Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 22(4): e3002259, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683873

ABSTRACT

Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.


Subject(s)
Antitubercular Agents , Disease Models, Animal , Macrophages , Mycobacterium tuberculosis , Oxadiazoles , Tuberculosis , Zinc , Animals , Oxadiazoles/pharmacology , Humans , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Mycobacterium tuberculosis/drug effects , Zinc/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Tuberculosis/drug therapy , Mice, Inbred C57BL , Female , Drug Synergism
2.
PLoS Pathog ; 19(7): e1011437, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37450466

ABSTRACT

The molecular factors and genetic adaptations that contributed to the emergence of Mycobacterium tuberculosis (MTB) from an environmental Mycobacterium canettii-like ancestor, remain poorly investigated. In MTB, the PhoPR two-component regulatory system controls production and secretion of proteins and lipid virulence effectors. Here, we describe that several mutations, present in phoR of M. canettii relative to MTB, impact the expression of the PhoP regulon and the pathogenicity of the strains. First, we establish a molecular model of PhoR and show that some substitutions found in PhoR of M. canettii are likely to impact the structure and activity of this protein. Second, we show that STB-K, the most attenuated available M. canettii strain, displays lower expression of PhoP-induced genes than MTB. Third, we demonstrate that genetic swapping of the phoPR allele from STB-K with the ortholog from MTB H37Rv enhances expression of PhoP-controlled functions and the capacities of the recombinant strain to colonize human macrophages, the MTB target cells, as well as to cause disease in several mouse infection models. Fourth, we extended these observations to other M. canettii strains and confirm that PhoP-controlled functions are expressed at lower levels in most M. canettii strains than in M. tuberculosis. Our findings suggest that distinct PhoR variants have been selected during the evolution of tuberculosis bacilli, contributing to higher pathogenicity and persistence of MTB in the mammalian host.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , Humans , Virulence/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutation , Tuberculosis/microbiology , Mammals
3.
Proc Natl Acad Sci U S A ; 119(11): e2122161119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35271388

ABSTRACT

SignificanceTuberculosis (TB), an ancient disease of humanity, continues to be a major cause of worldwide death. The causative agent of TB, Mycobacterium tuberculosis, and its close pathogenic relative Mycobacterium marinum, initially infect, evade, and exploit macrophages, a major host defense against invading pathogens. Within macrophages, mycobacteria reside within host membrane-bound compartments called phagosomes. Mycobacterium-induced damage of the phagosomal membranes is integral to pathogenesis, and this activity has been attributed to the specialized mycobacterial secretion system ESX-1, and particularly to ESAT-6, its major secreted protein. Here, we show that the integrity of the unstructured ESAT-6 C terminus is required for macrophage phagosomal damage, granuloma formation, and virulence.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Mycobacterium marinum , Mycobacterium tuberculosis , Phagosomes , Tuberculoma , Type VII Secretion Systems , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mycobacterium marinum/metabolism , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Phagosomes/metabolism , Phagosomes/microbiology , Protein Conformation , Tuberculoma/microbiology , Type VII Secretion Systems/metabolism , Virulence
4.
Proc Natl Acad Sci U S A ; 117(34): 20848-20859, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32778586

