Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
FASEB J ; 37(7): e23036, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37331005

RESUMO

Cholesterol is a crucial component in Mycobacterium tuberculosis virulence as it is required for phagocytosis of mycobacteria by macrophages. In addition, the tubercle bacilli can grow using cholesterol as the sole carbon source. Thus, cholesterol catabolism represents a valuable target for the development of new antitubercular drugs. However, the molecular partners of cholesterol catabolism remain elusive in mycobacteria. Here, we focused on HsaC and HsaD, enzymes involved in two consecutive steps of cholesterol ring degradation and identified putative partners, using a BirA-based proximity-dependent biotin identification (BioID) approach in Mycobacterium smegmatis. In rich medium, the fusion protein BirA-HsaD was able to fish the endogenous cognate HsaC, thus validating this approach to study protein-protein interactions and to infer metabolic channeling of cholesterol ring degradation. In chemically defined medium, both HsaC and HsaD interacted with four proteins, BkdA, BkdB, BkdC, and MSMEG_1634. BkdA, BkdB, and BkdC are enzymes that participate in the degradation of branched-chain amino acids. As cholesterol and branched-chain amino acid catabolism both generate propionyl-CoA, which is a toxic metabolite for mycobacteria, this interconnection suggests a compartmentalization to avoid dissemination of propionyl-CoA into the mycobacterial cytosol. Moreover, the BioID approach allowed us to decipher the interactome of MSMEG_1634 and MSMEG_6518, two proteins of unknown function, which are proximal to the enzymes involved in cholesterol and branched-chain amino acid catabolism. In conclusion, BioID is a powerful tool to characterize protein-protein interactions and to decipher the interconnections between different metabolic pathways, thereby facilitating the identification of new mycobacterial targets.


Assuntos
Mycobacterium smegmatis , Mycobacterium tuberculosis , Animais , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Biotina/metabolismo , Colesterol/metabolismo , Mycobacterium tuberculosis/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
2.
Environ Microbiol ; 23(6): 3212-3224, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33913567

RESUMO

Deciphering protein-protein interactions is a critical step in the identification and the understanding of biological mechanisms deployed by pathogenic bacteria. The development of in vivo technologies to characterize these interactions is still in its infancy, especially for bacteria whose subcellular organization is particularly complex, such as mycobacteria. In this work, we used the proximity-dependent biotin identification (BioID) to define the mycobacterial heparin-binding hemagglutinin (HbhA) interactome in the saprophytic bacterium Mycobacterium smegmatis. M. smegmatis is a commonly used model to study and characterize the physiology of pathogenic mycobacteria, such as Mycobacterium tuberculosis. Here, we adapted the BioID technology to in vivo protein-protein interactions studies in M. smegmatis, which presents several advantages, such as maintaining the complex organization of the mycomembrane, offering the possibility to study membrane or cell wall-associated proteins, including HbhA, in the presence of cofactors and post-translational modifications, such as the complex methylation pattern of HbhA. Using this technology, we found that HbhA is interconnected with cholesterol degradation and heme/iron pathways. These results are in line with previous studies showing the dual localization of HbhA, associated with the cell wall and intracytoplasmic lipid inclusions, and its induction under high iron growth conditions.


Assuntos
Mycobacterium smegmatis , Mycobacterium tuberculosis , Proteínas de Bactérias/genética , Biotina , Colesterol , Heme , Ferro , Lectinas
3.
EMBO Rep ; 19(1): 29-42, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141986

RESUMO

The interaction of Mycobacterium tuberculosis (Mtb) with pulmonary epithelial cells is critical for early stages of bacillus colonization and during the progression of tuberculosis. Entry of Mtb into epithelial cells has been shown to depend on F-actin polymerization, though the molecular mechanisms are still unclear. Here, we demonstrate that mycobacterial uptake into epithelial cells requires rearrangements of the actin cytoskeleton, which are regulated by ADP-ribosylation factor 1 (Arf1) and phospholipase D1 (PLD1), and is dependent on the M3 muscarinic receptor (M3R). We show that this pathway is controlled by Arf GTPase-activating protein 1 (ArfGAP1), as its silencing has an impact on actin cytoskeleton reorganization leading to uncontrolled uptake and replication of Mtb. Furthermore, we provide evidence that this pathway is critical for mycobacterial entry, while the cellular infection with other pathogens, such as Shigella flexneri and Yersinia pseudotuberculosis, is not affected. Altogether, these results reveal how cortical actin plays the role of a barrier to prevent mycobacterial entry into epithelial cells and indicate a novel role for ArfGAP1 as a restriction factor of host-pathogen interactions.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/genética , Proteínas Ativadoras de GTPase/genética , Interações Hospedeiro-Patógeno , Mycobacterium tuberculosis/patogenicidade , Alvéolos Pulmonares/metabolismo , Células A549 , Fator 1 de Ribosilação do ADP/genética , Fator 1 de Ribosilação do ADP/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Mycobacterium tuberculosis/fisiologia , Fosfolipase D/genética , Fosfolipase D/metabolismo , Polimerização , Alvéolos Pulmonares/microbiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Shigella flexneri/fisiologia , Transdução de Sinais , Especificidade da Espécie , Yersinia pseudotuberculosis/fisiologia
4.
Cell Microbiol ; 19(1)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27220037

