Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nature ; 593(7859): 445-448, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33981042

RESUMO

Mycobacterium tuberculosis is the cause of one of the most important infectious diseases in humans, which leads to 1.4 million deaths every year1. Specialized protein transport systems-known as type VII secretion systems (T7SSs)-are central to the virulence of this pathogen, and are also crucial for nutrient and metabolite transport across the mycobacterial cell envelope2,3. Here we present the structure of an intact T7SS inner-membrane complex of M. tuberculosis. We show how the 2.32-MDa ESX-5 assembly, which contains 165 transmembrane helices, is restructured and stabilized as a trimer of dimers by the MycP5 protease. A trimer of MycP5 caps a central periplasmic dome-like chamber that is formed by three EccB5 dimers, with the proteolytic sites of MycP5 facing towards the cavity. This chamber suggests a central secretion and processing conduit. Complexes without MycP5 show disruption of the EccB5 periplasmic assembly and increased flexibility, which highlights the importance of MycP5 for complex integrity. Beneath the EccB5-MycP5 chamber, dimers of the EccC5 ATPase assemble into three bundles of four transmembrane helices each, which together seal the potential central secretion channel. Individual cytoplasmic EccC5 domains adopt two distinctive conformations that probably reflect different secretion states. Our work suggests a previously undescribed mechanism of protein transport and provides a structural scaffold to aid in the development of drugs against this major human pathogen.


Assuntos
Microscopia Crioeletrônica , Mycobacterium tuberculosis , Sistemas de Secreção Tipo VII/metabolismo , Sistemas de Secreção Tipo VII/ultraestrutura , Citosol/química , Citosol/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/ultraestrutura , Periplasma/química , Periplasma/metabolismo , Domínios Proteicos , Multimerização Proteica , Estabilidade Proteica , Tuberculose/virologia , Sistemas de Secreção Tipo VII/química
2.
J Biol Chem ; 295(18): 5960-5969, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32184351

RESUMO

Bacterial type VII secretion systems secrete a wide range of extracellular proteins that play important roles in bacterial viability and in interactions of pathogenic mycobacteria with their hosts. Mycobacterial type VII secretion systems consist of five subtypes, ESX-1-5, and have four substrate classes, namely, Esx, PE, PPE, and Esp proteins. At least some of these substrates are secreted as heterodimers. Each ESX system mediates the secretion of a specific set of Esx, PE, and PPE proteins, raising the question of how these substrates are recognized in a system-specific fashion. For the PE/PPE heterodimers, it has been shown that they interact with their cognate EspG chaperone and that this chaperone determines the designated secretion pathway. However, both structural and pulldown analyses have suggested that EspG cannot interact with the Esx proteins. Therefore, the determining factor for system specificity of the Esx proteins remains unknown. Here, we investigated the secretion specificity of the ESX-1 substrate pair EsxB_1/EsxA_1 in Mycobacterium marinum Although this substrate pair was hardly secreted when homologously expressed, it was secreted when co-expressed together with the PE35/PPE68_1 pair, indicating that this pair could stimulate secretion of the EsxB_1/EsxA_1 pair. Surprisingly, co-expression of EsxB_1/EsxA_1 with a modified PE35/PPE68_1 version that carried the EspG5 chaperone-binding domain, previously shown to redirect this substrate pair to the ESX-5 system, also resulted in redirection and co-secretion of the Esx pair via ESX-5. Our results suggest a secretion model in which PE35/PPE68_1 determines the system-specific secretion of EsxB_1/EsxA_1.


Assuntos
Ácido Glutâmico/química , Mycobacterium marinum/metabolismo , Prolina/química , Sistemas de Secreção Tipo VII/metabolismo , Regulação Bacteriana da Expressão Gênica , Mycobacterium marinum/genética , Domínios Proteicos , Sistemas de Secreção Tipo VII/química
3.
Mol Microbiol ; 114(1): 66-76, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32096294

RESUMO

Mycobacteria use type VII secretion systems (T7SSs) to translocate a wide range of proteins across their diderm cell envelope. These systems, also called ESX systems, are crucial for the viability and/or virulence of mycobacterial pathogens, including Mycobacterium tuberculosis and the fish pathogen Mycobacterium marinum. We have previously shown that the M. tuberculosis ESX-5 system is unable to fully complement secretion in an M. marinum esx-5 mutant, suggesting species specificity in secretion. In this study, we elaborated on this observation and established that the membrane ATPase EccC5 , possessing four (putative) nucleotide-binding domains (NBDs), is responsible for this. By creating M. marinum-M. tuberculosis EccC5 chimeras, we observed both in M. marinum and in M. tuberculosis that secretion specificity of PE_PGRS proteins depends on the presence of the cognate linker 2 domain of EccC5 . This region connects NBD1 and NBD2 of EccC5 and is responsible for keeping NBD1 in an inhibited state. Notably, the ESX-5 substrate EsxN, predicted to bind to NBD3 on EccC5 , showed a distinct secretion profile. These results indicate that linker 2 is involved in species-specific substrate recognition and might therefore be an additional substrate recognition site of EccC5 .


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Bactérias/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/metabolismo , Sistemas de Secreção Tipo VII/genética , Fatores de Virulência/metabolismo , Membrana Celular/metabolismo , Quimera/genética , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Domínios Proteicos/genética , Especificidade da Espécie , Virulência/genética
4.
J Biol Chem ; 294(13): 4806-4814, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30692196

RESUMO

Mycobacteria use type VII secretion systems to secrete proteins across their highly hydrophobic diderm cell envelope. Pathogenic mycobacteria, such as Mycobacterium tuberculosis and Mycobacterium marinum, have up to five of these systems, named ESX-1 to ESX-5. Most of these systems contain a set of five conserved membrane components, of which the four Ecc proteins form the core membrane-embedded secretion complex. The fifth conserved membrane protein, mycosin protease (MycP), is not part of the core complex but is essential for secretion, as it stabilizes this membrane complex. Here we investigated which MycP domains are required for this stabilization by producing hybrid constructs between MycP1 and MycP5 in M. marinum and analyzed their effect on ESX-1 and ESX-5 secretion. We found that both the protease and transmembrane domain are required for the ESX system-specific function of mycosins. In addition, we observed that the transmembrane domain strongly affects MycP protein levels. We also show that the extended loops 1 and 2 in the protease domain are probably primarily involved in MycP stability, whereas loop 3 and the MycP5-specific loop 5 are dispensable. The atypical propeptide, or N-terminal extension, is required only for MycP stability. Finally, we show that the protease domain of MycPP1, encoded by the esx-P1 locus on the pRAW plasmid, is functionally redundant to the protease domain of MycP5 These results provide the first insight into the regions of mycosins involved in interaction with and stabilization of their respective ESX complexes.


Assuntos
Proteínas de Bactérias , Mycobacterium marinum , Mycobacterium tuberculosis , Subtilisinas , Sistemas de Secreção Tipo IV , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Mycobacterium marinum/enzimologia , Mycobacterium marinum/genética , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , Domínios Proteicos , Estrutura Secundária de Proteína , Subtilisinas/química , Subtilisinas/genética , Subtilisinas/metabolismo , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo
5.
PLoS Pathog ; 14(6): e1007139, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29912964

RESUMO

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.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/imunologia , Proteínas de Membrana/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/imunologia , Animais , Proteínas de Bactérias/genética , Feminino , Genoma Bacteriano , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Família Multigênica , Tuberculose/prevenção & controle , Virulência
6.
PLoS Pathog ; 14(8): e1007247, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30102741

RESUMO

The pathogen Mycobacterium tuberculosis employs a range of ESX-1 substrates to manipulate the host and build a successful infection. Although the importance of ESX-1 secretion in virulence is well established, the characterization of its individual components and the role of individual substrates is far from complete. Here, we describe the functional characterization of the Mycobacterium marinum accessory ESX-1 proteins EccA1, EspG1 and EspH, i.e. proteins that are neither substrates nor structural components. Proteomic analysis revealed that EspG1 is crucial for ESX-1 secretion, since all detectable ESX-1 substrates were absent from the cell surface and culture supernatant in an espG1 mutant. Deletion of eccA1 resulted in minor secretion defects, but interestingly, the severity of these secretion defects was dependent on the culture conditions. Finally, espH deletion showed a partial secretion defect; whereas several ESX-1 substrates were secreted in normal amounts, secretion of EsxA and EsxB was diminished and secretion of EspE and EspF was fully blocked. Interaction studies showed that EspH binds EspE and therefore could function as a specific chaperone for this substrate. Despite the observed differences in secretion, hemolytic activity was lost in all M. marinum mutants, implying that hemolytic activity is not strictly correlated with EsxA secretion. Surprisingly, while EspH is essential for successful infection of phagocytic host cells, deletion of espH resulted in a significantly increased virulence phenotype in zebrafish larvae, linked to poor granuloma formation and extracellular outgrowth. Together, these data show that different sets of ESX-1 substrates play different roles at various steps of the infection cycle of M. marinum.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidade , Sistemas de Secreção Tipo VII/genética , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/genética , Células Cultivadas , Embrião não Mamífero , Larva , Camundongos , Mycobacterium marinum/genética , Células RAW 264.7 , Ovinos , Sistemas de Secreção Tipo VII/metabolismo , Virulência/genética , Fatores de Virulência/genética , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento
7.
PLoS Genet ; 13(12): e1007131, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29281637

RESUMO

The cell envelope of mycobacteria is a highly unique and complex structure that is functionally equivalent to that of Gram-negative bacteria to protect the bacterial cell. Defects in the integrity or assembly of this cell envelope must be sensed to allow the induction of stress response systems. The promoter that is specifically and most strongly induced upon exposure to ethambutol and isoniazid, first line drugs that affect cell envelope biogenesis, is the iniBAC promoter. In this study, we set out to identify the regulator of the iniBAC operon in Mycobacterium marinum using an unbiased transposon mutagenesis screen in a constitutively iniBAC-expressing mutant background. We obtained multiple mutants in the mce1 locus as well as mutants in an uncharacterized putative transcriptional regulator (MMAR_0612). This latter gene was shown to function as the iniBAC regulator, as overexpression resulted in constitutive iniBAC induction, whereas a knockout mutant was unable to respond to the presence of ethambutol and isoniazid. Experiments with the M. tuberculosis homologue (Rv0339c) showed identical results. RNAseq experiments showed that this regulatory gene was exclusively involved in the regulation of the iniBAC operon. We therefore propose to name this dedicated regulator iniBAC Regulator (IniR). IniR belongs to the family of signal transduction ATPases with numerous domains, including a putative sugar-binding domain. Upon testing different sugars, we identified trehalose as an activator and metabolic cue for iniBAC activation, which could also explain the effect of the mce1 mutations. In conclusion, cell envelope stress in mycobacteria is regulated by IniR in a cascade that includes trehalose.


Assuntos
Adenosina Trifosfatases/genética , Mycobacterium marinum/genética , Mycobacterium marinum/metabolismo , Trealose/metabolismo , Proteínas de Bactérias/genética , Membrana Celular/metabolismo , Parede Celular/genética , Parede Celular/metabolismo , Elementos de DNA Transponíveis , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Mutagênese Insercional , Óperon , Regiões Promotoras Genéticas , Transdução de Sinais , Transcrição Gênica
8.
PLoS Genet ; 11(5): e1005190, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25938982

RESUMO

Mycobacteria possess different type VII secretion (T7S) systems to secrete proteins across their unusual cell envelope. One of these systems, ESX-5, is only present in slow-growing mycobacteria and responsible for the secretion of multiple substrates. However, the role of ESX-5 substrates in growth and/or virulence is largely unknown. In this study, we show that esx-5 is essential for growth of both Mycobacterium marinum and Mycobacterium bovis. Remarkably, this essentiality can be rescued by increasing the permeability of the outer membrane, either by altering its lipid composition or by the introduction of the heterologous porin MspA. Mutagenesis of the first nucleotide-binding domain of the membrane ATPase EccC5 prevented both ESX-5-dependent secretion and bacterial growth, but did not affect ESX-5 complex assembly. This suggests that the rescuing effect is not due to pores formed by the ESX-5 membrane complex, but caused by ESX-5 activity. Subsequent proteomic analysis to identify crucial ESX-5 substrates confirmed that all detectable PE and PPE proteins in the cell surface and cell envelope fractions were routed through ESX-5. Additionally, saturated transposon-directed insertion-site sequencing (TraDIS) was applied to both wild-type M. marinum cells and cells expressing mspA to identify genes that are not essential anymore in the presence of MspA. This analysis confirmed the importance of esx-5, but we could not identify essential ESX-5 substrates, indicating that multiple of these substrates are together responsible for the essentiality. Finally, examination of phenotypes on defined carbon sources revealed that an esx-5 mutant is strongly impaired in the uptake and utilization of hydrophobic carbon sources. Based on these data, we propose a model in which the ESX-5 system is responsible for the transport of cell envelope proteins that are required for nutrient uptake. These proteins might in this way compensate for the lack of MspA-like porins in slow-growing mycobacteria.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Mycobacterium bovis/genética , Mycobacterium marinum/genética , Sistemas de Secreção Tipo VII/metabolismo , Ampicilina/farmacologia , Proteínas de Bactérias/genética , Permeabilidade da Membrana Celular , Cromatografia Líquida , Elementos de DNA Transponíveis , DNA Bacteriano/genética , Farmacorresistência Bacteriana , Mutação , Mycobacterium bovis/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Análise de Sequência de DNA , Espectrometria de Massas em Tandem , Sistemas de Secreção Tipo VII/genética
9.
Infect Immun ; 83(5): 1778-88, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25690095

RESUMO

The interaction of environmental bacteria with unicellular eukaryotes is generally considered a major driving force for the evolution of intracellular pathogens, allowing them to survive and replicate in phagocytic cells of vertebrate hosts. To test this hypothesis on a genome-wide level, we determined for the intracellular pathogen Mycobacterium marinum whether it uses conserved strategies to exploit host cells from both protozoan and vertebrate origin. Using transposon-directed insertion site sequencing (TraDIS), we determined differences in genetic requirements for survival and replication in phagocytic cells of organisms from different kingdoms. In line with the general hypothesis, we identified a number of general virulence mechanisms, including the type VII protein secretion system ESX-1, biosynthesis of polyketide lipids, and utilization of sterols. However, we were also able to show that M. marinum contains an even larger set of host-specific virulence determinants, including proteins involved in the modification of surface glycolipids and, surprisingly, the auxiliary proteins of the ESX-1 system. Several of these factors were in fact counterproductive in other hosts. Therefore, M. marinum contains different sets of virulence factors that are tailored for specific hosts. Our data imply that although amoebae could function as a training ground for intracellular pathogens, they do not fully prepare pathogens for crossing species barriers.


Assuntos
Genoma Bacteriano , Viabilidade Microbiana , Mutagênese Insercional , Mycobacterium marinum/genética , Mycobacterium marinum/fisiologia , Fatores de Virulência/metabolismo , Acanthamoeba castellanii/microbiologia , Animais , Elementos de DNA Transponíveis , Dictyostelium/microbiologia , Humanos , Mycobacterium marinum/crescimento & desenvolvimento , Fagócitos/microbiologia , Virulência , Fatores de Virulência/genética
10.
Mol Microbiol ; 94(2): 367-82, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25155747

RESUMO

The growth or virulence of Mycobacterium tuberculosis bacilli depends on homologous type VII secretion systems, ESX-1, ESX-3 and ESX-5, which export a number of protein effectors across membranes to the bacterial surface and environment. PE and PPE proteins represent two large families of highly polymorphic proteins that are secreted by these ESX systems. Recently, it was shown that these proteins require system-specific cytoplasmic chaperones for secretion. Here, we report the crystal structure of M. tuberculosis ESX-5-secreted PE25-PPE41 heterodimer in complex with the cytoplasmic chaperone EspG(5). EspG(5) represents a novel fold that is unrelated to previously characterized secretion chaperones. Functional analysis of the EspG(5) -binding region uncovered a hydrophobic patch on PPE41 that promotes dimer aggregation, and the chaperone effectively abolishes this process. We show that PPE41 contains a characteristic chaperone-binding sequence, the hh motif, which is highly conserved among ESX-1-, ESX-3- and ESX-5-specific PPE proteins. Disrupting the interaction between EspG(5) and three different PPE target proteins by introducing different point mutations generally affected protein secretion. We further demonstrate that the EspG(5) chaperone plays an important role in the ESX secretion mechanism by keeping aggregation-prone PE-PPE proteins in their soluble state.


Assuntos
Proteínas de Bactérias/química , Sistemas de Secreção Bacterianos , Chaperonas Moleculares/química , Mycobacterium tuberculosis/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Análise Mutacional de DNA , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Mycobacterium tuberculosis/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA