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.
Artículo en Inglés | MEDLINE | ID: mdl-33981042

RESUMEN

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.


Asunto(s)
Microscopía por Crioelectrón , Mycobacterium tuberculosis , Sistemas de Secreción Tipo VII/metabolismo , Sistemas de Secreción Tipo VII/ultraestructura , Citosol/química , Citosol/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/ultraestructura , Periplasma/química , Periplasma/metabolismo , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica , Tuberculosis/virología , Sistemas de Secreción Tipo VII/química
2.
J Biol Chem ; 295(18): 5960-5969, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32184351

RESUMEN

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.


Asunto(s)
Ácido Glutámico/química , Mycobacterium marinum/metabolismo , Prolina/química , Sistemas de Secreción Tipo VII/metabolismo , Regulación Bacteriana de la Expresión Génica , Mycobacterium marinum/genética , Dominios Proteicos , Sistemas de Secreción Tipo VII/química
3.
Mol Microbiol ; 114(1): 66-76, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32096294

RESUMEN

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 .


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas Bacterianas/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/metabolismo , Sistemas de Secreción Tipo VII/genética , Factores de Virulencia/metabolismo , Membrana Celular/metabolismo , Quimera/genética , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Dominios Proteicos/genética , Especificidad de la Especie , Virulencia/genética
4.
J Biol Chem ; 294(13): 4806-4814, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30692196

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Mycobacterium marinum , Mycobacterium tuberculosis , Subtilisinas , Sistemas de Secreción Tipo IV , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mycobacterium marinum/enzimología , Mycobacterium marinum/genética , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Dominios Proteicos , Estructura Secundaria de Proteína , Subtilisinas/química , Subtilisinas/genética , Subtilisinas/metabolismo , Sistemas de Secreción Tipo IV/química , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo
5.
PLoS Pathog ; 14(6): e1007139, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29912964

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/inmunología , Proteínas de la Membrana/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Proteínas Bacterianas/genética , Femenino , Genoma Bacteriano , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Familia de Multigenes , Tuberculosis/prevención & control , Virulencia
6.
PLoS Pathog ; 14(8): e1007247, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30102741

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidad , Sistemas de Secreción Tipo VII/genética , Factores de Virulencia/fisiología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Embrión no Mamífero , Larva , Ratones , Mycobacterium marinum/genética , Células RAW 264.7 , Ovinos , Sistemas de Secreción Tipo VII/metabolismo , Virulencia/genética , Factores de Virulencia/genética , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo
7.
PLoS Genet ; 13(12): e1007131, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29281637

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/genética , Mycobacterium marinum/genética , Mycobacterium marinum/metabolismo , Trehalosa/metabolismo , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Pared Celular/genética , Pared Celular/metabolismo , Elementos Transponibles de ADN , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Mutagénesis Insercional , Operón , Regiones Promotoras Genéticas , Transducción de Señal , Transcripción Genética
8.
PLoS Genet ; 11(5): e1005190, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25938982

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Mycobacterium bovis/genética , Mycobacterium marinum/genética , Sistemas de Secreción Tipo VII/metabolismo , Ampicilina/farmacología , Proteínas Bacterianas/genética , Permeabilidad de la Membrana Celular , Cromatografía Liquida , Elementos Transponibles de ADN , ADN Bacteriano/genética , Farmacorresistencia Bacteriana , Mutación , Mycobacterium bovis/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Análisis de Secuencia de ADN , Espectrometría de Masas en Tándem , Sistemas de Secreción Tipo VII/genética
9.
Infect Immun ; 83(5): 1778-88, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25690095

RESUMEN

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.


Asunto(s)
Genoma Bacteriano , Viabilidad Microbiana , Mutagénesis Insercional , Mycobacterium marinum/genética , Mycobacterium marinum/fisiología , Factores de Virulencia/metabolismo , Acanthamoeba castellanii/microbiología , Animales , Elementos Transponibles de ADN , Dictyostelium/microbiología , Humanos , Mycobacterium marinum/crecimiento & desarrollo , Fagocitos/microbiología , Virulencia , Factores de Virulencia/genética
10.
Mol Microbiol ; 94(2): 367-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25155747

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Sistemas de Secreción Bacterianos , Chaperonas Moleculares/química , Mycobacterium tuberculosis/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Análisis Mutacional de ADN , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Mycobacterium tuberculosis/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Transporte de Proteínas
11.
Cell Microbiol ; 16(2): 280-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24119166