RESUMO

Mycobacterium tuberculosis produces several bacterial effectors impacting the colonization of phagocytes. Here, we report that the putative lipoprotein LppM hinders phagocytosis by macrophages in a toll-like receptor 2-dependent manner. Moreover, recombinant LppM is able to functionally complement the phenotype of the mutant, when exogenously added during macrophage infection. LppM is also implicated in the phagosomal maturation, as a lppM deletion mutant is more easily addressed towards the acidified compartments of the macrophage than its isogenic parental strain. In addition, this mutant was affected in its ability to induce the secretion of pro-inflammatory chemokines, interferon-gamma-inducible protein-10, monocyte chemoattractant protein-1 and macrophage inflammatory protein-1α. Thus, our results describe a new mycobacterial protein involved in the early trafficking of the tubercle bacillus and its manipulation of the host immune response.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Lipoproteínas/metabolismo , Macrófagos/microbiologia , Macrófagos/fisiologia , Mycobacterium tuberculosis/patogenicidade , Fagocitose , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Células Cultivadas , Deleção de Genes , Lipoproteínas/genética , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/genética , Fatores de Virulência/genética
5.
Int J Mol Sci ; 19(6)2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29874861

RESUMO

Heparin-binding haemagglutinin (HBHA) is a surface-exposed virulence factor of Mycobacterium tuberculosis and is involved in the binding of mycobacteria to non-phagocytic cells, allowing for extra-pulmonary dissemination of the bacilli. Despite its surface exposure, HBHA is not produced as a pre-protein containing a typical cleavable N-terminal signal peptide and is thus likely secreted by a Sec-independent, as of yet unknown mechanism. Here, we used the bacterial adenylate cyclase two-hybrid system to identify the proteins encoded by rv0613c and mmpL14 as being able to interact with HBHA. Our study was focused on Rv0613c, as it showed more consistent interactions with HBHA than MmpL14. Deletion of its orthologous gene MSMEG_1285 in recombinant Mycobacterium smegmatis producing HBHA from M. tuberculosis resulted in the loss of proper surface exposure of HBHA, as evidenced by atomic force microscopy. Furthermore, the lack of MSMEG_1285 also abolished the clumping phenotype and rough colony morphology of the recombinant M. smegmatis and reduced its adherence to A549 epithelial cells. These phenotypes have previously been associated with surface-exposed HBHA. Thus, MSMEG_1285 is directly involved in the proper cell-surface exposure of HBHA. These observations identify MSMEG_1285/Rv0613c as the first accessory protein involved in the cell surface exposure of HBHA.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Membrana/genética , Mycobacterium tuberculosis/genética , Tuberculose/genética , Células A549 , Sequência de Aminoácidos/genética , Membrana Celular/genética , Células Epiteliais/metabolismo , Humanos , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Fatores de Virulência/genética
6.
PLoS Pathog ; 10(5): e1004115, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809459