ABSTRACT

Tuberculosis (TB) claims 1.5 million lives per year. This situation is largely due to the low efficacy of the only licensed TB vaccine, Bacillus Calmette-Guérin (BCG) against pulmonary TB. The metabolic disease type 2 diabetes (T2D) is a risk factor for TB and the mechanisms underlying increased TB susceptibility in T2D are not well understood. Furthermore, it is unknown if new TB vaccines will provide protection in the context of T2D. Here we used a diet-induced murine model of T2D to investigate the underlying mechanisms of TB/T2D comorbidity and to evaluate the protective capacity of two experimental TB vaccines in comparison to conventional BCG. Our data reveal a distinct immune dysfunction that is associated with diminished recognition of mycobacterial antigens in T2D. More importantly, we provide compelling evidence that mucosal delivery of recombinant BCG strains expressing the Mycobacterium tuberculosis (Mtb) ESX-1 secretion system (BCG::RD1 and BCG::RD1 ESAT-6 ∆92-95) are safe and confer superior immunity against aerosol Mtb infection in the context of T2D. Our findings suggest that the remarkable anti-TB immunity by these recombinant BCG strains is achieved via augmenting the numbers and functional capacity of antigen presenting cells in the lungs of diabetic mice.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Proteins/pharmacology , Diabetes Mellitus, Type 2/metabolism , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , BCG Vaccine , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Diabetes Mellitus, Experimental , Disease Models, Animal , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis Vaccines/immunology , Vaccination
5.
Cell Microbiol ; 23(7): e13344, 2021 07.
Article in English | MEDLINE | ID: mdl-33860624

ABSTRACT

The interactions between microbes and their hosts are among the most complex biological phenomena known today. The interaction may reach from overall beneficial interaction, as observed for most microbiome/microbiota related interactions to interaction with virulent pathogens, against which host cells have evolved sophisticated defence strategies. Among the latter, the confinement of invading pathogens in a phagosome plays a key role, which often results in the destruction of the invader, whereas some pathogens may counteract phagosomal arrest and survive by gaining access to the cytosol of the host cell. In the current review, we will discuss recent insights into this dynamic process of host-pathogen interaction, using Mycobacterium tuberculosis and related pathogenic mycobacteria as main examples.


Subject(s)
Host-Pathogen Interactions , Mycobacterium tuberculosis/pathogenicity , Phagosomes/microbiology , Tuberculosis/microbiology , Humans , Phagocytosis
6.
PLoS Pathog ; 15(11): e1008069, 2019 11.
Article in English | MEDLINE | ID: mdl-31703112

ABSTRACT

Free-living amoebae are thought to represent an environmental niche in which amoeba-resistant bacteria may evolve towards pathogenicity. To get more insights into factors playing a role for adaptation to intracellular life, we characterized the transcriptomic activities of the emerging pathogen Mycobacterium abscessus in amoeba and murine macrophages (Mϕ) and compared them with the intra-amoebal transcriptome of the closely related, but less pathogenic Mycobacterium chelonae. Data on up-regulated genes in amoeba point to proteins that allow M. abscessus to resist environmental stress and induce defense mechanisms, as well as showing a switch from carbohydrate carbon sources to fatty acid metabolism. For eleven of the most upregulated genes in amoeba and/or Mϕ, we generated individual gene knock-out M. abscessus mutant strains, from which ten were found to be attenuated in amoeba and/or Mϕ in subsequence virulence analyses. Moreover, transfer of two of these genes into the genome of M. chelonae increased the intra-Mϕ survival of the recombinant strain. One knock-out mutant that had the gene encoding Eis N-acetyl transferase protein (MAB_4532c) deleted, was particularly strongly attenuated in Mϕ. Taken together, M. abscessus intra-amoeba and intra-Mϕ transcriptomes revealed the capacity of M. abscessus to adapt to an intracellular lifestyle, with amoeba largely contributing to the enhancement of M. abscessus intra-Mϕ survival.


Subject(s)
Amoeba/genetics , Macrophages/metabolism , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium abscessus/pathogenicity , Transcriptome , Virulence Factors/genetics , Virulence/genetics , Amoeba/growth & development , Amoeba/microbiology , Animals , Bacterial Proteins/genetics , Macrophages/microbiology , Mice , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/genetics , Mycobacterium abscessus/isolation & purification
7.
Proc Natl Acad Sci U S A ; 115(5): E1002-E1011, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29343644

ABSTRACT

Mycobacterium abscessus, a rapidly growing mycobacterium (RGM) and an opportunistic human pathogen, is responsible for a wide spectrum of clinical manifestations ranging from pulmonary to skin and soft tissue infections. This intracellular organism can resist the bactericidal defense mechanisms of amoebae and macrophages, an ability that has not been observed in other RGM. M. abscessus can up-regulate several virulence factors during transient infection of amoebae, thereby becoming more virulent in subsequent respiratory infections in mice. Here, we sought to identify the M. abscessus genes required for replication within amoebae. To this end, we constructed and screened a transposon (Tn) insertion library of an M. abscessus subspecies massiliense clinical isolate for attenuated clones. This approach identified five genes within the ESX-4 locus, which in M. abscessus encodes an ESX-4 type VII secretion system that exceptionally also includes the ESX conserved EccE component. To confirm the screening results and to get further insight into the contribution of ESX-4 to M. abscessus growth and survival in amoebae and macrophages, we generated a deletion mutant of eccB4 that encodes a core structural element of ESX-4. This mutant was less efficient at blocking phagosomal acidification than its parental strain. Importantly, and in contrast to the wild-type strain, it also failed to damage phagosomes and showed reduced signs of phagosome-to-cytosol contact, as demonstrated by a combination of cellular and immunological assays. This study attributes an unexpected and genuine biological role to the underexplored mycobacterial ESX-4 system and its substrates.


Subject(s)
Amoeba/microbiology , Mycobacterium abscessus/pathogenicity , Phagosomes/microbiology , Type IV Secretion Systems/genetics , Virulence Factors/genetics , Bacterial Proteins/genetics , Caspase 1/metabolism , Chromatography, Thin Layer , Cytosol/metabolism , Enzyme Activation , Flow Cytometry , Galectin 3/metabolism , Gene Deletion , Genomics , Humans , Lipids/chemistry , Macrophages/microbiology , Mutation , Mycobacterium abscessus/genetics , Mycobacterium tuberculosis/pathogenicity , THP-1 Cells , Virulence
8.
PLoS Pathog ; 14(6): e1007139, 2018 06.
Article in English | MEDLINE | ID: mdl-29912964

ABSTRACT

Tuberculosis is the deadliest infectious disease worldwide. Although the BCG vaccine is widely used, it does not efficiently protect against pulmonary tuberculosis and an improved tuberculosis vaccine is therefore urgently needed. Mycobacterium tuberculosis uses different ESX/Type VII secretion (T7S) systems to transport proteins important for virulence and host immune responses. We recently reported that secretion of T7S substrates belonging to the mycobacteria-specific Pro-Glu (PE) and Pro-Pro-Glu (PPE) proteins of the PGRS (polymorphic GC-rich sequences) and MPTR (major polymorphic tandem repeat) subfamilies required both a functional ESX-5 system and a functional PPE38/71 protein for secretion. Inactivation of ppe38/71 and the resulting loss of PE_PGRS/PPE-MPTR secretion were linked to increased virulence of M. tuberculosis strains. Here, we show that a predicted total of 89 PE_PGRS/PPE-MPTR surface proteins are not exported by certain animal-adapted strains of the M. tuberculosis complex including M. bovis. This Δppe38/71-associated secretion defect therefore also occurs in the M. bovis-derived tuberculosis vaccine BCG and could be partially restored by introduction of the M. tuberculosis ppe38-locus. Epitope mapping of the PPE-MPTR protein PPE10, further allowed us to monitor T-cell responses in splenocytes from BCG/M. tuberculosis immunized mice, confirming the dependence of PPE10-specific immune-induction on ESX-5/PPE38-mediated secretion. Restoration of PE_PGRS/PPE-MPTR secretion in recombinant BCG neither altered global antigenic presentation or activation of innate immune cells, nor protective efficacy in two different mouse vaccination-infection models. This unexpected finding stimulates a reassessment of the immunomodulatory properties of PE_PGRS/PPE-MPTR proteins, some of which are contained in vaccine formulations currently in clinical evaluation.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/metabolism , Bacterial Secretion Systems/immunology , Membrane Proteins/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Animals , Bacterial Proteins/genetics , Female , Genome, Bacterial , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Multigene Family , Tuberculosis/prevention & control , Virulence
9.
Proc Natl Acad Sci U S A ; 114(6): 1371-1376, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28119503