RESUMEN

The pathogenicity of mycobacteria is closely associated with their ability to export virulence factors. For this purpose, mycobacteria possess different protein secretion systems, including the accessory Sec translocation pathway, SecA2. Although this pathway is associated with intracellular survival and virulence, the SecA2-dependent effector proteins remain largely undefined. In this work, we studied a Mycobacterium marinum secA2 mutant with an impaired capacity to initiate granuloma formation in zebrafish embryos. By comparing the proteomic profile of cell envelope fractions from the secA2 mutant with wild type M. marinum, we identified putative SecA2-dependent substrates. Immunoblotting procedures confirmed SecA2-dependent membrane localization for several of these proteins, including the virulence factor protein kinase G (PknG). Interestingly, phenotypical defects of the secA2 mutant are similar to those described for ΔpknG, including phagosomal maturation. Overexpression of PknG in the secA2 mutant restored its localization to the cell envelope. Importantly, PknG-overexpression also partially restored the virulence of the secA2 mutant, as indicated by enhanced infectivity in zebrafish embryos and restored inhibition of phagosomal maturation. These results suggest that SecA2-dependent membrane localization of PknG is an important determinant for M. marinum virulence.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mycobacterium marinum/metabolismo , Factores de Virulencia/metabolismo , Animales , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Immunoblotting , Mutagénesis Insercional , Infecciones por Mycobacterium/microbiología , Mycobacterium marinum/patogenicidad , Especificidad por Sustrato , Pez Cebra
12.
Proc Natl Acad Sci U S A ; 109(28): 11342-7, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22733768

RESUMEN

Mycobacterial pathogens use specialized type VII secretion (T7S) systems to transport crucial virulence factors across their unusual cell envelope into infected host cells. These virulence factors lack classical secretion signals and the mechanism of substrate recognition is not well understood. Here we demonstrate that the model T7S substrates PE25/PPE41, which form a heterodimer, are targeted to the T7S pathway ESX-5 by a signal located in the C terminus of PE25. Site-directed mutagenesis of residues within this C terminus resulted in the identification of a highly conserved motif, i.e., YxxxD/E, which is required for secretion. This motif was also essential for the secretion of LipY, another ESX-5 substrate. Pathogenic mycobacteria have several different T7S systems and we identified a PE protein that is secreted by the ESX-1 system, which allowed us to compare substrate recognition of these two T7S systems. Surprisingly, this ESX-1 substrate contained a C-terminal signal functionally equivalent to that of PE25. Exchange of these C-terminal secretion signals between the PE proteins restored secretion, but each PE protein remained secreted via its own ESX secretion system, indicating that an additional signal(s) provides system specificity. Remarkably, the YxxxD/E motif was also present in and required for efficient secretion of the ESX-1 substrates CFP-10 and EspB. Therefore, our data show that the YxxxD/E motif is a general secretion signal that is present in all known mycobacterial T7S substrates or substrate complexes.


Asunto(s)
Antígenos Bacterianos/metabolismo , Mycobacterium marinum/genética , Mycobacterium tuberculosis/genética , Mycobacterium/metabolismo , Factores de Virulencia/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Genoma Bacteriano , Modelos Biológicos , Datos de Secuencia Molecular , Familia de Multigenes , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/metabolismo , Estructura Terciaria de Proteína , Vías Secretoras , Homología de Secuencia de Aminoácido
13.
Cell Microbiol ; 15(4): 660-74, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23121245

RESUMEN

Mannose-capped lipoarabinomannan (ManLAM) is considered an important virulence factor of Mycobacterium tuberculosis. However, while mannose caps have been reported to be responsible for various immunosuppressive activities of ManLAM observed in vitro, there is conflicting evidence about their contribution to mycobacterial virulence in vivo. Therefore, we used Mycobacterium bovis BCG and M. tuberculosis mutants that lack the mannose cap of LAM to assess the role of ManLAM in the interaction of mycobacteria with the host cells, to evaluate vaccine-induced protection and to determine its importance in M. tuberculosis virulence. Deletion of the mannose cap did not affect BCG survival and replication in macrophages, although the capless mutant induced a somewhat higher production of TNF. In dendritic cells, the capless mutant was able to induce the upregulation of co-stimulatory molecules and the only difference we detected was the secretion of slightly higher amounts of IL-10 as compared to the wild type strain. In mice, capless BCG survived equally well and induced an immune response similar to the parental strain. Furthermore, the efficacy of vaccination against a M. tuberculosis challenge in low-dose aerosol infection models in mice and guinea pigs was not affected by the absence of the mannose caps in the BCG. Finally, the lack of the mannose cap in M. tuberculosis did not affect its virulence in mice nor its interaction with macrophages in vitro. Thus, these results do not support a major role for the mannose caps of LAM in determining mycobacterial virulence and immunogenicity in vivo in experimental animal models of infection, possibly because of redundancy of function.


Asunto(s)
Interacciones Huésped-Patógeno , Lipopolisacáridos/análisis , Manosa/análisis , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Animales , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Cobayas , Macrófagos/microbiología , Ratones , Viabilidad Microbiana , Mycobacterium bovis/química , Mycobacterium bovis/genética , Mycobacterium bovis/crecimiento & desarrollo , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis Pulmonar/microbiología , Factores de Virulencia/análisis
14.
Proc Natl Acad Sci U S A ; 108(20): 8438-43, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21536872

RESUMEN

A major step in the biogenesis of newly synthesized precursor proteins in bacteria is their targeting to the Sec translocon at the inner membrane. In gram-negative bacteria, the chaperone SecB binds nonnative forms of precursors and specifically transfers them to the SecA motor component of the translocase, thus facilitating their export. The major human pathogen Mycobacterium tuberculosis is an unusual gram-positive bacterium with a well-defined outer membrane and outer membrane proteins. Assistance to precursor proteins by chaperones in this bacterium remains largely unexplored. Here we show that the product of the previously uncharacterized Rv1957 gene of M. tuberculosis can substitute for SecB functions in Escherichia coli and prevent preprotein aggregation in vitro. Interestingly, in M. tuberculosis, Rv1957 is clustered with a functional stress-responsive higB-higA toxin-antitoxin (TA) locus of unknown function. Further in vivo experiments in E. coli and in Mycobacterium marinum strains that do not possess the TA-chaperone locus show that the severe toxicity of the toxin was entirely inhibited when the antitoxin and the chaperone were jointly expressed. We found that Rv1957 acts directly on the antitoxin by preventing its aggregation and protecting it from degradation. Taken together, our results show that the SecB-like chaperone Rv1957 specifically controls a stress-responsive TA system relevant for M. tuberculosis adaptive response.


Asunto(s)
Proteínas Bacterianas/fisiología , Chaperonas Moleculares/fisiología , Mycobacterium tuberculosis/fisiología , Estrés Fisiológico , Antitoxinas , Toxinas Bacterianas , Genes Bacterianos
15.
Microbiol Spectr ; 12(6): e0316823, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38722177

RESUMEN

Vitamin B12 (B12) serves as a critical cofactor within mycobacterial metabolism. While some pathogenic strains can synthesize B12 de novo, others rely on host-acquired B12. In this investigation, we studied the transport of vitamin B12 in Mycobacterium marinum using B12-auxotrophic and B12-sensitive strains by deleting metH or metE, respectively. These two enzymes rely on B12 in different ways to function as methionine synthases. We used these strains to select mutants affecting B12 scavenging and confirmed their phenotypes during growth experiments in vitro. Our analysis of B12 uptake mechanisms revealed that membrane lipids and cell wall integrity play an essential role in cell envelope transport. Furthermore, we identified a potential transcription regulator that responds to B12. Our study demonstrates that M. marinum can take up exogenous B12 and that altering mycobacterial membrane integrity affects B12 uptake. Finally, during zebrafish infection using B12-auxotrophic and B12-sensitive strains, we found that B12 is available for virulent mycobacteria in vivo.IMPORTANCEOur study investigates how mycobacteria acquire essential vitamin B12. These microbes, including those causing tuberculosis, face challenges in nutrient uptake due to their strong outer layer. We focused on Mycobacterium marinum, similar to TB bacteria, to uncover its vitamin B12 absorption. We used modified strains unable to produce their own B12 and discovered that M. marinum can indeed absorb it from the environment, even during infections. Changes in the outer layer composition affect this process, and genes related to membrane integrity play key roles. These findings illuminate the interaction between mycobacteria and their environment, offering insights into combatting diseases like tuberculosis through innovative strategies. Our concise research underscores the pivotal role of vitamin B12 in microbial survival and its potential applications in disease control.


Asunto(s)
Membrana Externa Bacteriana , Mycobacterium marinum , Vitamina B 12 , Pez Cebra , Mycobacterium marinum/genética , Mycobacterium marinum/metabolismo , Vitamina B 12/metabolismo , Animales , Pez Cebra/microbiología , Membrana Externa Bacteriana/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Permeabilidad de la Membrana Celular , Transporte Biológico , Membrana Celular/metabolismo , Infecciones por Mycobacterium no Tuberculosas/microbiología
16.
J Bacteriol ; 195(9): 2050-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23457249

RESUMEN

A striking characteristic of mycobacteria is the presence of an unusual outer membrane which forms a thick permeability barrier and provides resistance to many antibiotics. Although specialized proteins must reside in this layer, only few mycolate outer membrane (MOM) proteins have been identified to date. Their discovery is complicated by difficulties in obtaining good separation of mycobacterial inner and outer membranes. During our efforts to identify novel mycobacterial outer membrane proteins (MOMPs), we discovered that we can enrich for MOMPs using differential solubilization of mycobacterial cell envelopes. Subsequently, these different fractions were analyzed by nano liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS). This proteomic analysis confirmed that our marker proteins for inner membrane and MOM were found in their expected fractions and revealed a few interesting candidate MOMPs. A number of these putative MOMPs were further analyzed for their expression and localization in the cell envelope. One identified MOMP, MMAR_0617 of Mycobacterium marinum, was purified and demonstrated to form a large oligomeric complex. Importantly, this protein showed a clear single-channel conductance of 0.8 ± 0.1 ns upon reconstitution into artificial planar lipid bilayers. The most surprising feature of MMAR_0617 is a long C-terminal threonine-rich domain with extensive modifications. In summary, we have identified a novel mycobacterial outer membrane porin with unusual properties.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Mycobacterium marinum/metabolismo , Porinas/metabolismo , Treonina/metabolismo , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/aislamiento & purificación , Membrana Celular/química , Membrana Celular/genética , Detergentes/química , Datos de Secuencia Molecular , Mycobacterium marinum/química , Mycobacterium marinum/genética , Porinas/química , Porinas/genética , Porinas/aislamiento & purificación , Estructura Terciaria de Proteína , Alineación de Secuencia , Treonina/análisis
17.
J Biol Chem ; 287(38): 31939-47, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22843727

RESUMEN

Mycobacteria use the dedicated type VII protein secretion systems ESX-1 and ESX-5 to secrete virulence factors across their highly hydrophobic cell envelope. The substrates of these systems include the large mycobacterial PE and PPE protein families, which are named after their characteristic Pro-Glu and Pro-Pro-Glu motifs. Pathogenic mycobacteria secrete large numbers of PE/PPE proteins via the major export pathway, ESX-5. In addition, a few PE/PPE proteins have been shown to be exported by ESX-1. It is not known how ESX-1 and ESX-5 recognize their cognate PE/PPE substrates. In this work, we investigated the function of the cytosolic protein EspG(5), which is essential for ESX-5-mediated secretion in Mycobacterium marinum, but for which the role in secretion is not known. By performing protein co-purifications, we show that EspG(5) interacts with several PPE proteins and a PE/PPE complex that is secreted by ESX-5, but not with the unrelated ESX-5 substrate EsxN or with PE/PPE proteins secreted by ESX-1. Conversely, the ESX-1 paralogue EspG(1) interacted with a PE/PPE couple secreted by ESX-1, but not with PE/PPE substrates of ESX-5. Furthermore, structural analysis of the complex formed by EspG(5) and PE/PPE indicates that these proteins interact in a 1:1:1 ratio. In conclusion, our study shows that EspG(5) and EspG(1) interact specifically with PE/PPE proteins that are secreted via their own ESX systems and suggests that EspG proteins are specific chaperones for the type VII pathway.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli/química , Chaperonas Moleculares/química , Mycobacterium marinum/enzimología , Clonación Molecular , Citosol/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Prueba de Complementación Genética , Espectrometría de Masas/métodos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Mycobacterium marinum/metabolismo , Níquel/química , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Vías Secretoras , Espectrometría de Masas en Tándem/métodos
18.
Mol Microbiol ; 86(2): 472-84, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22925462

RESUMEN

Pathogenic mycobacteria require type VII secretion (T7S) systems to transport virulence factors across their complex cell envelope. These bacteria have up to five of these systems, termed ESX-1 to ESX-5. Here, we show that ESX-5 of Mycobacterium tuberculosis mediates the secretion of EsxN, PPE and PE_PGRS proteins, indicating that ESX-5 is a major secretion pathway in this important pathogen. Using the ESX-5 system of Mycobacterium marinum and Mycobacterium bovis BCG as a model, we have purified and analysed the T7S membrane complex under native conditions. blue native-PAGE and immunoprecipitation experiments showed that the ESX-5 membrane complex of both species has a size of ~ 1500 kDa and is composed of four conserved membrane proteins, i.e. EccB(5) , EccC(5) , EccD(5) and EccE(5) . Subsequent limited proteolysis suggests that EccC(5) and EccE(5) mostly reside on the periphery of the complex. We also observed that EccC(5) and EccD(5) expression is essential for the formation of a stable membrane complex. These are the first data on a T7S membrane complex and, given the high conservation of its components, our data can likely be generalized to most mycobacterial T7S systems.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Membrana Celular/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Membrana Celular/química , Membrana Celular/genética , Mycobacterium marinum/química , Mycobacterium marinum/genética , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Estructura Terciaria de Proteína , Transporte de Proteínas
19.
J Biol Chem ; 286(21): 19024-34, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21471225

RESUMEN

The type VII secretion system ESX-5 is a major pathway for export of PE and PPE proteins in pathogenic mycobacteria. These mycobacteria-specific protein families are characterized by conserved N-terminal domains of 100 and 180 amino acids, which contain the proline-glutamic acid (PE) and proline-proline-glutamic acid (PPE) motifs after which they are named. Here we investigated secretion of the triacylglycerol lipase LipY, which in fast-growing mycobacteria contains a signal sequence, but in slow-growing species appears to have replaced the signal peptide with a PE or PPE domain. Selected LipY homologues were expressed in wild-type Mycobacterium marinum and its corresponding ESX-5 mutant, and localization of the proteins was investigated by immunoblotting and electron microscopy. Our study shows that Mycobacterium tuberculosis PE-LipY (LipY(tub)) and M. marinum PPE-LipY (LipY(mar)) are both secreted to the bacterial surface in an ESX-5-dependent fashion. After transport, the PE/PPE domains are removed by proteolytic cleavage. In contrast, Mycobacterium gilvum LipY, which has a signal sequence, is not transported to the cell surface. Furthermore, we show that LipY(tub) and LipY(mar) require their respective PE and PPE domains for ESX-5-dependent secretion. The role of the PE domain in ESX-5 secretion was confirmed in a whole cell lipase assay, in which wild-type bacteria expressing full-length LipY(tub), but not LipY(tub) lacking its PE domain, were shown to hydrolyze extracellular lipids. In conclusion, both PE and PPE domains contain a signal required for secretion of LipY by the ESX-5 system, and these domains are proteolytically removed upon translocation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/fisiología , Lipasa/metabolismo , Mycobacterium marinum/enzimología , Mycobacterium tuberculosis/enzimología , Señales de Clasificación de Proteína/fisiología , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Lipasa/genética , Estructura Terciaria de Proteína , Especificidad de la Especie
20.
Nat Microbiol ; 7(12): 2089-2100, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36329197

RESUMEN

So far, only members of the bacterial phyla Proteobacteria and Verrucomicrobia are known to grow methanotrophically under aerobic conditions. Here we report that this metabolic trait is also observed within the Actinobacteria. We enriched and cultivated a methanotrophic Mycobacterium from an extremely acidic biofilm growing on a cave wall at a gaseous chemocline interface between volcanic gases and the Earth's atmosphere. This Mycobacterium, for which we propose the name Candidatus Mycobacterium methanotrophicum, is closely related to well-known obligate pathogens such as M. tuberculosis and M. leprae. Genomic and proteomic analyses revealed that Candidatus M. methanotrophicum expresses a full suite of enzymes required for aerobic growth on methane, including a soluble methane monooxygenase that catalyses the hydroxylation of methane to methanol and enzymes involved in formaldehyde fixation via the ribulose monophosphate pathway. Growth experiments combined with stable isotope probing using 13C-labelled methane confirmed that Candidatus M. methanotrophicum can grow on methane as a sole carbon and energy source. A broader survey based on 16S metabarcoding suggests that species closely related to Candidatus M. methanotrophicum may be abundant in low-pH, high-methane environments.


Asunto(s)
Ecosistema , Mycobacterium , Proteómica , Filogenia , Metano/metabolismo , Mycobacterium/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA