1
|
Yabaji SM, Zhernovkov V, Araveti PB, Lata S, Rukhlenko OS, Abdullatif SA, Vanvalkenburg A, Alekseev Y, Ma Q, Dayama G, Lau NC, Johnson WE, Bishai WR, Crossland NA, Campbell JD, Kholodenko BN, Gimelbrant AA, Kobzik L, Kramnik I. Lipid Peroxidation and Type I Interferon Coupling Fuels Pathogenic Macrophage Activation Causing Tuberculosis Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.05.583602. [PMID: 38496444 PMCID: PMC10942339 DOI: 10.1101/2024.03.05.583602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
A quarter of human population is infected with Mycobacterium tuberculosis, but less than 10% of those infected develop pulmonary TB. We developed a genetically defined sst1-susceptible mouse model that uniquely reproduces a defining feature of human TB: the development of necrotic lung granulomas and determined that the sst1-susceptible phenotype was driven by the aberrant macrophage activation. This study demonstrates that the aberrant response of the sst1-susceptible macrophages to prolonged stimulation with TNF is primarily driven by conflicting Myc and antioxidant response pathways leading to a coordinated failure 1) to properly sequester intracellular iron and 2) to activate ferroptosis inhibitor enzymes. Consequently, iron-mediated lipid peroxidation fueled Ifn-beta superinduction and sustained the Type I Interferon (IFN-I) pathway hyperactivity that locked the sst1-susceptible macrophages in a state of unresolving stress and compromised their resistance to Mtb. The accumulation of the aberrantly activated, stressed, macrophages within granuloma microenvironment led to the local failure of anti-tuberculosis immunity and tissue necrosis. The upregulation of Myc pathway in peripheral blood cells of human TB patients was significantly associated with poor outcomes of TB treatment. Thus, Myc dysregulation in activated macrophages results in an aberrant macrophage activation and represents a novel target for host-directed TB therapies.
Collapse
|
2
|
Yabaji SM, Lata S, Tseng AE, Araveti PB, Lo M, Gavrish I, O’Connell AK, Gertje HP, Belkina AC, Thurman CE, Kiyokawa H, Kotton D, Tan S, Endsley JJ, Bishai WR, Crossland N, Kobzik L, Kramnik I. Aberrant macrophage activation and maladaptive lung repair promote tuberculosis progression uniquely in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.17.562695. [PMID: 40093068 PMCID: PMC11908135 DOI: 10.1101/2023.10.17.562695] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Pulmonary tuberculosis (PTB) represents 85% of the disease burden caused by Mycobacterium tuberculosis (Mtb) and promotes aerosol transmission infecting about a quarter of people globally. Most Mtb infections are effectively limited within primary granulomatous lesions. Containment failures lead to hematogenous spread and the formation of post-primary destructive PTB lesions. Factors that favor Mtb survival and replication in the lungs after hematogenous spread despite systemic immunity represent appealing targets for host-directed TB therapies, but are currently unknown. We developed a novel mouse model that mimics progression of chronic post-primary PTB in humans: wherein PTB lesions form after hematogenous spread from a remote primary lesion in immunocompetent but TB-susceptible B6.Sst1S mice. The B6.Sst1S mice developed PTB lesions featuring granulomatous pneumonia, bronchogenic expansion and broncho-occlusion closely resembling post-primary PTB in humans. Using spatial transcriptomic and fluorescent multiplexed immunochemistry, we demonstrated the expansion of myeloid cell populations with the appearance of alternatively activated macrophages, dissolution of initial lymphoid follicles, and accumulation of de-differentiated lung epithelial cells in the advanced PTB lesions. To determine whether lung parenchymal cells or lung oxygenation were necessary for the pulmonary TB progression, we implanted lung and spleen fragments subcutaneously to serve as potential targets for hematogenous spread. The lung (but not spleen) implants displayed characteristic organized granulomas with necrosis and Mtb replication demonstrating that deleterious interactions of aberrantly activated macrophages with the inflammation-injured lung resident cells, and possibly hypoxia, not oxygenation, are critical determinants of PTB progression in immunocompetent hosts. Necrotic TB lesions also developed in subcutaneous implants of human lung tissue in mice with human immune system after respiratory infection. These animal models may serve to further dissect the lung-specific mechanisms of host susceptibility to virulent Mtb and for testing therapeutic interventions targeting these mechanisms.
Collapse
Affiliation(s)
- Shivraj M. Yabaji
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Suruchi Lata
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna E. Tseng
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | | | - Ming Lo
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Igor Gavrish
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Aoife K O’Connell
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hans P Gertje
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna C. Belkina
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Colleen E Thurman
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hirofumi Kiyokawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Janice J. Endsley
- Departments of Microbiology & Immunology and Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William R Bishai
- Center for Tuberculosis Research School of Medicine, John Hopkins University Baltimore, Maryland
| | - Nicholas Crossland
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
| | | | - Igor Kramnik
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- Pulmonary Center, The Department of Medicine, Boston University Chobanian & Aveedisian School of Medicine
| |
Collapse
|
3
|
Kayukova L, Bismilda V, Turgenbayev K, Uzakova A, Baitursynova G, Jussipbekov U, Mukanova M, Chingissova L, Dyussembayeva G, Borsynbayeva A, Yerlanuly A, Auyezov A. β-Aminopropioamidoximes derivatives as potential antitubercular agents against anthropozoonotic infections caused by Mycobacterium tuberculosis and Mycobacterium bovis. Vet World 2025; 18:731-745. [PMID: 40342751 PMCID: PMC12056900 DOI: 10.14202/vetworld.2025.731-745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/20/2025] [Indexed: 05/11/2025] Open
Abstract
Background and Aim Tuberculosis (TB) remains a significant global health challenge, with increasing incidences of drug-sensitive (DS) and multidrug-resistant (MDR) TB. In addition, Mycobacterium bovis-induced zoonotic TB (zTB) presents treatment difficulties due to its resistance to pyrazinamide and the prolonged treatment duration required. This study aims to evaluate the antitubercular potential of β-aminopropioamidoxime derivatives against DS and MDR M. tuberculosis and M. bovis strains, and utilizing the SwissADME prognostic tool to predict the drug- and lead-likeness of the described compounds. Materials and Methods Six β-aminopropioamidoxime derivatives were synthesized through O-aroylation of amidoxime followed by dehydration to form 1,2,4-oxadiazoles. The compounds were tested in vitro against DS, MDR M. tuberculosis, and M. bovis using Sotton's liquid medium and subcultured on dense Lowenstein-Jensen medium. SwissADME was used to predict drug-likeness and pharmacokinetic properties. Results The derivatives exhibited significant antitubercular activity, with in vitro efficacy 5-100 times greater than rifampicin. 1,2,4-oxadiazoles with para-bromo and meta-chloro substituents demonstrated the highest activity against DS and MDR M. tuberculosis, while O-para-toluoyl-β-(morpholin-1-yl)propioamidoxime salts (hydrochloride, oxalate and citrate) were 10 times more active against M. bovis. SwissADME analysis confirmed favorable pharmacokinetic properties, including high gastrointestinal absorption and drug-likeness, with lead-likeness identified in four compounds. Conclusion The study presents β-aminopropioamidoxime derivatives as promising candidates for antitubercular therapy against both human and zTB. Their enhanced activity, oral bioavailability, and potential integration into new treatment regimens underscore their therapeutic relevance. Further in vivo studies are recommended to validate their efficacy and safety for clinical applications.
Collapse
Affiliation(s)
- Lyudmila Kayukova
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Venera Bismilda
- National Scientific Center of Phthisiopulmonology, Ministry of Health of the Republic of Kazakhstan, National Reference Bacteriological Laboratory, Almaty, Kazakhstan
| | | | - Assem Uzakova
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Gulnur Baitursynova
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Umirzak Jussipbekov
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Meruyert Mukanova
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Lyailya Chingissova
- National Scientific Center of Phthisiopulmonology, Ministry of Health of the Republic of Kazakhstan, National Reference Bacteriological Laboratory, Almaty, Kazakhstan
| | - Gulnur Dyussembayeva
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | | | - Azamat Yerlanuly
- JSC A.B. Bekturov Institute of Chemical Sciences, Laboratory of Chemistry of Synthetic and Natural Drug Substances, Almaty, Kazakhstan
| | - Ablay Auyezov
- National Scientific Center of Phthisiopulmonology, Ministry of Health of the Republic of Kazakhstan, National Reference Bacteriological Laboratory, Almaty, Kazakhstan
| |
Collapse
|
4
|
Davies-Bolorunduro OF, Jaemsai B, Ruangchai W, Noppanamas T, Boonbangyang M, Bodharamik T, Sawaengdee W, Mahasirimongkol S, Palittapongarnpim P. Analysis of complete genomes of Mycobacterium tuberculosis sublineage 2.1 (Proto-Beijing) revealed the presence of three pe_pgrs3-pe_pgrs4-like genes. Sci Rep 2024; 14:30702. [PMID: 39730410 DOI: 10.1038/s41598-024-79351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/08/2024] [Indexed: 12/29/2024] Open
Abstract
Mycobacterium tuberculosis Complex (MTBC), the etiological agent of tuberculosis (TB), demonstrates considerable genotypic diversity with distinct geographic distributions and variable virulence profiles. The pe-ppe gene family is especially noteworthy for its extensive variability and roles in host immune response modulation and virulence enhancement. We sequenced an Mtb genotype L2.1 isolate from Chiangrai, Northern Thailand, using second and third-generation sequencing technologies. Comparative genomic analysis with two additional L2.1 isolates and two L2.2.AA3 (Asia Ancestral 3 Beijing) isolates revealed significant pe-ppe gene variations. Notably, all L2.1 isolates harbored three copies of pe_pgrs3-pe_pgrs4-like genes (pe_pgrs3*, pe_pgrs4*, and pe_pgrs4), different from L2.2.AA3 and H37Rv strains. Additionally, ppe53 was duplicated in all but H37Rv, and ppe50 was deleted in L2.1 isolates, contrasting with an extended ppe50 in an L2.2 isolate (Mtb 18b), which contains an additional SVP motif. Complete deletion of ppe66 and loss of wag22 were observed in L2.1 isolates. These findings highlight the high structural variability of the pe-ppe gene family, emphasizing its complex roles in Mtb-host immune interactions. This genetic complexity offers potentially critical insights into mycobacterial pathogenesis, with significant implications for vaccine development and diagnostics.
Collapse
Affiliation(s)
- Olabisi Flora Davies-Bolorunduro
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
- Department of Microbiology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
- Floret Center for Advanced Genomics and Bioinformatics Research, Lagos, Nigeria
| | - Bharkbhoom Jaemsai
- Department of Microbiology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Wuthiwat Ruangchai
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Thanakron Noppanamas
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Manon Boonbangyang
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Thavin Bodharamik
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Waritta Sawaengdee
- Department of Medical Sciences, Medical Life Science Institute, Ministry of Public Health, Nonthaburi, Thailand
| | - Surakameth Mahasirimongkol
- Department of Medical Sciences, Medical Life Science Institute, Ministry of Public Health, Nonthaburi, Thailand
| | - Prasit Palittapongarnpim
- Pornchai Matangkasombut Center for Microbial Genomics, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand.
- Department of Microbiology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand.
| |
Collapse
|
5
|
Yan L, Lai HY, Leung TCN, Cheng HF, Chen X, Tsui SKW, Ngai SM, Au SWN. PE/PPE Proteome and ESX-5 Substrate Spectrum in Mycobacterium marinum. Int J Mol Sci 2024; 25:9550. [PMID: 39273496 PMCID: PMC11395111 DOI: 10.3390/ijms25179550] [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: 07/13/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024] Open
Abstract
PE/PPE proteins secreted by the ESX-5 type VII secretion system constitute a major protein repertoire in pathogenic mycobacteria and are essential for bacterial survival, pathogenicity, and host-pathogen interaction; however, little is known about their expression and secretion. The scarcity of arginine and lysine residues in PE/PPE protein sequences and the high homology of their N-terminal domains limit protein identification using classical trypsin-based proteomic methods. This study used endoproteinase AspN and trypsin to characterize the proteome of Mycobacterium marinum. Twenty-seven PE/PPE proteins were uniquely identified in AspN digests, especially PE_PGRS proteins. These treatments allowed the identification of approximately 50% of the PE/PPE pool encoded in the genome. Moreover, EspG5 pulldown assays retrieved 44 ESX-5-associated PPE proteins, covering 85% of the PPE pool in the identified proteome. The identification of PE/PE_PGRS proteins in the EspG5 interactome suggested the presence of PE-PPE pairs. The correlation analysis between protein abundance and phylogenetic relationships found potential PE/PPE pairs, indicating the presence of multiple PE/PE_PGRS partners in one PPE. We validated that EspG5 interacted with PPE31 and PPE32 and mapped critical residues for complex formation. The modified proteomic platform increases the coverage of PE/PPE proteins and elucidates the expression and localization of these proteins.
Collapse
Affiliation(s)
- Lili Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Hiu Ying Lai
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Thomas Chun Ning Leung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Hiu Fu Cheng
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Xin Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Stephen Kwok Wing Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Sai Ming Ngai
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Shannon Wing Ngor Au
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| |
Collapse
|
6
|
Zheng W, Diao NC, Wang Q, Wang CY, Su N, Yin JY, Tian T, Shi K, Du R. Worldwide Swine Tuberculosis-Positive Rate and Associated Risk Factors, 1966-2020: A Systematic Review and Meta-Analysis. Vector Borne Zoonotic Dis 2024; 24:181-195. [PMID: 38306180 DOI: 10.1089/vbz.2023.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024] Open
Abstract
Background: Tuberculosis (TB) is a chronic, zoonotic infectious disease caused by Mycobacterium tuberculosis that infects not only humans but also animals such as pigs, cows, buffaloes, sheep, and goats. Among them, pigs are one of the main food animals in the world. If pigs are infected with M. tuberculosis, meat products will be negatively affected, causing economic losses to the livestock industry. There is currently no systematic epidemiological assessment of swine TB in the world, so it is important to know the prevalence of swine, and these data are currently lacking, so we performed a statistical analysis. Results: We searched 6791 articles and finally included data from 35,303 pigs from 15 countries or territories, showing a combined prevalence of TB in pigs of 12.1% (95% confidence interval [CI]: 9.2 to 15.9). Among them, the prevalence rate of swine TB in Europe was 15.2% (95% CI: 11.1 to 20.7, 2491/25,050), which was higher compared with other continents, and the difference was significant; the positive rate of PCR method was higher in the detection method subgroup, which was 15.7% (95% CI: 8.0 to 31.0, 376/2261); Mycobacterium bovis was detected in pigs in the M. tuberculosis typing group (9.5%, 95% CI: 6.7 to 13.5, 1364/21,430). The positive rate is higher compared with Mycobacterium capris. Conclusion: This systematic review and meta-analysis is the first to determine the global prevalence of TB in swine herds. Although the seroprevalence of swine TB in this article is very low, the harm of TB cannot be ignored. It is important to take effective control and preventive measures to stop the spread of TB to reduce the impact of diseased pigs on animal husbandry and human health.
Collapse
Affiliation(s)
- Wei Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, P.R. China
| | - Nai-Chao Diao
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, P.R. China
| | - Qi Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, P.R. China
| | - Chun-Yu Wang
- Jilin Heyuan Bioengineering Co., Ltd., Songyuan, P.R. China
| | - Nuo Su
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, P.R. China
| | - Ji-Ying Yin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, P.R. China
| | - Tian Tian
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, P.R. China
| | - Kun Shi
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, P.R. China
| | - Rui Du
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, P.R. China
- Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, P.R. China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, P.R. China
| |
Collapse
|
7
|
Orgeur M, Sous C, Madacki J, Brosch R. Evolution and emergence of Mycobacterium tuberculosis. FEMS Microbiol Rev 2024; 48:fuae006. [PMID: 38365982 PMCID: PMC10906988 DOI: 10.1093/femsre/fuae006] [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: 11/09/2023] [Revised: 01/12/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024] Open
Abstract
Tuberculosis (TB) remains one of the deadliest infectious diseases in human history, prevailing even in the 21st century. The causative agents of TB are represented by a group of closely related bacteria belonging to the Mycobacterium tuberculosis complex (MTBC), which can be subdivided into several lineages of human- and animal-adapted strains, thought to have shared a last common ancestor emerged by clonal expansion from a pool of recombinogenic Mycobacterium canettii-like tubercle bacilli. A better understanding of how MTBC populations evolved from less virulent mycobacteria may allow for discovering improved TB control strategies and future epidemiologic trends. In this review, we highlight new insights into the evolution of mycobacteria at the genus level, describing different milestones in the evolution of mycobacteria, with a focus on the genomic events that have likely enabled the emergence and the dominance of the MTBC. We also review the recent literature describing the various MTBC lineages and highlight their particularities and differences with a focus on host preferences and geographic distribution. Finally, we discuss on putative mechanisms driving the evolution of tubercle bacilli and mycobacteria in general, by taking the mycobacteria-specific distributive conjugal transfer as an example.
Collapse
Affiliation(s)
- Mickael Orgeur
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Camille Sous
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Jan Madacki
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Unit for Human Evolutionary Genetics, 75015 Paris, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| |
Collapse
|
8
|
Born SEM, Reichlen MJ, Bartek IL, Benoit JB, Frank DN, Voskuil MI. Population heterogeneity in Mycobacterium smegmatis and Mycobacterium abscessus. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001402. [PMID: 37862100 PMCID: PMC10634367 DOI: 10.1099/mic.0.001402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
Bacteria use population heterogeneity, the presence of more than one phenotypic variant in a clonal population, to endure diverse environmental challenges - a 'bet-hedging' strategy. Phenotypic variants have been described in many bacteria, but the phenomenon is not well-understood in mycobacteria, including the environmental factors that influence heterogeneity. Here, we describe three reproducible morphological variants in M. smegmatis - smooth, rough, and an intermediate morphotype that predominated under typical laboratory conditions. M. abscessus has two recognized morphotypes, smooth and rough. Interestingly, M. tuberculosis exists in only a rough form. The shift from smooth to rough in both M. smegmatis and M. abscessus was observed over time in extended static culture, however the frequency of the rough morphotype was high in pellicle preparations compared to planktonic culture, suggesting a role for an aggregated microenvironment in the shift to the rough form. Differences in growth rate, biofilm formation, cell wall composition, and drug tolerance were noted among M. smegmatis and M. abscessus variants. Deletion of the global regulator lsr2 shifted the M. smegmatis intermediate morphotype to a smooth form but did not fully phenocopy the naturally generated smooth morphotype, indicating Lsr2 is likely downstream of the initiating regulatory cascade that controls these morphotypes. Rough forms typically correlate with higher invasiveness and worse outcomes during infection and our findings indicate the shift to this rough form is promoted by aggregation. Our findings suggest that mycobacterial population heterogeneity, reflected in colony morphotypes, is a reproducible, programmed phenomenon that plays a role in adaptation to unique environments and this heterogeneity may influence infection progression and response to treatment.
Collapse
Affiliation(s)
- Sarah E. M. Born
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Matthew J. Reichlen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Iona L. Bartek
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeanne B. Benoit
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Martin I. Voskuil
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
9
|
Barrera-Rosales A, Rodríguez-Sanoja R, Hernández-Pando R, Moreno-Mendieta S. The Use of Particulate Systems for Tuberculosis Prophylaxis and Treatment: Opportunities and Challenges. Microorganisms 2023; 11:1988. [PMID: 37630548 PMCID: PMC10459556 DOI: 10.3390/microorganisms11081988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
The use of particles to develop vaccines and treatments for a wide variety of diseases has increased, and their success has been demonstrated in preclinical investigations. Accurately targeting cells and minimizing doses and adverse side effects, while inducing an adequate biological response, are important advantages that particulate systems offer. The most used particulate systems are liposomes and their derivatives, immunostimulatory complexes, virus-like particles, and organic or inorganic nano- and microparticles. Most of these systems have been proven using therapeutic or prophylactic approaches to control tuberculosis, one of the most important infectious diseases worldwide. This article reviews the progress and current state of the use of particles for the administration of TB vaccines and treatments in vitro and in vivo, with a special emphasis on polymeric particles. In addition, we discuss the challenges and benefits of using these particulate systems to provide researchers with an overview of the most promising strategies in current preclinical trials, offering a perspective on their progress to clinical trials.
Collapse
Affiliation(s)
- Alejandra Barrera-Rosales
- Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, México;
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, México; (R.R.-S.)
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México 14080, México
| | - Silvia Moreno-Mendieta
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, México; (R.R.-S.)
- CONAHCyT, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, México
| |
Collapse
|
10
|
Karakousis ND, Gourgoulianis KI, Kotsiou OS. Sarcopenia and Tuberculosis: Is There Any Connection? J Pers Med 2023; 13:1102. [PMID: 37511715 PMCID: PMC10381550 DOI: 10.3390/jpm13071102] [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: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Tuberculosis (TB) infection is a life-threatening infection caused by certain bacteria belonging to the Mycobacterium tuberculosis complex. More than 10 million subjects are newly sick from this infection every year globally. At the same time, TB is quite prevalent among subjects who come from lower socioeconomic layers of general population, and marginalized sections and areas. Sarcopenia is a muscle disease that derives from adverse muscle alterations and is related to the loss of muscle strength and mass. It is a major medical issue due to its increased adverse outcomes including falls, functional decline, frailty, hospitalizations, increased mortality, and healthcare costs. METHODS This study examined the potential interplay between the TB infection and sarcopenia through conducting a non-systematic review of the current literature. RESULTS It has been recorded that the prevalence of sarcopenia among TB survivors is high, whilst the danger of TB among the elderly increases with sarcopenia and physical inactivity. Nevertheless, sufficient protein and total energy intake are associated with a low risk of sarcopenia in TB survivors. CONCLUSIONS Further studies are needed to validate these findings and shed more light on the upcoming different aspects of this intriguing association.
Collapse
Affiliation(s)
- Nikolaos D Karakousis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Ourania S Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41110 Larissa, Greece
- Laboratory of Human Pathophysiology, Faculty of Nursing, University of Thessaly, Gaiopolis, 41500 Larissa, Greece
| |
Collapse
|
11
|
Gonzalo X, Yrah S, Broda A, Laurenson I, Claxton P, Kostrzewa M, Drobniewski F, Larrouy-Maumus G. Performance of lipid fingerprint by routine matrix-assisted laser desorption/ionization time of flight for the diagnosis of Mycobacterium tuberculosis complex species. Clin Microbiol Infect 2023; 29:387.e1-387.e6. [PMID: 36270589 DOI: 10.1016/j.cmi.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/21/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Rapid detection of bacterial pathogens at species and sub-species levels is crucial for appropriate treatment, infection control, and public health management. Currently, one of the challenges in clinical microbiology is the discrimination of mycobacterial sub-species within the M. tuberculosis complex (MTBC). Our objective was to evaluate the ability of a biosafe mycobacterial lipid-based approach to identify MTBC cultures and sub-species. METHODS A blinded study was conducted using 90 mycobacterial clinical isolate strains comprising MTBC strains sub-cultured in Middlebrook 7H11 medium supplemented with 10% oleic-acid, dextrose, catalase growth supplement and incubated for up to 6 weeks at 37°C and using the following seven reference strains (M. tuberculosis H37Rv, M canettii, M. africanum, M. pinnipedii, M. caprae, M. bovis, and M. bovis BCG) grown under the same conditions, to set the reference lipid database and test it against the 90 MTBC clinical isolates. Cultured mycobacteria were heat-inactivated and loaded onto the matrix-assisted laser desorption/ionization target followed by the addition of the matrix. Acquisition of the data was performed using the positive ion mode. RESULTS Based on the identification of clear and defined lipid signatures from the seven reference strains, the method that we developed was fast (<10 minutes) and produced interpretable profiles for all but four isolates, caused by poor ionization giving an n = 86 with interpretable spectra. The sensitivity and specificity of the matrix-assisted laser desorption/ionization time of flight were 94.4 (95% CI, 86.4-98.5) and 94.4 (95% CI, 72.7-99.9), respectively. CONCLUSIONS Mycobacterial lipid profiling provides a means of rapid, safe, and accurate discrimination of species within the MTBC.
Collapse
Affiliation(s)
- Ximena Gonzalo
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Shih Yrah
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Agnieszka Broda
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ian Laurenson
- Scottish Mycobacteria Reference Laboratory, Edinburgh, United Kingdom
| | - Pauline Claxton
- Scottish Mycobacteria Reference Laboratory, Edinburgh, United Kingdom
| | | | - Francis Drobniewski
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
12
|
Saelens JW, Sweeney MI, Viswanathan G, Xet-Mull AM, Jurcic Smith KL, Sisk DM, Hu DD, Cronin RM, Hughes EJ, Brewer WJ, Coers J, Champion MM, Champion PA, Lowe CB, Smith CM, Lee S, Stout JE, Tobin DM. An ancestral mycobacterial effector promotes dissemination of infection. Cell 2022; 185:4507-4525.e18. [PMID: 36356582 PMCID: PMC9691622 DOI: 10.1016/j.cell.2022.10.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/27/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.
Collapse
Affiliation(s)
- Joseph W Saelens
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mollie I Sweeney
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ana María Xet-Mull
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kristen L Jurcic Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dana M Sisk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel D Hu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel M Cronin
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sunhee Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jason E Stout
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Division of Infectious Diseases and International Health, Duke University School of Medicine, Durham, NC 27710, USA.
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
13
|
Weng Y, Shepherd D, Liu Y, Krishnan N, Robertson BD, Platt N, Larrouy-Maumus G, Platt FM. Inhibition of the Niemann-Pick C1 protein is a conserved feature of multiple strains of pathogenic mycobacteria. Nat Commun 2022; 13:5320. [PMID: 36085278 PMCID: PMC9463166 DOI: 10.1038/s41467-022-32553-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 08/04/2022] [Indexed: 11/12/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) survives and replicates within host macrophages (MΦ) and subverts multiple antimicrobial defense mechanisms. Previously, we reported that lipids shed by pathogenic mycobacteria inhibit NPC1, the lysosomal membrane protein deficient in the lysosomal storage disorder Niemann-Pick disease type C (NPC). Inhibition of NPC1 leads to a drop in lysosomal calcium levels, blocking phagosome-lysosome fusion leading to mycobacterial survival. We speculated that the production of specific cell wall lipid(s) that inhibit NPC1 could have been a critical step in the evolution of pathogenicity. We therefore investigated whether lipid extracts from clinical Mtb strains from multiple Mtb lineages, Mtb complex (MTBC) members and non-tubercular mycobacteria (NTM) inhibit the NPC pathway. We report that inhibition of the NPC pathway was present in all clinical isolates from Mtb lineages 1, 2, 3 and 4, Mycobacterium bovis and the NTM, Mycobacterium abscessus and Mycobacterium avium. However, lipid extract from Mycobacterium canettii, which is considered to resemble the common ancestor of the MTBC did not inhibit the NPC1 pathway. We conclude that the evolution of NPC1 inhibitory mycobacterial cell wall lipids evolved early and post divergence from Mycobacterium canettii-related mycobacteria and that this activity contributes significantly to the promotion of disease.
Collapse
Affiliation(s)
- Yuzhe Weng
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Dawn Shepherd
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Yi Liu
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Nitya Krishnan
- MRC Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, Flowers Building, London, SW7 2AZ, UK
| | - Brian D Robertson
- MRC Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, Flowers Building, London, SW7 2AZ, UK
| | - Nick Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
14
|
Kirsch SH, Haeckl FPJ, Müller R. Beyond the approved: target sites and inhibitors of bacterial RNA polymerase from bacteria and fungi. Nat Prod Rep 2022; 39:1226-1263. [PMID: 35507039 DOI: 10.1039/d1np00067e] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Covering: 2016 to 2022RNA polymerase (RNAP) is the central enzyme in bacterial gene expression representing an attractive and validated target for antibiotics. Two well-known and clinically approved classes of natural product RNAP inhibitors are the rifamycins and the fidaxomycins. Rifampicin (Rif), a semi-synthetic derivative of rifamycin, plays a crucial role as a first line antibiotic in the treatment of tuberculosis and a broad range of bacterial infections. However, more and more pathogens such as Mycobacterium tuberculosis develop resistance, not only against Rif and other RNAP inhibitors. To overcome this problem, novel RNAP inhibitors exhibiting different target sites are urgently needed. This review includes recent developments published between 2016 and today. Particular focus is placed on novel findings concerning already known bacterial RNAP inhibitors, the characterization and development of new compounds isolated from bacteria and fungi, and providing brief insights into promising new synthetic compounds.
Collapse
Affiliation(s)
- Susanne H Kirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany. .,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - F P Jake Haeckl
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany. .,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany. .,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany.,Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
15
|
Zhang J, Hu L, Zhang H, He Z. Cyclic
di‐GMP
triggers the hypoxic adaptation of
Mycobacterium bovis
through a metabolic switching regulator
ArgR. Environ Microbiol 2022; 24:4382-4400. [DOI: 10.1111/1462-2920.15987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Jiaxun Zhang
- College of Life Science and Technology Huazhong Agricultural University Wuhan 430070 China
| | - Lihua Hu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐bioresources, College of Life Science and Technology Guangxi University Nanning 530004 China
| | - Hua Zhang
- College of Life Science and Technology Huazhong Agricultural University Wuhan 430070 China
| | - Zheng‐Guo He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐bioresources, College of Life Science and Technology Guangxi University Nanning 530004 China
| |
Collapse
|
16
|
Tang P, Liang E, Zhang X, Feng Y, Song H, Xu J, Wu M, Pang Y. Prevalence and Risk Factors of Subclinical Tuberculosis in a Low-Incidence Setting in China. Front Microbiol 2022; 12:731532. [PMID: 35087480 PMCID: PMC8787132 DOI: 10.3389/fmicb.2021.731532] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: Subclinical tuberculosis (TB) represents a substantial proportion of individuals with TB disease, although limited evidence is available to understand the epidemiological characteristics of these cases. We aimed to explore the prevalence of subclinical patients with TB and identify the underlying association between the subclinical TB cases in the study setting and the Beijing genotype. Methods: A retrospective study was conducted among patients with incident TB at the Fifth People’s Hospital of Suzhou between January and December 2018. A total of 380 patients with TB were included in our analysis. Results: Of the 380 patients, 81.8% were active TB cases, whereas the other 18.2% were subclinical TB cases. Compared with patients aged 65 years and older, the risk of having subclinical TB is higher among younger patients. The use of smear, culture, and Xpert identified 3, 16, and 13 subclinical TB cases, respectively. When using a combination of positive culture and Xpert results, the sensitivity improved to 33.3%. In addition, the neutrophil-to-lymphocyte ratio was significantly elevated in the active TB group compared with that in the subclinical TB group. We also observed that the proportion of the Beijing genotype in the subclinical TB group was significantly lower than that in the active TB group. Conclusion: To conclude, our data demonstrate that approximately one-fifth of patients with TB were subclinical in Suzhou. Mycobacterium tuberculosis could be detected by the existing microbiologic diagnostics in one-third of patients with subclinical TB. The patients with subclinical TB are more prone to having low neutrophil-to-lymphocyte ratio values than those with active TB. Additionally, non-Beijing genotype strains are associated with subclinical TB.
Collapse
Affiliation(s)
- Peijun Tang
- Department of Tuberculosis, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Ermin Liang
- Department of Tuberculosis, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Xuxia Zhang
- Department of Bacteriology and Immunology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yanjun Feng
- Department of Tuberculosis, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Huafeng Song
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Junchi Xu
- Department of Clinical Laboratory, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Meiying Wu
- Department of Tuberculosis, The Fifth People's Hospital of Suzhou, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, China
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Wang Y, Tang Y, Lin C, Zhang J, Mai J, Jiang J, Gao X, Li Y, Zhao G, Zhang L, Liu J. Crosstalk between the ancestral type VII secretion system ESX-4 and other T7SS in Mycobacterium marinum. iScience 2022; 25:103585. [PMID: 35005535 PMCID: PMC8718981 DOI: 10.1016/j.isci.2021.103585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/30/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
The type VII secretion system (T7SS) of Mycobacterium tuberculosis secretes three substrate classes: Esx, Esp, and PE/PPE proteins, that play important roles in bacterial physiology and host interaction. Five subtypes of T7SS, namely ESX-1 to ESX-5, are present in M. tb. ESX-4 is the progenitor of T7SS but its function is not understood. We investigated the ESX-4 system in Mycobacterium marinum. We show that ESX-4 of M. marinum does not secrete its cognate substrates, EsxT and EsxU, under the conditions tested. Paradoxically, the deletion of eccC4, an essential component of ESX-4, resulted in elevated secretion of protein substrates of ESX-1 and ESX-5. Consequently, the ΔeccC4 mutant was more efficient in inducing actin cytoskeleton rearrangement, which led to enhanced phagocytosis by macrophages. Our results reveal an intimate crosstalk between the progenitor of T7SS and its more recent duplication and expansion, and provide new insight into the evolution of T7SS in mycobacteria.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Microbiology, School of Life Science, Fudan University, Shanghai 200090, China.,Guizhou institute of Biotechnology, Guiyang 550025, Guizhou, China
| | - Yuting Tang
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Chen Lin
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Junli Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Juntao Mai
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G1M1, Canada
| | - Jun Jiang
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Xiaoxiao Gao
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Yao Li
- State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China
| | - Guoping Zhao
- Department of Microbiology, School of Life Science, Fudan University, Shanghai 200090, China
| | - Lu Zhang
- Department of Microbiology, School of Life Science, Fudan University, Shanghai 200090, China.,State Key Laboratory of Genetic Engineering, School of Life Science, Institute of Genetics, Fudan University, Shanghai 200090, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai 200090, China
| | - Jun Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G1M1, Canada
| |
Collapse
|
18
|
Yoon Y, Seo H, Kim S, Lee Y, Rahim MDA, Lee S, Song HY. Anti-Tuberculosis Activity of Pediococcus acidilactici Isolated from Young Radish Kimchi against Mycobacterium tuberculosis. J Microbiol Biotechnol 2021; 31:1632-1642. [PMID: 34584040 PMCID: PMC9705845 DOI: 10.4014/jmb.2107.07044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022]
Abstract
Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis. It affects about 10 million people each year and is still one of the leading causes of death worldwide. About 2 to 3 billion people (equivalent to 1 in 3 people in the world) are infected with latent tuberculosis. Moreover, as the number of multidrug-resistant, extensively drug-resistant, and totally drug-resistant strains of M. tuberculosis continues to increase, there is an urgent need to develop new anti-tuberculosis drugs that are different from existing drugs to combat antibiotic-resistant M. tuberculosis. Against this background, we aimed to develop new anti-tuberculosis drugs using probiotics. Here, we report the anti-tuberculosis effect of Pediococcus acidilactici PMC202 isolated from young radish kimchi, a traditional Korean fermented food. Under coculture conditions, PMC202 inhibited the growth of M. tuberculosis. In addition, PMC202 inhibited the growth of drug-sensitive and -resistant M. tuberculosis- infected macrophages at a concentration that did not show cytotoxicity and showed a synergistic effect with isoniazid. In a 2-week, repeated oral administration toxicity study using mice, PMC202 did not cause weight change or specific clinical symptoms. Furthermore, the results of 16S rRNA-based metagenomics analysis confirmed that dysbiosis was not induced in bronchoalveolar lavage fluid after oral administration of PMC202. The anti-tuberculosis effect of PMC202 was found to be related to the reduction of nitric oxide. Our findings indicate that PMC202 could be used as an anti-tuberculosis drug candidate with the potential to replace current chemicalbased drugs. However, more extensive toxicity, mechanism of action, and animal efficacy studies with clinical trials are needed.
Collapse
Affiliation(s)
- Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - MD Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Saebim Lee
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea,Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan 31538, Republic of Korea,Corresponding author Phone: +82-41-570-2412 Fax : +82-41-577-2415 E-mail:
| |
Collapse
|
19
|
Mahghani GA, Kargar M, Ghaemi EA, Kafilzadeh F, Davoodi H. Role of ESAT-6 in pathogenicity of Beijing and non-Beijing Mycobacterium tuberculosis isolates. Microb Pathog 2021; 162:105366. [PMID: 34968645 DOI: 10.1016/j.micpath.2021.105366] [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: 08/26/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis Beijing genotype was associated with tuberculosis outbreaks and increased transmissibility. To understand the variation in virulence between Beijing and non-Beijing clinical isolates of M.tuberculosis genotypes, the esat-6 gene sequencing, and its expression was compared in the macrophage environment. MATERIALS & METHODS Among 64 nonrepetitive, culture-positive M.tuberculosis, DNA extraction of 24 and 40 pure confirmed Beijing and non-Beijing isolates was accompanied by the boiling method. esat-6 gene PCR amplification and their sequencing were carried out by specific primers and its expression was performed on human macrophage cell line U937 after 6, 12, and 18 h of exposure to bacilli. The esat-6 mRNA transcription and expression in M. tuberculosis treated macrophage by Real-Time PCR and Western blot method. RESULTS Data analysis based on sequencing of the east-6 gene PCR product showed that this gene exists in all isolates and there are no changes or single nucleotide variation between the Beijing and non-Beijing isolates. In Beijing strains, the esat-6 expression was increased during the study times, but it was constant in non-Beijing isolates. esat-6 gene expression in Beijing isolates reached to about 44.9 times more than non-Beijing isolates after 18 h incubation on the macrophages cell line. CONCLUSION esat-6 is a conserved gene both in Beijing and non-Beijing isolates of M.tuberculosis. More expression of the east-6 gene in the macrophage model may indicate that this gene is likely to play a more important role in increasing the pathogenicity of Beijing strains.
Collapse
Affiliation(s)
- Ghorban Ali Mahghani
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Mohammad Kargar
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran.
| | - Ezzat Allah Ghaemi
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farshid Kafilzadeh
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Homa Davoodi
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
20
|
Iszatt JJ, Larcombe AN, Chan HK, Stick SM, Garratt LW, Kicic A. Phage Therapy for Multi-Drug Resistant Respiratory Tract Infections. Viruses 2021; 13:v13091809. [PMID: 34578390 PMCID: PMC8472870 DOI: 10.3390/v13091809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
The emergence of multi-drug resistant (MDR) bacteria is recognised today as one of the greatest challenges to public health. As traditional antimicrobials are becoming ineffective and research into new antibiotics is diminishing, a number of alternative treatments for MDR bacteria have been receiving greater attention. Bacteriophage therapies are being revisited and present a promising opportunity to reduce the burden of bacterial infection in this post-antibiotic era. This review focuses on the current evidence supporting bacteriophage therapy against prevalent or emerging multi-drug resistant bacterial pathogens in respiratory medicine and the challenges ahead in preclinical data generation. Starting with efforts to improve delivery of bacteriophages to the lung surface, the current developments in animal models for relevant efficacy data on respiratory infections are discussed before finishing with a summary of findings from the select human trials performed to date.
Collapse
Affiliation(s)
- Joshua J. Iszatt
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Alexander N. Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, Sydney Pharmacy School, University of Sydney, Camperdown 2006, Australia;
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Nedlands 6009, Australia
| | - Luke W. Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
21
|
Boom WH, Schaible UE, Achkar JM. The knowns and unknowns of latent Mycobacterium tuberculosis infection. J Clin Invest 2021; 131:136222. [PMID: 33529162 DOI: 10.1172/jci136222] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Humans have been infected with Mycobacterium tuberculosis (Mtb) for thousands of years. While tuberculosis (TB), one of the deadliest infectious diseases, is caused by uncontrolled Mtb infection, over 90% of presumed infected individuals remain asymptomatic and contain Mtb in a latent TB infection (LTBI) without ever developing disease, and some may clear the infection. A small number of heavily Mtb-exposed individuals appear to resist developing traditional LTBI. Because Mtb has mechanisms for intracellular survival and immune evasion, successful control involves all of the arms of the immune system. Here, we focus on immune responses to Mtb in humans and nonhuman primates and discuss new concepts and outline major knowledge gaps in our understanding of LTBI, ranging from the earliest events of exposure and infection to success or failure of Mtb control.
Collapse
Affiliation(s)
- W Henry Boom
- Department of Medicine.,Department of Pathology, and.,Department of Molecular Biology and Microbiology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ulrich E Schaible
- Division of Cellular Microbiology, Research Center Borstel-Leibniz Lung Center, Borstel, Germany.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Jacqueline M Achkar
- Department of Medicine and.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
22
|
Natural products from Brazilian biodiversity identified as potential inhibitors of PknA and PknB of M. tuberculosis using molecular modeling tools. Comput Biol Med 2021; 136:104694. [PMID: 34365277 DOI: 10.1016/j.compbiomed.2021.104694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 11/21/2022]
Abstract
Mycobacterium tuberculosis was discovered in 1882 by Robert Koch but, since its discovery, the tuberculosis (TB) epidemic has endured, being one of the top 10 causes of death worldwide. Drug-resistant TB continues to be a public health threat and bioactive compounds with a new mode of action (MoA) are needed to overcome this. Since natural products are described as important sources for the development of new drugs, the objective of this work was to identify potential ligands from Brazilian natural products (NPs) for M. tuberculosis targets using molecular modeling tools. Using chemogenomics we identified the Serine/Threonine Protein Kinase PknB as a putative target for 13 NPs from a database from Brazilian biodiversity (NuBBE). Literature data supported further investigation of NuBBE105, NuBBE598, NuBBE936, NuBBE964, NuBBE1045, and NuBBE1180 by molecular docking and dynamics. Key interactions were observed with PknB and simulations confirmed stability and favorable binding energies. Considering structural similarity with PknB, we further explored binding of the NPs to PknA, critical for M. tuberculosis survival, and all of them resembled important interactions with the enzyme, showing stable and favorable binding energies, whilst van der Waals interactions seem to play a key role for binding to PknA and PknB. NuBBE936 and NuBBE1180 have already had their antimycobacterial activity reported and our results can provide a basis for their MoA. Finally, the other NPs which have not been tested against M. tuberculosis deserve further investigation, aiming at the discovery of antimycobacterial drug candidates with innovative MoA.
Collapse
|
23
|
Beckham KSH, Ritter C, Chojnowski G, Ziemianowicz DS, Mullapudi E, Rettel M, Savitski MM, Mortensen SA, Kosinski J, Wilmanns M. Structure of the mycobacterial ESX-5 type VII secretion system pore complex. SCIENCE ADVANCES 2021; 7:eabg9923. [PMID: 34172453 PMCID: PMC8232910 DOI: 10.1126/sciadv.abg9923] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/10/2021] [Indexed: 05/28/2023]
Abstract
The ESX-5 type VII secretion system is a membrane-spanning protein complex key to the virulence of mycobacterial pathogens. However, the overall architecture of the fully assembled translocation machinery and the composition of the central secretion pore have remained unknown. Here, we present the high-resolution structure of the 2.1-megadalton ESX-5 core complex. Our structure captured a dynamic, secretion-competent conformation of the pore within a well-defined transmembrane section, sandwiched between two flexible protein layers at the cytosolic entrance and the periplasmic exit. We propose that this flexibility endows the ESX-5 machinery with large conformational plasticity required to accommodate targeted protein secretion. Compared to known secretion systems, a highly dynamic state of the pore may represent a fundamental principle of bacterial secretion machineries.
Collapse
Affiliation(s)
- Katherine 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
| | - Grzegorz Chojnowski
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany
| | - Daniel S Ziemianowicz
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany
| | - Edukondalu Mullapudi
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany
| | - Mandy Rettel
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Simon A Mortensen
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany
| | - Jan Kosinski
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany.
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Notkestrasse 85, 22607 Hamburg, Germany.
- University Hamburg Clinical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
24
|
Bryant JM, Brown KP, Burbaud S, Everall I, Belardinelli JM, Rodriguez-Rincon D, Grogono DM, Peterson CM, Verma D, Evans IE, Ruis C, Weimann A, Arora D, Malhotra S, Bannerman B, Passemar C, Templeton K, MacGregor G, Jiwa K, Fisher AJ, Blundell TL, Ordway DJ, Jackson M, Parkhill J, Floto RA. Stepwise pathogenic evolution of Mycobacterium abscessus. Science 2021; 372:372/6541/eabb8699. [PMID: 33926925 DOI: 10.1126/science.abb8699] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
Although almost all mycobacterial species are saprophytic environmental organisms, a few, such as Mycobacterium tuberculosis, have evolved to cause transmissible human infection. By analyzing the recent emergence and spread of the environmental organism M. abscessus through the global cystic fibrosis population, we have defined key, generalizable steps involved in the pathogenic evolution of mycobacteria. We show that epigenetic modifiers, acquired through horizontal gene transfer, cause saltational increases in the pathogenic potential of specific environmental clones. Allopatric parallel evolution during chronic lung infection then promotes rapid increases in virulence through mutations in a discrete gene network; these mutations enhance growth within macrophages but impair fomite survival. As a consequence, we observe constrained pathogenic evolution while person-to-person transmission remains indirect, but postulate accelerated pathogenic adaptation once direct transmission is possible, as observed for M. tuberculosis Our findings indicate how key interventions, such as early treatment and cross-infection control, might restrict the spread of existing mycobacterial pathogens and prevent new, emergent ones.
Collapse
Affiliation(s)
- Josephine M Bryant
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,University of Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Karen P Brown
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - Sophie Burbaud
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Isobel Everall
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,Wellcome Sanger Institute, Hinxton, UK
| | - Juan M Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Daniela Rodriguez-Rincon
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Dorothy M Grogono
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - Chelsea M Peterson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Ieuan E Evans
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - Christopher Ruis
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,University of Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Aaron Weimann
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,University of Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Divya Arora
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Sony Malhotra
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK.,Scientific Computing Department, Science and Technology Facilities Council, Harwell, UK
| | - Bridget Bannerman
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK.,University of Cambridge Centre for AI in Medicine, Cambridge, UK
| | - Charlotte Passemar
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Kerra Templeton
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, Scotland, UK
| | - Gordon MacGregor
- Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, Glasgow, Scotland, UK
| | - Kasim Jiwa
- Newcastle University Translational and Clinical Research Institute and Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute and Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Julian Parkhill
- Wellcome Sanger Institute, Hinxton, UK. .,Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - R Andres Floto
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK. .,University of Cambridge Centre for AI in Medicine, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| |
Collapse
|
25
|
Zakham F, Sironen T, Vapalahti O, Kant R. Pan and Core Genome Analysis of 183 Mycobacterium tuberculosis Strains Revealed a High Inter-Species Diversity among the Human Adapted Strains. Antibiotics (Basel) 2021; 10:antibiotics10050500. [PMID: 33924811 PMCID: PMC8145561 DOI: 10.3390/antibiotics10050500] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/24/2021] [Accepted: 04/25/2021] [Indexed: 11/16/2022] Open
Abstract
Tuberculosis (TB) is an airborne communicable disease with high morbidity and mortality rates, especially in developing countries. The causal agents of TB belong to the complex Mycobacterium tuberculosis (MTBc), which is composed of different human and animal TB associated species. Some animal associated species have zoonotic potential and add to the burden of TB management. The BCG ("Bacillus Calmette-Guérin") vaccine is widely used for the prevention against TB, but its use is limited in immunocompromised patients and animals due to the adverse effects and disseminated life-threatening complications. In this study, we aimed to carry out a comparative genome analysis between the human adapted species including BCG vaccine strains to identify and pinpoint the conserved genes related to the virulence across all the species, which could add a new value for vaccine development. For this purpose, the sequences of 183 Mycobacterium tuberculosis (MTB) strains were retrieved from the freely available WGS dataset at NCBI. The species included: 168 sensu stricto MTB species with other human MTB complex associated strains: M. tuberculosis var. africanum (3), M. tuberculosis var. bovis (2 draft genomes) and 10 BCG species, which enabled the analysis of core genome which contains the conserved genes and some virulence factor determinants. Further, a phylogenetic tree was constructed including the genomes of human (183); animals MTB adapted strains (6) and the environmental Mycobacterium strain "M. canettii". Our results showed that the core genome consists of 1166 conserved genes among these species, which represents a small portion of the pangenome (7036 genes). The remaining genes in the pangenome (5870) are accessory genes, adding a high inter-species diversity. Further, the core genome includes several virulence-associated genes and this could explain the rare infectiousness potential of some attenuated vaccine strains in some patients. This study reveals that low number of conserved genes in human adapted MTBc species and high inter-species diversity of the pan-genome could be considered for vaccine candidate development.
Collapse
Affiliation(s)
- Fathiah Zakham
- Department of Virology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (F.Z.); (T.S.); (O.V.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Tarja Sironen
- Department of Virology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (F.Z.); (T.S.); (O.V.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (F.Z.); (T.S.); (O.V.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- HUSLAB, Hospital District of Helsinki and Uusimaa, 00260 Helsinki, Finland
| | - Ravi Kant
- Department of Virology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (F.Z.); (T.S.); (O.V.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- Correspondence:
| |
Collapse
|
26
|
Park JH, Shim D, Kim KES, Lee W, Shin SJ. Understanding Metabolic Regulation Between Host and Pathogens: New Opportunities for the Development of Improved Therapeutic Strategies Against Mycobacterium tuberculosis Infection. Front Cell Infect Microbiol 2021; 11:635335. [PMID: 33796480 PMCID: PMC8007978 DOI: 10.3389/fcimb.2021.635335] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/01/2021] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) causes chronic granulomatous lung disease in humans. Recently, novel strategies such as host-directed therapeutics and adjunctive therapies that enhance the effect of existing antibiotics have emerged to better control Mtb infection. Recent advances in understanding the metabolic interplay between host immune cells and pathogens have provided new insights into how their interactions ultimately influence disease outcomes and antibiotic-treatment efficacy. In this review, we describe how metabolic cascades in immune environments and relevant metabolites produced from immune cells during Mtb infection play critical roles in the progression of diseases and induction of anti-Mtb protective immunity. In addition, we introduce how metabolic alterations in Mtb itself can lead to the development of persister cells that are resistant to host immunity and can eventually evade antibiotic attacks. Further understanding of the metabolic link between host cells and Mtb may contribute to not only the prevention of Mtb persister development but also the optimization of host anti-Mtb immunity together with enhanced efficacy of existing antibiotics. Overall, this review highlights novel approaches to improve and develop host-mediated therapeutic strategies against Mtb infection by restoring and switching pathogen-favoring metabolic conditions with host-favoring conditions.
Collapse
Affiliation(s)
- Ji-Hae Park
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Dahee Shim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Keu Eun San Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
27
|
De Novo Cobalamin Biosynthesis, Transport, and Assimilation and Cobalamin-Mediated Regulation of Methionine Biosynthesis in Mycobacterium smegmatis. J Bacteriol 2021; 203:JB.00620-20. [PMID: 33468593 PMCID: PMC8088520 DOI: 10.1128/jb.00620-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in cobalamin-dependent metabolism have marked the evolution of Mycobacterium tuberculosis into a human pathogen. However, the role(s) of cobalamin in mycobacterial physiology remain poorly understood. Cobalamin is an essential cofactor in all domains of life, yet its biosynthesis is restricted to some bacteria and archaea. Mycobacterium smegmatis, an environmental saprophyte frequently used as surrogate for the obligate human pathogen M. tuberculosis, carries approximately 30 genes predicted to be involved in de novo cobalamin biosynthesis. M. smegmatis also encodes multiple cobalamin-dependent enzymes, including MetH, a methionine synthase that catalyzes the final reaction in methionine biosynthesis. In addition to metH, M. smegmatis possesses a cobalamin-independent methionine synthase, metE, suggesting that enzyme use—MetH versus MetE—is regulated by cobalamin availability. Consistent with this notion, we previously described a cobalamin-sensing riboswitch controlling metE expression in M. tuberculosis. Here, we apply a targeted mass spectrometry-based approach to confirm de novo cobalamin biosynthesis in M. smegmatis during aerobic growth in vitro. We also demonstrate that M. smegmatis can transport and assimilate exogenous cyanocobalamin (CNCbl; also known as vitamin B12) and its precursor, dicyanocobinamide ([CN]2Cbi). However, the uptake of CNCbl and (CN)2Cbi in this organism is restricted and seems dependent on the conditional essentiality of the cobalamin-dependent methionine synthase. Using gene and protein expression analyses combined with single-cell growth kinetics and live-cell time-lapse microscopy, we show that transcription and translation of metE are strongly attenuated by endogenous cobalamin. These results support the inference that metH essentiality in M. smegmatis results from riboswitch-mediated repression of MetE expression. Moreover, differences observed in cobalamin-dependent metabolism between M. smegmatis and M. tuberculosis provide some insight into the selective pressures which might have shaped mycobacterial metabolism for pathogenicity. IMPORTANCE Alterations in cobalamin-dependent metabolism have marked the evolution of Mycobacterium tuberculosis into a human pathogen. However, the role(s) of cobalamin in mycobacterial physiology remains poorly understood. Using the nonpathogenic saprophyte M. smegmatis, we investigated the production of cobalamin, transport and assimilation of cobalamin precursors, and the role of cobalamin in regulating methionine biosynthesis. We confirm constitutive de novo cobalamin biosynthesis in M. smegmatis, in contrast with M. tuberculosis, which appears to lack de novo cobalamin biosynthetic capacity. We also show that uptake of cyanocobalamin (vitamin B12) and its precursors is restricted in M. smegmatis, apparently depending on the cofactor requirements of the cobalamin-dependent methionine synthase. These observations establish M. smegmatis as an informative foil to elucidate key metabolic adaptations enabling mycobacterial pathogenicity.
Collapse
|
28
|
Orgeur M, Frigui W, Pawlik A, Clark S, Williams A, Ates LS, Ma L, Bouchier C, Parkhill J, Brodin P, Brosch R. Pathogenomic analyses of Mycobacterium microti, an ESX-1-deleted member of the Mycobacterium tuberculosis complex causing disease in various hosts. Microb Genom 2021; 7:000505. [PMID: 33529148 PMCID: PMC8208694 DOI: 10.1099/mgen.0.000505] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/16/2020] [Indexed: 01/03/2023] Open
Abstract
Mycobacterium microti is an animal-adapted member of the Mycobacterium tuberculosis complex (MTBC), which was originally isolated from voles, but has more recently also been isolated from other selected mammalian hosts, including occasionally from humans. Here, we have generated and analysed the complete genome sequences of five representative vole and clinical M. microti isolates using PacBio- and Illumina-based technologies, and have tested their virulence and vaccine potential in SCID (severe combined immune deficient) mouse and/or guinea pig infection models. We show that the clinical isolates studied here cluster separately in the phylogenetic tree from vole isolates and other clades from publicly available M. microti genome sequences. These data also confirm that the vole and clinical M. microti isolates were all lacking the specific RD1mic region, which in other tubercle bacilli encodes the ESX-1 type VII secretion system. Biochemical analysis further revealed marked phenotypic differences between isolates in type VII-mediated secretion of selected PE and PPE proteins, which in part were attributed to specific genetic polymorphisms. Infection experiments in the highly susceptible SCID mouse model showed that the clinical isolates were significantly more virulent than the tested vole isolates, but still much less virulent than the M. tuberculosis H37Rv control strain. The strong attenuation of the ATCC 35872 vole isolate in immunocompromised mice, even compared to the attenuated BCG (bacillus Calmette-Guérin) vaccine, and its historic use in human vaccine trials encouraged us to test this strain's vaccine potential in a guinea pig model, where it demonstrated similar protective efficacy as a BCG control, making it a strong candidate for vaccination of immunocompromised individuals in whom BCG vaccination is contra-indicated. Overall, we provide new insights into the genomic and phenotypic variabilities and particularities of members of an understudied clade of the MTBC, which all share a recent common ancestor that is characterized by the deletion of the RD1mic region.
Collapse
Affiliation(s)
- Mickael Orgeur
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Paris 75015, France
| | - Wafa Frigui
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Paris 75015, France
| | - Alexandre Pawlik
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Paris 75015, France
| | - Simon Clark
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Ann Williams
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Louis S. Ates
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Paris 75015, France
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection & Immunity, Meibergdreef 9, Amsterdam, Netherlands
| | - Laurence Ma
- Institut Pasteur, Biomics, C2RT, Paris 75015, France
| | | | - Julian Parkhill
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Priscille Brodin
- CIIL - Center for Infection and Immunity of Lille, Université de Lille/CNRS UMR 9017/INSERM U1019/CHU Lille/Institut Pasteur de Lille, Lille 59000, France
| | - Roland Brosch
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Paris 75015, France
| |
Collapse
|
29
|
[Predictive factors and biomarkers of exudative lymphocytic tuberculous pleurisy]. Rev Mal Respir 2021; 38:231-239. [PMID: 33531187 DOI: 10.1016/j.rmr.2020.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 10/02/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Tuberculosis is the leading cause of exudative lymphocytic pleural effusion. OBJECTIVE To determine the predictive factors and assess the diagnostic performance of biomarkers for exudative lymphocytic tuberculous pleural effusion found in our practice. METHODS Case-control study from 01 January 2015 to 30 August 2019, including patients admitted to the day-hospital facility for pleural biopsy. Histopathology of pleural biopsy fragments was the gold standard for assessing the diagnostic performance of the biomarkers studied (pleural adenosine deaminase, geneXpert and pleural fluid culture). RESULTS One hundred and nine patients were included. The average age was 35.7±18.1 years. In all, 72.5% of them were diagnosed with tuberculosis. After multivariate analysis, only patients aged under 35 years old were found to be at risk for exudative lymphocytic tuberculous pleural effusion (adjusted odds ratio: 9.18 [1.99; 42.28], P=0.004). Histopathology was suggestive of tuberculosis in 84.8% of cases (P<0.0001). The sensitivity of geneXpert was 42.1% and the specificity was 100%, with a mean concordance rate with histopathology (k=0.46). One case of rifampicin-resistant tuberculosis was detected by geneXpert (2.2%). The diagnostic performance of pleural adenosine deaminase varies according to the defined positivity threshold. For values greater than 70UI/L, its sensitivity was 25% and its specificity was 86.7%. The area under the ROC curve of the pleural adenosine deaminase was 0.70. CONCLUSION In patients with exudative lymphocytic pleural effusion, young age is a risk factor for pleural tuberculosis. The geneXpert, although not very sensitive, is a moderately efficient and specific test for tuberculosis. Pleural adenosine deaminase is a discriminating and useful biomarker for the diagnosis of tuberculosis.
Collapse
|
30
|
Korol CB, Shallom SJ, Arora K, Boshoff HI, Freeman AF, King A, Agrawal S, Daugherty SC, Jancel T, Kabat J, Ganesan S, Torrero MN, Sampaio EP, Barry C, Holland SM, Tettelin H, Rosenzweig SD, Zelazny AM. Tissue specific diversification, virulence and immune response to Mycobacterium bovis BCG in a patient with an IFN-γ R1 deficiency. Virulence 2020; 11:1656-1673. [PMID: 33356838 PMCID: PMC7781554 DOI: 10.1080/21505594.2020.1848108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/25/2020] [Accepted: 11/05/2020] [Indexed: 11/16/2022] Open
Abstract
Summary: We characterized Mycobacterium bovis BCG isolates found in lung and brain samples from a previously vaccinated patient with IFNγR1 deficiency. The isolates collected displayed distinct genomic and phenotypic features consistent with host adaptation and associated changes in antibiotic susceptibility and virulence traits. Background: We report a case of a patient with partial recessive IFNγR1 deficiency who developed disseminated BCG infection after neonatal vaccination (BCG-vaccine). Distinct M. bovis BCG-vaccine derived clinical strains were recovered from the patient's lungs and brain. Methods: BCG strains were phenotypically (growth, antibiotic susceptibility, lipid) and genetically (whole genome sequencing) characterized. Mycobacteria cell infection models were used to assess apoptosis, necrosis, cytokine release, autophagy, and JAK-STAT signaling. Results: Clinical isolates BCG-brain and BCG-lung showed distinct Rv0667 rpoB mutations conferring high- and low-level rifampin resistance; the latter displayed clofazimine resistance through Rv0678 gene (MarR-like transcriptional regulator) mutations. BCG-brain and BCG-lung showed mutations in fadA2, fadE5, and mymA operon genes, respectively. Lipid profiles revealed reduced levels of PDIM in BCG-brain and BCG-lung and increased TAGs and Mycolic acid components in BCG-lung, compared to parent BCG-vaccine. In vitro infected cells showed that the BCG-lung induced a higher cytokine release, necrosis, and cell-associated bacterial load effect when compared to BCG-brain; conversely, both strains inhibited apoptosis and altered JAK-STAT signaling. Conclusions: During a chronic-disseminated BCG infection, BCG strains can evolve independently at different sites likely due to particular microenvironment features leading to differential antibiotic resistance, virulence traits resulting in dissimilar responses in different host tissues.
Collapse
Affiliation(s)
- Cecilia B. Korol
- Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, USA
| | | | - Kriti Arora
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Helena I. Boshoff
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Alejandra King
- Department of Pediatric Immunology, Hospital Luis Calvo MacKenna, Universidad De, Chile, Chile
| | - Sonia Agrawal
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA
| | - Sean C. Daugherty
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA
| | - Timothy Jancel
- Department of Pharmacy, Clinical Center, NIH, Bethesda, USA
| | - Juraj Kabat
- Department Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Sundar Ganesan
- Department Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Marina N. Torrero
- Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, USA
| | - Elizabeth P. Sampaio
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Clifton Barry
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Steve M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA
| | | | - Adrian M. Zelazny
- Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, USA
| |
Collapse
|
31
|
Kanipe C, Palmer MV. Mycobacterium bovis and you: A comprehensive look at the bacteria, its similarities to Mycobacterium tuberculosis, and its relationship with human disease. Tuberculosis (Edinb) 2020; 125:102006. [PMID: 33032093 DOI: 10.1016/j.tube.2020.102006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022]
Abstract
While Mycobacterium tuberculosis is the primary cause of tuberculosis in people, multiple other mycobacteria are capable of doing so. With the World Health Organization's goal of a 90% reduction in tuberculosis by 2035, all tuberculous mycobacteria need to be addressed. Understanding not only the similarities, but importantly the differences between the different species is crucial if eradication is ever to be achieved. Mycobacterium bovis, while typically thought of as a disease of cattle, remains a possible source of human infection worldwide. Although this species' genome differs from Mycobacterium tuberculosis by only 0.05%, significant differences are present, creating unique challenges to address. This review focuses on features which distinguish this bacterium from Mycobacterium tuberculosis, including differences in origin, structure, environmental persistence, host preferences, infection and disease, host immune response, diagnostics and treatment.
Collapse
Affiliation(s)
- Carly Kanipe
- Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA; Immunobiology Graduate Program, Iowa State University, Ames, IA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA.
| | - Mitchell V Palmer
- Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| |
Collapse
|
32
|
Comprehensive analysis of protein acetyltransferases of human pathogen Mycobacterium tuberculosis. Biosci Rep 2020; 39:221456. [PMID: 31820790 PMCID: PMC6923341 DOI: 10.1042/bsr20191661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB), a leading infectious disease caused by Mycobacterium tuberculosis strain, takes four human lives every minute globally. Paucity of knowledge on M. tuberculosis virulence and antibiotic resistance is the major challenge for tuberculosis control. We have identified 47 acetyltransferases in the M. tuberculosis, which use diverse substrates including antibiotic, amino acids, and other chemical molecules. Through comparative analysis of the protein file of the virulent M. tuberculosis H37Rv strain and the avirulent M. tuberculosis H37Ra strain, we identified one acetyltransferase that shows significant variations with N-terminal deletion, possibly influencing its physicochemical properties. We also found that one acetyltransferase has three types of post-translation modifications (lysine acetylation, succinylation, and glutarylation). The genome context analysis showed that many acetyltransferases with their neighboring genes belong to one operon. By data mining from published transcriptional profiles of M. tuberculosis exposed to diverse treatments, we revealed that several acetyltransferases may be functional during M. tuberculosis infection. Insights obtained from the present study can potentially provide clues for developing novel TB therapeutic interventions.
Collapse
|
33
|
Peters JS, Ismail N, Dippenaar A, Ma S, Sherman DR, Warren RM, Kana BD. Genetic Diversity in Mycobacterium tuberculosis Clinical Isolates and Resulting Outcomes of Tuberculosis Infection and Disease. Annu Rev Genet 2020; 54:511-537. [PMID: 32926793 DOI: 10.1146/annurev-genet-022820-085940] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tuberculosis claims more human lives than any other bacterial infectious disease and represents a clear and present danger to global health as new tools for vaccination, treatment, and interruption of transmission have been slow to emerge. Additionally, tuberculosis presents with notable clinical heterogeneity, which complicates diagnosis, treatment, and the establishment of nonrelapsing cure. How this heterogeneity is driven by the diversity ofclinical isolates of the causative agent, Mycobacterium tuberculosis, has recently garnered attention. Herein, we review advances in the understanding of how naturally occurring variation in clinical isolates affects transmissibility, pathogenesis, immune modulation, and drug resistance. We also summarize how specific changes in transcriptional responses can modulate infection or disease outcome, together with strain-specific effects on gene essentiality. Further understanding of how this diversity of M. tuberculosis isolates affects disease and treatment outcomes will enable the development of more effective therapeutic options and vaccines for this dreaded disease.
Collapse
Affiliation(s)
- Julian S Peters
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg 2000, South Africa; ,
| | - Nabila Ismail
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; ,
| | - Anzaan Dippenaar
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; , .,Family Medicine and Population Health (FAMPOP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, 2000, Belgium;
| | - Shuyi Ma
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - David R Sherman
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Robin M Warren
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; ,
| | - Bavesh D Kana
- Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg 2000, South Africa; ,
| |
Collapse
|
34
|
Performance and Application of 16S rRNA Gene Cycle Sequencing for Routine Identification of Bacteria in the Clinical Microbiology Laboratory. Clin Microbiol Rev 2020; 33:33/4/e00053-19. [PMID: 32907806 DOI: 10.1128/cmr.00053-19] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review provides a state-of-the-art description of the performance of Sanger cycle sequencing of the 16S rRNA gene for routine identification of bacteria in the clinical microbiology laboratory. A detailed description of the technology and current methodology is outlined with a major focus on proper data analyses and interpretation of sequences. The remainder of the article is focused on a comprehensive evaluation of the application of this method for identification of bacterial pathogens based on analyses of 16S multialignment sequences. In particular, the existing limitations of similarity within 16S for genus- and species-level differentiation of clinically relevant pathogens and the lack of sequence data currently available in public databases is highlighted. A multiyear experience is described of a large regional clinical microbiology service with direct 16S broad-range PCR followed by cycle sequencing for direct detection of pathogens in appropriate clinical samples. The ability of proteomics (matrix-assisted desorption ionization-time of flight) versus 16S sequencing for bacterial identification and genotyping is compared. Finally, the potential for whole-genome analysis by next-generation sequencing (NGS) to replace 16S sequencing for routine diagnostic use is presented for several applications, including the barriers that must be overcome to fully implement newer genomic methods in clinical microbiology. A future challenge for large clinical, reference, and research laboratories, as well as for industry, will be the translation of vast amounts of accrued NGS microbial data into convenient algorithm testing schemes for various applications (i.e., microbial identification, genotyping, and metagenomics and microbiome analyses) so that clinically relevant information can be reported to physicians in a format that is understood and actionable. These challenges will not be faced by clinical microbiologists alone but by every scientist involved in a domain where natural diversity of genes and gene sequences plays a critical role in disease, health, pathogenicity, epidemiology, and other aspects of life-forms. Overcoming these challenges will require global multidisciplinary efforts across fields that do not normally interact with the clinical arena to make vast amounts of sequencing data clinically interpretable and actionable at the bedside.
Collapse
|
35
|
Lekko YM, Ooi PT, Omar S, Mazlan M, Ramanoon SZ, Jasni S, Jesse FFA, Che-Amat A. Mycobacterium tuberculosis complex in wildlife: Review of current applications of antemortem and postmortem diagnosis. Vet World 2020; 13:1822-1836. [PMID: 33132593 PMCID: PMC7566238 DOI: 10.14202/vetworld.2020.1822-1836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/16/2020] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) is a chronic inflammatory and zoonotic disease caused by Mycobacterium tuberculosis complex (MTBC) members, which affects various domestic animals, wildlife, and humans. Some wild animals serve as reservoir hosts in the transmission and epidemiology of the disease. Therefore, the monitoring and surveillance of both wild and domestic hosts are critical for prevention and control strategies. For TB diagnosis, the single intradermal tuberculin test or the single comparative intradermal tuberculin test, and the gamma-interferon test, which is regarded as an ancillary test, are used. Postmortem examination can identify granulomatous lesions compatible with a diagnosis of TB. In contrast, smears of the lesions can be stained for acid-fast bacilli, and samples of the affected organs can be subjected to histopathological analyses. Culture is the gold standard test for isolating mycobacterial bacilli because it has high sensitivity and specificity compared with other methods. Serology for antibody detection allows the testing of many samples simply, rapidly, and inexpensively, and the protocol can be standardized in different laboratories. Molecular biological analyses are also applicable to trace the epidemiology of the disease. In conclusion, reviewing the various techniques used in MTBC diagnosis can help establish guidelines for researchers when choosing a particular diagnostic method depending on the situation at hand, be it disease outbreaks in wildlife or for epidemiological studies. This is because a good understanding of various diagnostic techniques will aid in monitoring and managing emerging pandemic threats of infectious diseases from wildlife and also preventing the potential spread of zoonotic TB to livestock and humans. This review aimed to provide up-to-date information on different techniques used for diagnosing TB at the interfaces between wildlife, livestock, and humans.
Collapse
Affiliation(s)
- Yusuf Madaki Lekko
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Maiduguri, 1069 PMB, Maiduguri, Borno State, Nigeria
| | - Peck Toung Ooi
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sharina Omar
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Mazlina Mazlan
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Siti Zubaidah Ramanoon
- Department of Farm and Exotic Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sabri Jasni
- Department of Paraclinical, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, 16100 Kota Bharu, Kelantan, Malaysia
| | - Faez Firdaus Abdullah Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Azlan Che-Amat
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
36
|
Antimicrobial susceptibility testing of Mycobacterium tuberculosis complex isolates - the EUCAST broth microdilution reference method for MIC determination. Clin Microbiol Infect 2020; 26:1488-1492. [PMID: 32750539 DOI: 10.1016/j.cmi.2020.07.036] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/03/2023]
Abstract
SCOPE Several methods are used worldwide for antibiotic susceptibility testing (AST) for the Mycobacterium tuberculosis complex (MTBC). The variability in the results obtained with these methods hampers setting epidemiological cut-off (ECOFF) values and clinical breakpoints according to EUCAST guidelines. Methods for susceptibility testing and determination of the minimal inhibitory concentrations (MICs) need to be standardized for MTBC isolates for old and new agents. Our objective was to establish a standardized reference method for MIC determination for MTBC. METHODS The EUCAST antimycobacterial susceptibility testing subcommittee (AMST) compared protocols of MIC determination with regard to medium, inoculum preparation, antituberculous agent preparation, incubation, reading of the results and interpretation. RECOMMENDATIONS The EUCAST reference method of MIC determination for MTBC is the broth microdilution method in Middlebrook 7H9-10% OADC medium. The final inoculum is a 105 CFU/mL suspension, obtained from a 10-2 dilution of a 0.5 McFarland suspension prepared after vortexing bacterial colonies with glass beads before suspending them in sterile water. The culture is maintained in a U-shaped 96-well polystyrene microtitre sterile plate with a lid incubated at 36° ± 1°C. Reading is done using an inverted mirror as soon as the 1:100 diluted control (i.e. 103 CFU/mL suspension) shows visual growth. The MIC, expressed in mg/L, is the lowest concentration that inhibits visual growth. Mycobacterium tuberculosis H37Rv ATCC 27294 is used as the reference strain and its targeted MIC values are within the range 0.03-0.12 for isoniazid, 0.12-0.5 for levofloxacin and 0.25-1 mg/L for amikacin. CONCLUSIONS The EUCAST reference method for MTBC was endorsed by EUCAST after public consultation and will from now on be used to define EUCAST ECOFFs and clinical breakpoints. This reference method is not primarily intended to be used under routine conditions and the AST methods will need to be calibrated against this reference method to be used with EUCAST breakpoints.
Collapse
|
37
|
Sabio Y García J, Bigi MM, Klepp LI, García EA, Blanco FC, Bigi F. Does Mycobacterium bovis persist in cattle in a non-replicative latent state as Mycobacterium tuberculosis in human beings? Vet Microbiol 2020; 247:108758. [PMID: 32768211 DOI: 10.1016/j.vetmic.2020.108758] [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: 03/26/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/13/2022]
Abstract
Members of the Mycobacterium tuberculosis complex (MTBC) are responsible for tuberculosis in several mammals. In this complex, Mycobacterium tuberculosis and Mycobacterium bovis, which are closely related, show host preference for humans and cattle, respectively. Although human and bovine tuberculosis are clinically similar, M. tuberculosis mostly causes latent infection in humans, whereas M. bovis frequently leads to an acute infection in cattle. This review attempts to connect the pathology in experimental animal models as well as the cellular responses to M. bovis and M. tuberculosis regarding the differences in protein expression and regulatory mechanisms of both pathogens that could explain their apparent divergent latency behaviour. The occurrence of latent bovine tuberculosis (bTB) would represent a serious complication for the eradication of the disease in cattle, with the risk of onward transmission to humans. Thus, understanding the physiological events that may lead to the state of latency in bTB could assist in the development of appropriate prevention and control tools.
Collapse
Affiliation(s)
- Julia Sabio Y García
- (Instituto de Biotecnología-IABIMO, INTA-CONICET), Institute of Biotechnology-IABIMO, National Institute of Agricultural Technology (INTA) and National Scientific and Technical Research Council (CONICET), Argentina.
| | - María M Bigi
- (Universidad de Buenos Aires, Facultad de Agronomía), University of Buenos Aires, School of Agronomy Facultad de Agronomía, UBA, Buenos Aires Argentina.
| | - Laura I Klepp
- (Instituto de Biotecnología-IABIMO, INTA-CONICET), Institute of Biotechnology-IABIMO, National Institute of Agricultural Technology (INTA) and National Scientific and Technical Research Council (CONICET), Argentina.
| | - Elizabeth A García
- (Instituto de Biotecnología-IABIMO, INTA-CONICET), Institute of Biotechnology-IABIMO, National Institute of Agricultural Technology (INTA) and National Scientific and Technical Research Council (CONICET), Argentina.
| | - Federico C Blanco
- (Instituto de Biotecnología-IABIMO, INTA-CONICET), Institute of Biotechnology-IABIMO, National Institute of Agricultural Technology (INTA) and National Scientific and Technical Research Council (CONICET), Argentina.
| | - Fabiana Bigi
- (Instituto de Biotecnología-IABIMO, INTA-CONICET), Institute of Biotechnology-IABIMO, National Institute of Agricultural Technology (INTA) and National Scientific and Technical Research Council (CONICET), Argentina.
| |
Collapse
|
38
|
R NV, G H T, R DV, M H, H H, N S. Lateral Flow Genochromatographic Strip for Naked-Eye Detection of Mycobacterium Tuberculosis PCR Products with Gold Nanoparticles as a Reporter. J Biomed Phys Eng 2020; 10:307-318. [PMID: 32637375 PMCID: PMC7321395 DOI: 10.31661/jbpe.v0i0.1912-1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/14/2020] [Indexed: 11/16/2022]
Abstract
Background Mycobacterium tuberculosis (MTB) is a pathogen causing tuberculosis (TB) in human, and TB can cause enormous social and economic disruptions. Lateral flow test strips (LFTSs) are inexpensive, portable, disposable, rapid, and easy-to-use analytical tools. Objective LFTSs were prepared for the detection of MTB. LFTSs were fabricated using a new specific probe for MTB H37Rv, based on IS6110 sequence gene, and tailed with poly deoxyadenine (dA). Material and Methods In this experimental study, to create test and control zones, streptavidin (STP) and a 150-mer dA were dotted on a nitrocellolose membrane. Gold nanoparticles (GNPs) were conjugated with poly deoxythymidine sequence and placed on the conjugate pad. The composition of immersion buffers for sample pad and conjugate pad, running solution, solutions of GNPs-S-dT150 and STP were introduced. DNA genome of MTB and Mycobacterium bovis in clinical samples was amplified with PCR, and then detected by the LFTSs. During the assay, samples were firstly hybridized in two steps and then placed on a conjugate pad in a manner that positive and negative samples provided two and one red lines, respectively, on the detection pad. Results After PCR reaction with biotinylated primer, hybridization process with specific MTB probe-dA70-100 toke 10 min, and running process on the strip was performed within 5 min. Conclusion We showed that LFTS can discriminate a particular bacteria strain from others. The LFTSs can be redesigned for detection of other pathogenic genomes.
Collapse
Affiliation(s)
- Nazari-Vanani R
- MSc, Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tondro G H
- MSc, Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Dehdari Vais R
- MSc, Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haghkhah M
- PhD, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Heli H
- PhD, Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sattarahmady N
- PhD, Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- PhD, Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
39
|
Crosskey TD, Beckham KS, Wilmanns M. The ATPases of the mycobacterial type VII secretion system: Structural and mechanistic insights into secretion. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 152:25-34. [DOI: 10.1016/j.pbiomolbio.2019.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
|
40
|
Sousa J, Cá B, Maceiras AR, Simões-Costa L, Fonseca KL, Fernandes AI, Ramos A, Carvalho T, Barros L, Magalhães C, Chiner-Oms Á, Machado H, Veiga MI, Singh A, Pereira R, Amorim A, Vieira J, Vieira CP, Bhatt A, Rodrigues F, Rodrigues PNS, Gagneux S, Castro AG, Guimarães JT, Bastos HN, Osório NS, Comas I, Saraiva M. Mycobacterium tuberculosis associated with severe tuberculosis evades cytosolic surveillance systems and modulates IL-1β production. Nat Commun 2020; 11:1949. [PMID: 32327653 PMCID: PMC7181847 DOI: 10.1038/s41467-020-15832-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/23/2020] [Indexed: 01/26/2023] Open
Abstract
Genetic diversity of Mycobacterium tuberculosis affects immune responses and clinical outcomes of tuberculosis (TB). However, how bacterial diversity orchestrates immune responses to direct distinct TB severities is unknown. Here we study 681 patients with pulmonary TB and show that M. tuberculosis isolates from cases with mild disease consistently induce robust cytokine responses in macrophages across multiple donors. By contrast, bacteria from patients with severe TB do not do so. Secretion of IL-1β is a good surrogate of the differences observed, and thus to classify strains as probable drivers of different TB severities. Furthermore, we demonstrate that M. tuberculosis isolates that induce low levels of IL-1β production can evade macrophage cytosolic surveillance systems, including cGAS and the inflammasome. Isolates exhibiting this evasion strategy carry candidate mutations, generating sigA recognition boxes or affecting components of the ESX-1 secretion system. Therefore, we provide evidence that M. tuberculosis strains manipulate host-pathogen interactions to drive variable TB severities.
Collapse
Grants
- The authors thank the excellent support from the i3S scientific platforms, namely Animal facility, Advanced Light Microscopy and BioSciences Screening, member of the national infrastructure PPBI - Portuguese Platform of Bioimaging (PPBI-POCI-01-0145-FEDER-022122). This work was financed by FCT - Fundação para a Ciência e a Tecnologia/ Ministério da Ciência, Tecnologia e Inovação grant POCI-01-0145-FEDER-028955 (to MS) and by the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (FEDER) (NORTE-01-0145-FEDER-000013, to MIV, FR, AGC and NSO). IC acknowledges the support of Ministerio de Ciencia, Innovación y Universidades (SAF2016-77346-R) and the European Research Council (638553-TB-ACCELERATE). HNB acknowledges the support of Bolsa D. Manuel de Mello and of the Portuguese Society for Pneumology; AB and MS were also recipients of an International Exchanges Grant from the Royal Society. JS is funded by a research fellow NORTE-01-0145-FEDER-000012; BC and KLF are funded by FCT PhD scholarships SFRH/BD/114403/2016 and SFRH/BD/114405/2016, respectively; MIV is funded by FCT through DL 57/2016 (CRP) and MS through Estimulo Individual ao Emprego Científico.
Collapse
Affiliation(s)
- Jeremy Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
- Doctoral Program in Molecular and Cell Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Baltazar Cá
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
- Doctoral Program in Molecular and Cell Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Raquel Maceiras
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Luisa Simões-Costa
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Kaori L Fonseca
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
- Doctoral Program in Molecular and Cell Biology, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Isabel Fernandes
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Angélica Ramos
- São João Hospital Center & EPIUnit-Institute of Public Health, University of Porto, Porto, Portugal
| | - Teresa Carvalho
- São João Hospital Center & EPIUnit-Institute of Public Health, University of Porto, Porto, Portugal
| | - Leandro Barros
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Carlos Magalhães
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Henrique Machado
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Isabel Veiga
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Albel Singh
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Rui Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
| | - António Amorim
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
- Faculty of Sciences, University of Porto, Porto, Portugal
| | - Jorge Vieira
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Cristina P Vieira
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Apoorva Bhatt
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Fernando Rodrigues
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pedro N S Rodrigues
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Sebastien Gagneux
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - António Gil Castro
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Tiago Guimarães
- São João Hospital Center & EPIUnit-Institute of Public Health, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Helder Novais Bastos
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- São João Hospital Center, Porto, Portugal
| | - Nuno S Osório
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Iñaki Comas
- Biomedicine Institute of Valencia (CSIC), Valencia, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Margarida Saraiva
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
- IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal.
| |
Collapse
|
41
|
Sapriel G, Brosch R. Shared Pathogenomic Patterns Characterize a New Phylotype, Revealing Transition toward Host-Adaptation Long before Speciation of Mycobacterium tuberculosis. Genome Biol Evol 2020; 11:2420-2438. [PMID: 31368488 PMCID: PMC6736058 DOI: 10.1093/gbe/evz162] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Tuberculosis remains one of the deadliest infectious diseases of humanity. To better understand the evolutionary history of host-adaptation of tubercle bacilli (MTB), we sought for mycobacterial species that were more closely related to MTB than the previously used comparator species Mycobacterium marinum and Mycobacterium kansasii. Our phylogenomic approach revealed some recently sequenced opportunistic mycobacterial pathogens, Mycobacterium decipiens, Mycobacterium lacus, Mycobacterium riyadhense, and Mycobacterium shinjukuense, to constitute a common clade with MTB, hereafter called MTB-associated phylotype (MTBAP), from which MTB have emerged. Multivariate and clustering analyses of genomic functional content revealed that the MTBAP lineage forms a clearly distinct cluster of species that share common genomic characteristics, such as loss of core genes, shift in dN/dS ratios, and massive expansion of toxin–antitoxin systems. Consistently, analysis of predicted horizontal gene transfer regions suggests that putative functions acquired by MTBAP members were markedly associated with changes in microbial ecology, for example adaption to intracellular stress resistance. Our study thus considerably deepens our view on MTB evolutionary history, unveiling a decisive shift that promoted conversion to host-adaptation among ancestral founders of the MTBAP lineage long before Mycobacterium tuberculosis has adapted to the human host.
Collapse
Affiliation(s)
- Guillaume Sapriel
- UFR des Sciences de La Santé, Université de Versailles St. Quentin, Montigny le Bretonneux, France.,Atelier de Bioinformatique, ISYEB, UMR 7205, Paris, France
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, CNRS UMR 3525, Paris, France
| |
Collapse
|
42
|
TbD1 deletion as a driver of the evolutionary success of modern epidemic Mycobacterium tuberculosis lineages. Nat Commun 2020; 11:684. [PMID: 32019932 PMCID: PMC7000671 DOI: 10.1038/s41467-020-14508-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 01/09/2020] [Indexed: 11/29/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) strains are classified into different phylogenetic lineages (L), three of which (L2/L3/L4) emerged from a common progenitor after the loss of the MmpS6/MmpL6-encoding Mtb-specific deletion 1 region (TbD1). These TbD1-deleted “modern” lineages are responsible for globally-spread tuberculosis epidemics, whereas TbD1-intact “ancestral” lineages tend to be restricted to specific geographical areas, such as South India and South East Asia (L1) or East Africa (L7). By constructing and characterizing a panel of recombinant TbD1-knock-in and knock-out strains and comparison with clinical isolates, here we show that deletion of TbD1 confers to Mtb a significant increase in resistance to oxidative stress and hypoxia, which correlates with enhanced virulence in selected cellular, guinea pig and C3HeB/FeJ mouse infection models, the latter two mirroring in part the development of hypoxic granulomas in human disease progression. Our results suggest that loss of TbD1 at the origin of the L2/L3/L4 Mtb lineages was a key driver for their global epidemic spread and outstanding evolutionary success. Mycobacterium tuberculosis (Mtb) modern strains emerged from a common progenitor after the loss of Mtb-specific deletion 1 region (TbD1). Here, the authors show that deletion of TbD1 correlates with enhanced Mtb virulence in animal models, mirroring the development of hypoxic granulomas in human disease progression.
Collapse
|
43
|
Abstract
Natural products from microorganisms are important small molecules that play roles in various biological processes like cellular growth, motility, nutrient acquisition, stress response, biofilm formation, and defense. It is hypothesized that pathogens exploit these molecules to regulate virulence and persistence during infections. Here, we present selected examples of signaling natural products from human pathogenic bacteria that use these metabolites to gain a competitive advantage. Targeting these signaling systems provides novel strategies to antimicrobial treatments.
Collapse
Affiliation(s)
- Zhijuan Hu
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, 201 Gilman Hall, Berkeley, California 94720, United States
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, 201 Gilman Hall, Berkeley, California 94720, United States
- Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| |
Collapse
|
44
|
Levillain F, Kim H, Woong Kwon K, Clark S, Cia F, Malaga W, Lanni F, Brodin P, Gicquel B, Guilhot C, Bancroft GJ, Williams A, Jae Shin S, Poquet Y, Neyrolles O. Preclinical assessment of a new live attenuated Mycobacterium tuberculosis Beijing-based vaccine for tuberculosis. Vaccine 2019; 38:1416-1423. [PMID: 31862194 DOI: 10.1016/j.vaccine.2019.11.085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/14/2019] [Accepted: 11/29/2019] [Indexed: 01/16/2023]
Abstract
Tuberculosis still claims more lives than any other pathogen, and a vaccine better than BCG is urgently needed. One of the challenges for novel TB vaccines is to protect against all Mycobacterium tuberculosis lineages, including the most virulent ones, such as the Beijing lineage. Here we developed a live attenuated M. tuberculosis mutant derived from GC1237, a Beijing strain responsible for tuberculosis outbreaks in the Canary Islands. The mutant strain is inactivated both in the Rv1503c gene, responsible for surface glycolipid synthesis, and in the two-component global regulator PhoPR. This double mutant is as safe as BCG in immunodeficient SCID mice. In immune-competent mice and guinea pigs, the mutant is as protective as BCG against M. tuberculosis strains of common lineage 4 (Euro-American). By contrast, in mice the vaccine is protective against a M. tuberculosis strain of lineage 2 (East-Asian, Beijing), while BCG is not. These results highlight differences in protection efficacy of live attenuated M. tuberculosis-derived vaccine candidates depending on their genetic background, and provide insights for the development of novel live vaccines against TB, especially in East-Asian countries where M. tuberculosis strains of the Beijing family are highly dominant.
Collapse
Affiliation(s)
- Florence Levillain
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, South Korea
| | - Simon Clark
- Public Health England, Salisbury, United Kingdom
| | - Felipe Cia
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Wladimir Malaga
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, South Korea
| | - Faye Lanni
- Public Health England, Salisbury, United Kingdom
| | - Priscille Brodin
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Brigitte Gicquel
- Unité de Génétique Mycobactérienne, Institut Pasteur, Paris, France; Department of Tuberculosis Control and Prevention, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Christophe Guilhot
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Ann Williams
- Public Health England, Salisbury, United Kingdom
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, South Korea
| | - Yannick Poquet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
45
|
Cerezo-Cortés MI, Rodríguez-Castillo JG, Hernández-Pando R, Murcia MI. Circulation of M. tuberculosis Beijing genotype in Latin America and the Caribbean. Pathog Glob Health 2019; 113:336-351. [PMID: 31903874 PMCID: PMC7006823 DOI: 10.1080/20477724.2019.1710066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lineage 2 (East Asian), which includes the Beijing genotype, is one of the most prevalent lineages of Mycobacterium tuberculosis (Mtb) throughout the world. The Beijing family is associated to hypervirulence and drug-resistant tuberculosis. The study of this genotype's circulation in Latin America is crucial for achieving total control of TB, the goal established by the World Health Organization, for the American sub-continent, before 2035. In this sense, the present work presents an overview of the status of the Beijing genotype for this region, with a bibliographical review, and data analysis of MIRU-VNTRs for available Beijing isolates. Certain countries present a prevalent trend of <5%, suggesting low transmissibility for the region, with the exception of Cuba (17.2%), Perú (16%) and Colombia (5%). Minimum Spanning Tree analysis, obtained from MIRU-VNTR data, shows distribution of specific clonal complex strains in each country. From this data, in most countries, we found that molecular epidemiology has not been a tool used for the control of TB, suggesting that the Beijing genotype may be underestimated in Latin America. It is recommended that countries with the highest incidence of the Beijing genotype use effective control strategies and increased care, as a requirement for public health systems.
Collapse
Affiliation(s)
- MI Cerezo-Cortés
- Grupo MICOBAC-UN, Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - JG Rodríguez-Castillo
- Grupo MICOBAC-UN, Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - R Hernández-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition, México D.F., Mexico
| | - MI Murcia
- Grupo MICOBAC-UN, Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
46
|
Palmer MV, Thacker TC, Rabideau MM, Jones GJ, Kanipe C, Vordermeier HM, Ray Waters W. Biomarkers of cell-mediated immunity to bovine tuberculosis. Vet Immunol Immunopathol 2019; 220:109988. [PMID: 31846797 DOI: 10.1016/j.vetimm.2019.109988] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/15/2019] [Accepted: 11/28/2019] [Indexed: 10/25/2022]
Abstract
Whole blood based assays, particularly interferon gamma (IFN-γ) release assays (IGRAs), are used for the diagnosis of both bovine and human tuberculosis (TB). The aim of the current study was to evaluate a panel of cytokines and chemokines for potential use as diagnostic readouts indicative of Mycobacterium bovis (M. bovis) infection in cattle. A gene expression assay was used to determine the kinetics of the response to M. bovis purified protein derivative and a fusion protein consisting of ESAT-6, CFP10, and Rv3615c upon aerosol infection with ∼104 cfu of M. bovis. The panel of biomarkers included: IFN-γ, CXCL9, CXCL10, CCL2, CCL3, TNF-α, IL-1α, IL-1β, IL-1Ra, IL-22, IL-21 and IL-13. Protein levels of IFN-γ, CXCL9, and CXCL10 were determined by ELISA. Findings suggest that CXCL9, CXCL10, IL-21, IL-13, and several acute phase cytokines may be worth pursuing as diagnostic biomarkers of M. bovis infection in cattle.
Collapse
Affiliation(s)
- Mitchell V Palmer
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA.
| | - Tyler C Thacker
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Meaghan M Rabideau
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Gareth J Jones
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - Carly Kanipe
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA; Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - H Martin Vordermeier
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| |
Collapse
|
47
|
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]
|
48
|
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: 31] [Impact Index Per Article: 5.2] [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
|
49
|
Mashabela GT, de Wet TJ, Warner DF. Mycobacterium tuberculosis Metabolism. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0067-2019. [PMID: 31350832 PMCID: PMC10957194 DOI: 10.1128/microbiolspec.gpp3-0067-2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis is the cause of tuberculosis (TB), a disease which continues to overwhelm health systems in endemic regions despite the existence of effective combination chemotherapy and the widespread use of a neonatal anti-TB vaccine. For a professional pathogen, M. tuberculosis retains a surprisingly large proportion of the metabolic repertoire found in nonpathogenic mycobacteria with very different lifestyles. Moreover, evidence that additional functions were acquired during the early evolution of the M. tuberculosis complex suggests the organism has adapted (and augmented) the metabolic pathways of its environmental ancestor to persistence and propagation within its obligate human host. A better understanding of M. tuberculosis pathogenicity, however, requires the elucidation of metabolic functions under disease-relevant conditions, a challenge complicated by limited knowledge of the microenvironments occupied and nutrients accessed by bacilli during host infection, as well as the reliance in experimental mycobacteriology on a restricted number of experimental models with variable relevance to clinical disease. Here, we consider M. tuberculosis metabolism within the framework of an intimate host-pathogen coevolution. Focusing on recent advances in our understanding of mycobacterial metabolic function, we highlight unusual adaptations or departures from the better-characterized model intracellular pathogens. We also discuss the impact of these mycobacterial "innovations" on the susceptibility of M. tuberculosis to existing and experimental anti-TB drugs, as well as strategies for targeting metabolic pathways. Finally, we offer some perspectives on the key gaps in the current knowledge of fundamental mycobacterial metabolism and the lessons which might be learned from other systems.
Collapse
Affiliation(s)
- Gabriel T Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Current address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch, South Africa
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, South Africa
| |
Collapse
|
50
|
Chiner-Oms Á, Sánchez-Busó L, Corander J, Gagneux S, Harris SR, Young D, González-Candelas F, Comas I. Genomic determinants of speciation and spread of the Mycobacterium tuberculosis complex. SCIENCE ADVANCES 2019; 5:eaaw3307. [PMID: 31448322 PMCID: PMC6691555 DOI: 10.1126/sciadv.aaw3307] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/10/2019] [Indexed: 06/10/2023]
Abstract
Models on how bacterial lineages differentiate increase our understanding of early bacterial speciation events and the genetic loci involved. Here, we analyze the population genomics events leading to the emergence of the tuberculosis pathogen. The emergence is characterized by a combination of recombination events involving core pathogenesis functions and purifying selection on early diverging loci. We identify the phoR gene, the sensor kinase of a two-component system involved in virulence, as a key functional player subject to pervasive positive selection after the divergence of the Mycobacterium tuberculosis complex from its ancestor. Previous evidence showed that phoR mutations played a central role in the adaptation of the pathogen to different host species. Now, we show that phoR mutations have been under selection during the early spread of human tuberculosis, during later expansions, and in ongoing transmission events. Our results show that linking pathogen evolution across evolutionary and epidemiological time scales points to past and present virulence determinants.
Collapse
Affiliation(s)
- Á. Chiner-Oms
- Unidad Mixta “Infección y Salud Pública” FISABIO-CSISP/Universidad de Valencia, Instituto de Biología Integrativa de Sistemas (ISysBio), Valencia, Spain
| | - L. Sánchez-Busó
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - J. Corander
- Pathogen Genomics, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
- Department of Biostatistics, University of Oslo, 0317 Oslo, Norway
- Helsinki Institute of Information Technology (HIIT), Department of Mathematics and Statistics, University of Helsinki, 00014 Helsinki, Finland
| | - S. Gagneux
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - S. R. Harris
- Microbiotica, BioData Innovation Centre, Wellcome Genome Campus, Cambridge CB10 1DR, UK
| | - D. Young
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - F. González-Candelas
- Unidad Mixta “Infección y Salud Pública” FISABIO-CSISP/Universidad de Valencia, Instituto de Biología Integrativa de Sistemas (ISysBio), Valencia, Spain
- CIBER en Epidemiología y Salud Pública, Valencia, Spain
| | - I. Comas
- CIBER en Epidemiología y Salud Pública, Valencia, Spain
- Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| |
Collapse
|