ABSTRACT

Mycobacterium tuberculosis and Mycobacterium marinum are thought to exert virulence, in part, through their ability to lyse host cell membranes. The type VII secretion system ESX-1 [6-kDa early secretory antigenic target (ESAT-6) secretion system 1] is required for both virulence and host cell membrane lysis. Both activities are attributed to the pore-forming activity of the ESX-1-secreted substrate ESAT-6 because multiple studies have reported that recombinant ESAT-6 lyses eukaryotic membranes. We too find ESX-1 of M. tuberculosis and M. marinum lyses host cell membranes. However, we find that recombinant ESAT-6 does not lyse cell membranes. The lytic activity previously attributed to ESAT-6 is due to residual detergent in the preparations. We report here that ESX-1-dependent cell membrane lysis is contact dependent and accompanied by gross membrane disruptions rather than discrete pores. ESX-1-mediated lysis is also morphologically distinct from the contact-dependent lysis of other bacterial secretion systems. Our findings suggest redirection of research to understand the mechanism of ESX-1-mediated lysis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Hemolysis , Animals , Antigens, Bacterial/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Cell Line , Cell Line, Tumor , Erythrocyte Membrane/microbiology , Erythrocytes/microbiology , Host-Pathogen Interactions , Humans , Larva/metabolism , Larva/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mycobacterium marinum/genetics , Mycobacterium marinum/metabolism , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Sheep , Virulence , Zebrafish
10.
PLoS Pathog ; 13(11): e1006752, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29176894

ABSTRACT

The unique ability of the tuberculosis (TB) bacillus, Mycobacterium tuberculosis, to persist for long periods of time in lung hypoxic lesions chiefly contributes to the global burden of latent TB. We and others previously reported that the M. tuberculosis ancestor underwent massive episodes of horizontal gene transfer (HGT), mostly from environmental species. Here, we sought to explore whether such ancient HGT played a part in M. tuberculosis evolution towards pathogenicity. We were interested by a HGT-acquired M. tuberculosis-specific gene set, namely moaA1-D1, which is involved in the biosynthesis of the molybdenum cofactor. Horizontal acquisition of this gene set was striking because homologues of these moa genes are present all across the Mycobacterium genus, including in M. tuberculosis. Here, we discovered that, unlike their paralogues, the moaA1-D1 genes are strongly induced under hypoxia. In vitro, a M. tuberculosis moaA1-D1-null mutant has an impaired ability to respire nitrate, to enter dormancy and to survive in oxygen-limiting conditions. Conversely, heterologous expression of moaA1-D1 in the phylogenetically closest non-TB mycobacterium, Mycobacterium kansasii, which lacks these genes, improves its capacity to respire nitrate and grants it with a marked ability to survive oxygen depletion. In vivo, the M. tuberculosis moaA1-D1-null mutant shows impaired survival in hypoxic granulomas in C3HeB/FeJ mice, but not in normoxic lesions in C57BL/6 animals. Collectively, our results identify a novel pathway required for M. tuberculosis resistance to host-imposed stress, namely hypoxia, and provide evidence that ancient HGT bolstered M. tuberculosis evolution from an environmental species towards a pervasive human-adapted pathogen.


Subject(s)
Coenzymes/biosynthesis , Gene Transfer, Horizontal , Metalloproteins/biosynthesis , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Oxygen/metabolism , Tuberculosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Gene Expression Regulation, Bacterial , Humans , Hypoxia/metabolism , Hypoxia/microbiology , Mice , Mice, Inbred C57BL , Molybdenum Cofactors , Mycobacterium/genetics , Mycobacterium/metabolism , Nitrates/metabolism , Pteridines , Tuberculosis/metabolism
11.
PLoS Pathog ; 13(5): e1006399, 2017 May.
Article in English | MEDLINE | ID: mdl-28545104

ABSTRACT

Sensing and response to changes in nutrient availability are essential for the lifestyle of environmental and pathogenic bacteria. Serine/threonine protein kinase G (PknG) is required for virulence of the human pathogen Mycobacterium tuberculosis, and its putative substrate GarA regulates the tricarboxylic acid cycle in M. tuberculosis and other Actinobacteria by protein-protein binding. We sought to understand the stimuli that lead to phosphorylation of GarA, and the roles of this regulatory system in pathogenic and non-pathogenic bacteria. We discovered that M. tuberculosis lacking garA was severely attenuated in mice and macrophages and furthermore that GarA lacking phosphorylation sites failed to restore the growth of garA deficient M. tuberculosis in macrophages. Additionally we examined the impact of genetic disruption of pknG or garA upon protein phosphorylation, nutrient utilization and the intracellular metabolome. We found that phosphorylation of GarA requires PknG and depends on nutrient availability, with glutamate and aspartate being the main stimuli. Disruption of pknG or garA caused opposing effects on metabolism: a defect in glutamate catabolism or depletion of intracellular glutamate, respectively. Strikingly, disruption of the phosphorylation sites of GarA was sufficient to recapitulate defects caused by pknG deletion. The results suggest that GarA is a cellular target of PknG and the metabolomics data demonstrate that the function of this signaling system is in metabolic regulation. This function in amino acid homeostasis is conserved amongst the Actinobacteria and provides an example of the close relationship between metabolism and virulence.


Subject(s)
Amino Acids/metabolism , Bacterial Proteins/metabolism , Metabolomics , Mycobacterium tuberculosis , Protein Serine-Threonine Kinases/metabolism , Animals , Aspartic Acid/metabolism , Bacterial Proteins/genetics , Glutamic Acid/metabolism , Homeostasis , Macrophages/microbiology , Mice , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Tuberculosis/microbiology , Virulence
12.
Proc Natl Acad Sci U S A ; 113(35): 9876-81, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27528665

ABSTRACT

Horizontal gene transfer (HGT) is a major driving force of bacterial diversification and evolution. For tuberculosis-causing mycobacteria, the impact of HGT in the emergence and distribution of dominant lineages remains a matter of debate. Here, by using fluorescence-assisted mating assays and whole genome sequencing, we present unique experimental evidence of chromosomal DNA transfer between tubercle bacilli of the early-branching Mycobacterium canettii clade. We found that the obtained recombinants had received multiple donor-derived DNA fragments in the size range of 100 bp to 118 kbp, fragments large enough to contain whole operons. Although the transfer frequency between M. canettii strains was low and no transfer could be observed among classical Mycobacterium tuberculosis complex (MTBC) strains, our study provides the proof of concept for genetic exchange in tubercle bacilli. This outstanding, now experimentally validated phenomenon presumably played a key role in the early evolution of the MTBC toward pathogenicity. Moreover, our findings also provide important information for the risk evaluation of potential transfer of drug resistance and fitness mutations among clinically relevant mycobacterial strains.


Subject(s)
DNA, Bacterial/genetics , Gene Transfer, Horizontal , Genome, Bacterial/genetics , Mycobacterium/genetics , Evolution, Molecular , Humans , Mycobacterium/classification , Mycobacterium/physiology , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/physiology , Species Specificity , Tuberculosis/microbiology , Whole Genome Sequencing/methods
13.
Immunol Rev ; 264(1): 25-45, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703550

ABSTRACT

Mycobacterium tuberculosis has evolved from a Mycobacterium canettii-like progenitor pool into one of the most successful and widespread human pathogens. The pathogenicity of M. tuberculosis is linked to its ability to secrete/export/release selected mycobacterial proteins, and it is also established that active release of mycobacterial antigens is a prerequisite for strong immune recognition. Recent research has enabled mycobacterial secretion systems and vesicle-based release of mycobacterial antigens to be elucidated, which together with host-related specificities constitute key variables that determine the outcome of infection. Here, we discuss recently discovered, novel aspects on the nature and the regulation of antigen release of the tuberculosis agent with particular emphasis on the biological characterization of mycobacteria-specific ESX/type VII secretion systems and their secreted proteins, belonging to the Esx, PE, and PPE categories. The importance of specific mycobacterial antigen release is probably best exemplified by the striking differences observed between the cellular events during infection with the ESX-1-deficient, attenuated Mycobacterium bovis BCG compared to the virulent M. tuberculosis, which are clearly important for design of more specific diagnostics and more efficient vaccines.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology , Adenosine Triphosphatases/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Biological Transport , Humans , Membrane Transport Proteins/metabolism , SEC Translocation Channels , SecA Proteins , Secretory Pathway , Secretory Vesicles/metabolism
14.
Mol Microbiol ; 103(1): 7-12, 2017 01.
Article in English | MEDLINE | ID: mdl-27859892

ABSTRACT

Mycobacterium tuberculosis, the etiological agent of human tuberculosis, harbours five ESAT-6/type VII secretion (ESX/T7S) systems. The first esx gene clusters were identified during the genome-sequencing project of M. tuberculosis H37Rv. Follow-up studies revealed additional genes playing important roles in ESX/T7S systems. Among the latter genes, one can find those that encode Pro-Glu (PE) and Pro-Pro-Glu (PPE) proteins as well as a gene cluster that is encoded >260 kb upstream of the esx-1 locus and encodes ESX-1 secretion-associated proteins EspA (Rv3616c), EspC (Rv3615c) and EspD (Rv3614c). The espACD cluster has been suggested to have an important function in ESX-1 secretion since EspA-EspC and EsxA-EsxB are mutually co-dependent on each other for secretion. However, the molecular mechanism of this co-dependence and interaction between the substrates remained unknown. In this issue of Molecular Microbiology, Lou and colleagues show that EspC forms high-molecular weight polymerization complexes that resemble selected components of type II, III and/or IV secretion systems of Gram-negative bacteria. Indeed, EspC-multimeric complexes form filamentous structures that could well represent a secretion needle of ESX-1 type VII secretion systems. This exciting observation opens new avenues for research to discover and characterize ESX/T7S components and elucidates the co-dependence of EsxA/B secretion with EspA/C.


Subject(s)
Type VII Secretion Systems/genetics , Type VII Secretion Systems/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Genome, Bacterial , Multigene Family , Mycobacterium tuberculosis/genetics , Protein Transport
15.
PLoS Pathog ; 12(7): e1005770, 2016 07.
Article in English | MEDLINE | ID: mdl-27467705

ABSTRACT

Mycobacterium tuberculosis (Mtb), possesses at least three type VII secretion systems, ESX-1, -3 and -5 that are actively involved in pathogenesis and host-pathogen interaction. We recently showed that an attenuated Mtb vaccine candidate (Mtb Δppe25-pe19), which lacks the characteristic ESX-5-associated pe/ppe genes, but harbors all other components of the ESX-5 system, induces CD4+ T-cell immune responses against non-esx-5-associated PE/PPE protein homologs. These T cells strongly cross-recognize the missing esx-5-associated PE/PPE proteins. Here, we characterized the fine composition of the functional cross-reactive Th1 effector subsets specific to the shared PE/PPE epitopes in mice immunized with the Mtb Δppe25-pe19 vaccine candidate. We provide evidence that the Mtb Δppe25-pe19 strain, despite its significant attenuation, is comparable to the WT Mtb strain with regard to: (i) its antigenic repertoire related to the different ESX systems, (ii) the induced Th1 effector subset composition, (iii) the differentiation status of the Th1 cells induced, and (iv) its particular features at stimulating the innate immune response. Indeed, we found significant contribution of PE/PPE-specific Th1 effector cells in the protective immunity against pulmonary Mtb infection. These results offer detailed insights into the immune mechanisms underlying the remarkable protective efficacy of the live attenuated Mtb Δppe25-pe19 vaccine candidate, as well as the specific potential of PE/PPE proteins as protective immunogens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Disease Models, Animal , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Th1 Cells
16.
Curr Top Microbiol Immunol ; 404: 235-265, 2017.
Article in English | MEDLINE | ID: mdl-26847354

ABSTRACT

Bacterial secretion systems are sophisticated molecular machines that fulfil a wide range of important functions, which reach from export/secretion of essential proteins or virulence factors to the implication in conjugation processes. In contrast to the widely distributed Sec and Twin Arginine Translocation (TAT) systems, the recently identified ESX/type VII systems show a more restricted distribution and are typical for mycobacteria and other high-GC Actinobacteria. Similarly, type VII-like secretion systems have been described in low-GC Gram-positive bacteria belonging to the phylum Firmicutes. While the most complex organization of type VII secretion systems currently known is found in slow-growing mycobacteria, which harbour up to 5 chromosomal-encoded systems (ESX-1 to ESX-5), much simpler organization is reported for type VII-like systems in Firmicutes. In this chapter, we describe common and divergent features of type VII- and type VII-like secretion pathways and also comment on their biological key roles, many of which are related to species-/genus-specific host-pathogen interactions and/or virulence mechanisms.


Subject(s)
Gram-Positive Bacteria/metabolism , Type VII Secretion Systems/physiology , Gram-Positive Bacteria/pathogenicity , Multigene Family , Virulence
17.
Cell Microbiol ; 19(7)2017 07.
Article in English | MEDLINE | ID: mdl-28095608

ABSTRACT

Although phthiocerol dimycocerosates (DIM) are major virulence factors of Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, little is known about their mechanism of action. Localized in the outer membrane of mycobacterial pathogens, DIM are predicted to interact with host cell membranes. Interaction with eukaryotic membranes is a property shared with another virulence factor of Mtb, the early secretory antigenic target EsxA (also known as ESAT-6). This small protein, which is secreted by the type VII secretion system ESX-1 (T7SS/ESX-1), is involved in phagosomal rupture and cell death induced by virulent mycobacteria inside host phagocytes. In this work, by the use of several knock-out or knock-in mutants of Mtb or Mycobacterium bovis BCG strains and different cell biological assays, we present conclusive evidence that ESX-1 and DIM act in concert to induce phagosomal membrane damage and rupture in infected macrophages, ultimately leading to host cell apoptosis. These results identify an as yet unknown function for DIM in the infection process and open up a new research field for the study of the interaction of lipid and protein virulence factors of Mtb.


Subject(s)
Antigens, Bacterial/metabolism , Apoptosis/physiology , Bacterial Proteins/metabolism , Lipids/physiology , Macrophages/metabolism , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Phagosomes/metabolism , Cell Line, Tumor , Cell Membrane/pathology , Humans , Macrophages/microbiology , Phagosomes/microbiology , THP-1 Cells , Virulence Factors
18.
Mol Microbiol ; 99(4): 794-807, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26538472

ABSTRACT

Understanding the molecular strategies used by Mycobacterium tuberculosis to invade and persist within the host is of paramount importance to tackle the tuberculosis pandemic. Comparative genomic surveys have revealed that hadC, encoding a subunit of the HadBC dehydratase, is mutated in the avirulent M. tuberculosis H37Ra strain. We show here that mutation or deletion of hadC affects the biosynthesis of oxygenated mycolic acids, substantially reducing their production level. Additionally, it causes the loss of atypical extra-long mycolic acids, demonstrating the involvement of HadBC in the late elongation steps of mycolic acid biosynthesis. These events have an impact on the morphotype, cording capacity and biofilm growth of the bacilli as well as on their sensitivity to agents such as rifampicin. Furthermore, deletion of hadC leads to a dramatic loss of virulence: an almost 4-log drop of the bacterial load in the lungs and spleens of infected immunodeficient mice. Both its unique function and importance for M. tuberculosis virulence make HadBC an attractive therapeutic target for tuberculosis drug development.


Subject(s)
Bacterial Proteins/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Mycolic Acids/chemistry , Tuberculosis/microbiology , Animals , Antitubercular Agents/pharmacology , Bacterial Load , Bacterial Proteins/metabolism , Biofilms/growth & development , Gene Deletion , Lung/microbiology , Mice , Mutation , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Spleen/microbiology , Virulence/genetics
19.
Mol Microbiol ; 99(5): 866-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585558

ABSTRACT

In mycobacteria, MmpL proteins represent key components that participate in the biosynthesis of the complex cell envelope. Whole genome analysis of a spontaneous rough morphotype variant of Mycobacterium abscessus subsp. bolletii identified a conserved tyrosine that is crucial for the function of MmpL family proteins. Isogenic smooth (S) and rough (R) variants differed by a single mutation linked to a Y842H substitution in MmpL4a. This mutation caused a deficiency in glycopeptidolipid production/transport in the R variant and a gain in the capacity to produce cords in vitro. In zebrafish, increased virulence of the M. bolletii R variant over the parental S strain was found, involving massive production of serpentine cords, abscess formation and rapid larval death. Importantly, this finding allowed us to demonstrate an essential role of Tyr842 in several different MmpL proteins, including Mycobacterium tuberculosis MmpL3. Structural homology models of MmpL4a and MmpL3 identified two additional critical residues located in the transmembrane regions TM10 and TM4 that are facing each other. We propose that these central residues are part of the proton-motive force that supplies the energy for substrate transport. Hence, we provide important insights into mechanistic/structural aspects of MmpL proteins as lipid transporters and virulence determinants in mycobacteria.


Subject(s)
Membrane Transport Proteins/metabolism , Mycobacterium/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Cell Wall/metabolism , Conserved Sequence , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mycobacterium/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Proton-Motive Force , Virulence , Virulence Factors/metabolism , Zebrafish
20.
Mol Microbiol ; 101(3): 515-29, 2016 08.
Article in English | MEDLINE | ID: mdl-27121350

ABSTRACT

The natural resistance of Mycobacterium abscessus to most commonly available antibiotics seriously limits chemotherapeutic treatment options, which is particularly challenging for cystic fibrosis patients infected with this rapid-growing mycobacterium. New drugs with novel molecular targets are urgently needed against this emerging pathogen. However, the discovery of such new chemotypes has not been appropriately performed. Here, we demonstrate the utility of a phenotypic screen for bactericidal compounds against M. abscessus using a library of compounds previously validated for activity against M. tuberculosis. We identified a new piperidinol-based molecule, PIPD1, exhibiting potent activity against clinical M. abscessus strains in vitro and in infected macrophages. Treatment of infected zebrafish with PIPD1 correlated with increased embryo survival and decreased bacterial burden. Whole genome analysis of M. abscessus strains resistant to PIPD1 identified several mutations in MAB_4508, encoding a protein homologous to MmpL3. Biochemical analyses demonstrated that while de novo mycolic acid synthesis was unaffected, PIPD1 strongly inhibited the transport of trehalose monomycolate, thereby abrogating mycolylation of arabinogalactan. Mapping the mutations conferring resistance to PIPD1 on a MAB_4508 tridimensional homology model defined a potential PIPD1-binding pocket. Our data emphasize a yet unexploited chemical structure class against M. abscessus infections with promising translational development possibilities.


Subject(s)
Antitubercular Agents/pharmacology , Mycolic Acids/metabolism , Nontuberculous Mycobacteria/drug effects , Piperidines/pharmacology , Animals , Binding Sites , Disease Models, Animal , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL