization of Nontuberculous Mycobacteria. Sci Rep 2017; 7:45258. [PMID: 28345639 PMCID: PMC5366915 DOI: 10.1038/srep45258] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/23/2017] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis and Mycobacterium leprae have remained, for many years, the primary species of the genus Mycobacterium of clinical and microbiological interest. The other members of the genus, referred to as nontuberculous mycobacteria (NTM), have long been underinvestigated. In the last decades, however, the number of reports linking various NTM species with human diseases has steadily increased and treatment difficulties have emerged. Despite the availability of whole genome sequencing technologies, limited effort has been devoted to the genetic characterization of NTM species. As a consequence, the taxonomic and phylogenetic structure of the genus remains unsettled and genomic information is lacking to support the identification of these organisms in a clinical setting. In this work, we widen the knowledge of NTMs by reconstructing and analyzing the genomes of 41 previously uncharacterized NTM species. We provide the first comprehensive characterization of the genomic diversity of NTMs and open new venues for the clinical identification of opportunistic pathogens from this genus.
Collapse
Affiliation(s)
| | - Conor J Meehan
- Mycobacteriology unit, Department of Biomedical Science, Institute of Tropical Medicine, Antwerp, Belgium
| | - Antonella Grottola
- Microbiology and Virology Unit, University Hospital Polyclinic, Modena, Italy
| | | | | | - Sara Tagliazucchi
- Microbiology and Virology Unit, University Hospital Polyclinic, Modena, Italy
| | - Anna Fabio
- Microbiology and Virology Unit, University Hospital Polyclinic, Modena, Italy
| | - Clotilde Bettua
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Roberto Bertorelli
- NGS Facility, Laboratory of Biomolecular Sequence and Structure Analysis for Health, Centre for Integrative Biology, University of Trento, Italy
| | - Veronica De Sanctis
- NGS Facility, Laboratory of Biomolecular Sequence and Structure Analysis for Health, Centre for Integrative Biology, University of Trento, Italy
| | - Fabio Rumpianesi
- Microbiology and Virology Unit, University Hospital Polyclinic, Modena, Italy
| | - Monica Pecorari
- Microbiology and Virology Unit, University Hospital Polyclinic, Modena, Italy
| | - Olivier Jousson
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Enrico Tortoli
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
50
|
Pandey H, Tripathi S, Srivastava K, Tripathi DK, Srivastava M, Kant S, Srivastava KK, Arora A. Characterization of culture filtrate proteins Rv1197 and Rv1198 of ESAT-6 family from Mycobacterium tuberculosis H37Rv. Biochim Biophys Acta Gen Subj 2017; 1861:396-408. [DOI: 10.1016/j.bbagen.2016.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 10/01/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022]
|