RESUMO

Mycobacterium tuberculosis bacilli display two signature features: acid-fast staining and the capacity to induce long-term latent infections in humans. However, the mechanisms governing these two important processes remain largely unknown. Ser/Thr phosphorylation has recently emerged as an important regulatory mechanism allowing mycobacteria to adapt their cell wall structure/composition in response to their environment. Herein, we evaluated whether phosphorylation of KasB, a crucial mycolic acid biosynthetic enzyme, could modulate acid-fast staining and virulence. Tandem mass spectrometry and site-directed mutagenesis revealed that phosphorylation of KasB occurred at Thr334 and Thr336 both in vitro and in mycobacteria. Isogenic strains of M. tuberculosis with either a deletion of the kasB gene or a kasB_T334D/T336D allele, mimicking constitutive phosphorylation of KasB, were constructed by specialized linkage transduction. Biochemical and structural analyses comparing these mutants to the parental strain revealed that both mutant strains had mycolic acids that were shortened by 4-6 carbon atoms and lacked trans-cyclopropanation. Together, these results suggested that in M. tuberculosis, phosphorylation profoundly decreases the condensing activity of KasB. Structural/modeling analyses reveal that Thr334 and Thr336 are located in the vicinity of the catalytic triad, which indicates that phosphorylation of these amino acids would result in loss of enzyme activity. Importantly, the kasB_T334D/T336D phosphomimetic and deletion alleles, in contrast to the kasB_T334A/T336A phosphoablative allele, completely lost acid-fast staining. Moreover, assessing the virulence of these strains indicated that the KasB phosphomimetic mutant was attenuated in both immunodeficient and immunocompetent mice following aerosol infection. This attenuation was characterized by the absence of lung pathology. Overall, these results highlight for the first time the role of Ser/Thr kinase-dependent KasB phosphorylation in regulating the later stages of mycolic acid elongation, with important consequences in terms of acid-fast staining and pathogenicity.


Assuntos
3-Oxoacil-(Proteína de Transporte de Acila) Sintase/metabolismo , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Ácidos Micólicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase/genética , Animais , Proteínas de Bactérias/genética , Domínio Catalítico/genética , Parede Celular/metabolismo , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Técnicas Microbiológicas/métodos , Modelos Moleculares , Mycobacterium tuberculosis/genética , Ácidos Micólicos/química , Fosforilação , Coloração e Rotulagem/métodos , Tuberculose/diagnóstico , Tuberculose/metabolismo , Tuberculose/microbiologia , Virulência
7.
Nat Chem Biol ; 9(8): 499-506, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770708

RESUMO

We report a new class of thiophene (TP) compounds that kill Mycobacterium tuberculosis by the previously uncharacterized mechanism of Pks13 inhibition. An F79S mutation near the catalytic Ser55 site in Pks13 conferred TP resistance in M. tuberculosis. Overexpression of wild-type Pks13 resulted in TP resistance, and overexpression of the Pks13(F79S) mutant conferred high resistance. In vitro, TP inhibited fatty acyl-AMP loading onto Pks13. TP inhibited mycolic acid biosynthesis in wild-type M. tuberculosis, but it did so to a much lesser extent in TP-resistant M. tuberculosis. TP treatment was bactericidal and equivalent to treatment with the first-line drug isoniazid, but it was less likely to permit emergent resistance. Combined isoniazid and TP treatment resulted in sterilizing activity. Computational docking identified a possible TP-binding groove within the Pks13 acyl carrier protein domain. This study confirms that M. tuberculosis Pks13 is required for mycolic acid biosynthesis, validates it as a druggable target and demonstrates the therapeutic potential of simultaneously inhibiting multiple targets in the same biosynthetic pathway.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/efeitos dos fármacos , Ácidos Micólicos/metabolismo , Policetídeo Sintases/antagonistas & inibidores , Policetídeo Sintases/metabolismo , Tiofenos/farmacologia , Antituberculosos/síntese química , Antituberculosos/química , Proteínas de Bactérias/genética , Biocatálise , Testes de Sensibilidade Microbiana , Estrutura Molecular , Mutação , Mycobacterium tuberculosis/citologia , Mycobacterium tuberculosis/metabolismo , Policetídeo Sintases/genética , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/química
8.
J Biol Chem ; 285(17): 12714-25, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20178986

RESUMO

Mycolic acids are key cell wall components for the survival, pathogenicity, and antibiotic resistance of the human tubercle bacillus. Although it was thought that Mycobacterium tuberculosis tightly regulates their production to adapt to prevailing environmental conditions, the molecular mechanisms governing mycolic acid biosynthesis remained extremely obscure. Meromycolic acids, the direct precursors of mycolic acids, are synthesized by a type II fatty acid synthase from acyl carrier protein-bound substrates that are extended iteratively, with a reductive cycle in each round of extension, the second step of which is catalyzed by the essential beta-ketoacyl-acyl carrier protein reductase, MabA. In this study, we investigated whether post-translational modifications of MabA might represent a strategy employed by M. tuberculosis to regulate mycolic acid biosynthesis. Indeed, we show here that MabA was efficiently phosphorylated in vitro by several M. tuberculosis Ser/Thr protein kinases, including PknB, as well as in vivo in mycobacteria. Mass spectrometric analyses using LC-ESI/MS/MS and site-directed mutagenesis identified three phosphothreonines, with Thr(191) being the primary phosphor-acceptor. A MabA_T191D mutant, designed to mimic constitutive phosphorylation, exhibited markedly decreased ketoacyl reductase activity compared with the wild-type protein, as well as impaired binding of the NADPH cofactor, as demonstrated by fluorescence spectroscopy. The hypothesis that phosphorylation of Thr(191) alters the enzymatic activity of MabA, and subsequently mycolic acid biosynthesis, was further supported by the fact that constitutive overexpression of the mabA_T191D allele in Mycobacterium bovis BCG strongly impaired mycobacterial growth. Importantly, conditional expression of the phosphomimetic MabA_T191D led to a significant inhibition of de novo biosynthesis of mycolic acids. This study provides the first information on the molecular mechanism(s) involved in mycolic acid regulation through Ser/Thr protein kinase-dependent phosphorylation of a type II fatty acid synthase enzyme.


Assuntos
Oxirredutases do Álcool/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Mycobacterium tuberculosis/enzimologia , Ácidos Micólicos/metabolismo , Oxirredutases/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , 3-Oxoacil-(Proteína Carreadora de Acil) Redutase , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Humanos , Mutagênese Sítio-Dirigida , Mycobacterium bovis/enzimologia , Mycobacterium bovis/genética , Mycobacterium tuberculosis/genética , Oxirredutases/química , Oxirredutases/genética , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Espectrometria de Fluorescência
9.
Mol Microbiol ; 78(6): 1591-605, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21143326

RESUMO

The remarkable survival ability of Mycobacterium tuberculosis in infected hosts is related to the presence of cell wall-associated mycolic acids. Despite their importance, the mechanisms that modulate expression of these lipids in response to environmental changes are unknown. Here we demonstrate that the enoyl-ACP reductase activity of InhA, an essential enzyme of the mycolic acid biosynthetic pathway and the primary target of the anti-tubercular drug isoniazid, is controlled via phosphorylation. Thr-266 is the unique kinase phosphoacceptor, both in vitro and in vivo. The physiological relevance of Thr-266 phosphorylation was demonstrated using inhA phosphoablative (T266A) or phosphomimetic (T266D/E) mutants. Enoyl reductase activity was severely impaired in the mimetic mutants in vitro, as a consequence of a reduced binding affinity to NADH. Importantly, introduction of inhA_T266D/E failed to complement growth and mycolic acid defects of an inhA-thermosensitive Mycobacterium smegmatis strain, in a similar manner to what is observed following isoniazid treatment. This study suggests that phosphorylation of InhA may represent an unusual mechanism that allows M. tuberculosis to regulate its mycolic acid content, thus offering a new approach to future anti-tuberculosis drug development.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Ácidos Micólicos/metabolismo , Oxirredutases/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Vias Biossintéticas , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/enzimologia , Oxirredutases/química , Oxirredutases/genética , Fosforilação
10.
FEBS J ; 287(20): 4415-4426, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31994828

RESUMO

Protein-protein interactions are key in mycobacterial physiology, notably during the biosynthesis of the very peculiar mycobacterial cell wall. In this paper, we demonstrate that MSMEG_1285 interacts with PonA1, a bifunctional penicillin-binding protein involved in peptidoglycan biosynthesis. Deletion of MSMEG_1285 enhances Mycobacterium smegmatis resistance to penicillin antibiotics, a phenotype that is exacerbated by the additional deletion of hbhA. This also led to a substantial decrease in the amounts of porins in the cell wall, which are necessary for the import of small and hydrophilic ß-lactams. Deletion of both MSMEG_1285 and hbhA provoked an over-representation of several enzymes involved in peptidoglycan degradation. Thus, we propose that MSMEG_1285 is part of a protein scaffold, which also involves PonA1 and HbhA, and that it is responsible for the tight regulation of peptidoglycan hydrolysis. This study provides a better understanding of the mycobacterial physiology, which is an essential step for strengthening the action of drugs that specifically target peptidoglycan biosynthesis.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Parede Celular/efeitos dos fármacos , Mycobacterium/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Penicilinas/farmacologia , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Hidrólise , Testes de Sensibilidade Microbiana , Mycobacterium/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/metabolismo
11.
Pathogens ; 8(4)2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31581602

RESUMO

Studies on protein-protein interactions (PPI) can be helpful for the annotation of unknown protein functions and for the understanding of cellular processes, such as specific virulence mechanisms developed by bacterial pathogens. In that context, several methods have been extensively used in recent years for the characterization of Mycobacterium tuberculosis PPI to further decipher tuberculosis (TB) pathogenesis. This review aims at compiling the most striking results based on in vivo methods (yeast and bacterial two-hybrid systems, protein complementation assays) for the specific study of PPI in mycobacteria. Moreover, newly developed methods, such as in-cell native mass resonance and proximity-dependent biotinylation identification, will have a deep impact on future mycobacterial research, as they are able to perform dynamic (transient interactions) and integrative (multiprotein complexes) analyses.

12.
J Mol Biol ; 353(4): 847-58, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16213523

RESUMO

Tuberculosis kills about two million people every year and remains one of the leading causes of mortality worldwide. As a result of the increasing antibiotic resistance of Mycobacterium tuberculosis (Mtb) strains, there is an urgent need for new antitubercular drugs. Several efficient antibiotics, including isoniazid, specifically target the fatty acid synthase-II (FAS-II) complex of mycolic acid biosynthesis. We have previously shown that there are protein-protein interactions between the components of FAS-II that are essential for mycobacterial survival. We have now looked at the potential partners of FAS-II, mtFabD, the methyltransferases MmaAs, and Pks13. A combination of yeast two-hybrid and co-immunoprecipitation experiments showed that mtFabD interacts with each beta-ketoacyl-synthase (KasA, KasB and mtFabH) and with the core of FAS-II (InhA and MabA). The methyltransferases have a greater affinity for KasA and KasB than for mtFabH, suggesting that modifications on the meromycolic chains may occur during their elongation. Finally, Pks13, which catalyzes the final Claisen condensation of mycolic acids, interacts specifically with KasB. These data allowed us to determine the architecture of the multiple specialized FAS-II complexes, giving us insights into the organization of the complete mycolic acids biosynthesis. Our studies suggest a new and crucial interaction (KasB-Pks13) as a putative target for peptidomimetic antibiotics.


Assuntos
Acetiltransferases/metabolismo , Complexos Multienzimáticos/metabolismo , Mycobacterium tuberculosis/enzimologia , Ácidos Micólicos/metabolismo , Mapeamento de Interação de Proteínas , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase/metabolismo , Proteínas de Bactérias/metabolismo , Ácido Graxo Sintase Tipo II , Imunoprecipitação , Metiltransferases/metabolismo , Proteínas Mitocondriais/metabolismo , Policetídeo Sintases/metabolismo , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
13.
Structure ; 24(10): 1788-1794, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27568926

RESUMO

Mycobacterium tuberculosis (Mtb) encodes several bacterial effectors impacting the colonization of phagocytes. LppM (Rv2171) is both implicated in phagocytosis and able to efficiently block phagosomal acidification in the macrophage, two key processes contributing to Mtb persistence. We show that LppM is anchored to the mycobacterial cell wall by a C-terminal membrane domain. However, the protein also exists as a truncated protein secreted into the culture medium. The LppM solution structure we solve here displays no similarity with other Mtb lipoproteins also involved in phagosomal maturation (i.e., LprG). In addition, we demonstrate that the protein may be able to bind rare molecular species of phosphatidylinositol mannosides, bacterial compounds known to affect the host immune response. Thus, our data demonstrate a dual localization of LppM and provide a unique perspective on the regulation of protein secretion and localization in Mtb.


Assuntos
Parede Celular/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Mycobacterium tuberculosis/metabolismo , Fosfatidilinositóis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Meios de Cultura/química , Regulação Bacteriana da Expressão Gênica , Espectrometria de Massas , Modelos Moleculares , Mycobacterium tuberculosis/química , Fagocitose , Domínios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína
14.
Sci Rep ; 6: 29297, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27384401

RESUMO

Mycobacterium tuberculosis is a successful intracellular pathogen. Numerous host innate immune responses signaling pathways are induced upon mycobacterium invasion, however their impact on M. tuberculosis replication is not fully understood. Here we reinvestigate the role of STAT3 specifically inside human macrophages shortly after M. tuberculosis uptake. We first show that STAT3 activation is mediated by IL-10 and occurs in M. tuberculosis infected cells as well as in bystander non-colonized cells. STAT3 activation results in the inhibition of IL-6, TNF-α, IFN-γ and MIP-1ß. We further demonstrate that STAT3 represses iNOS expression and NO synthesis. Accordingly, the inhibition of STAT3 is detrimental for M. tuberculosis intracellular replication. Our study thus points out STAT3 as a key host factor for M. tuberculosis intracellular establishment in the early stages of macrophage infection.


Assuntos
Macrófagos/metabolismo , Óxido Nítrico Sintase/metabolismo , Fator de Transcrição STAT3/metabolismo , Tuberculose/metabolismo , Animais , Linhagem Celular , Quimiocina CCL4/metabolismo , Humanos , Imunidade Inata/imunologia , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Macrófagos/microbiologia , Camundongos , Mycobacterium tuberculosis/imunologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Transdução de Sinais/fisiologia , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/metabolismo
15.
J Med Chem ; 57(17): 7382-95, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25158122

RESUMO

Mycolactone is a complex macrolide toxin produced by Mycobacterium ulcerans, the causative agent of skin lesions called Buruli ulcers. Mycolactone-mediated activation of neural (N) Wiskott-Aldrich syndrome proteins (WASP) induces defects in cell adhesion underpinning cytotoxicity and disease pathogenesis. We describe the chemical synthesis of 23 novel mycolactone analogues that differ in structure and modular assembly of the lactone core with its northern and southern polyketide side chains. The lactone core linked to southern chain was the minimal structure binding N-WASP and hematopoietic homolog WASP, where the number and configuration of hydroxyl groups on the acyl side chain impacted the degree of binding. A fluorescent derivative of this compound showed time-dependent accumulation in target cells. Furthermore, a simplified version of mycolactone mimicked the natural toxin for activation of WASP in vitro and induced comparable alterations of epithelial cell adhesion. Therefore, it constitutes a structural and functional surrogate of mycolactone for WASP/N-WASP-dependent effects.


Assuntos
Toxinas Bacterianas/química , Macrolídeos/química , Proteína da Síndrome de Wiskott-Aldrich/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Adesão Celular/efeitos dos fármacos , Células HeLa , Humanos , Cinética , Macrolídeos/metabolismo , Macrolídeos/farmacologia , Modelos Químicos , Estrutura Molecular , Mycobacterium ulcerans/química , Ligação Proteica , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
16.
J Vis Exp ; (83): e51114, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24473237

RESUMO

Despite the availability of therapy and vaccine, tuberculosis (TB) remains one of the most deadly and widespread bacterial infections in the world. Since several decades, the sudden burst of multi- and extensively-drug resistant strains is a serious threat for the control of tuberculosis. Therefore, it is essential to identify new targets and pathways critical for the causative agent of the tuberculosis, Mycobacterium tuberculosis (Mtb) and to search for novel chemicals that could become TB drugs. One approach is to set up methods suitable for the genetic and chemical screens of large scale libraries enabling the search of a needle in a haystack. To this end, we developed a phenotypic assay relying on the detection of fluorescently labeled Mtb within fluorescently labeled host cells using automated confocal microscopy. This in vitro assay allows an image based quantification of the colonization process of Mtb into the host and was optimized for the 384-well microplate format, which is proper for screens of siRNA-, chemical compound- or Mtb mutant-libraries. The images are then processed for multiparametric analysis, which provides read out inferring on the pathogenesis of Mtb within host cells.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Mycobacterium tuberculosis/isolamento & purificação , Células Epiteliais Alveolares/microbiologia , Humanos , Microscopia Confocal/métodos , Mycobacterium tuberculosis/genética , Fenótipo , RNA Interferente Pequeno/análise , RNA Interferente Pequeno/genética , Tuberculose/microbiologia
17.
J Clin Invest ; 123(4): 1501-12, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23549080

RESUMO

Mycolactone is a diffusible lipid secreted by the human pathogen Mycobacterium ulcerans, which induces the formation of open skin lesions referred to as Buruli ulcers. Here, we show that mycolactone operates by hijacking the Wiskott-Aldrich syndrome protein (WASP) family of actin-nucleating factors. By disrupting WASP autoinhibition, mycolactone leads to uncontrolled activation of ARP2/3-mediated assembly of actin in the cytoplasm. In epithelial cells, mycolactone-induced stimulation of ARP2/3 concentrated in the perinuclear region, resulting in defective cell adhesion and directional migration. In vivo injection of mycolactone into mouse ears consistently altered the junctional organization and stratification of keratinocytes, leading to epidermal thinning, followed by rupture. This degradation process was efficiently suppressed by coadministration of the N-WASP inhibitor wiskostatin. These results elucidate the molecular basis of mycolactone activity and provide a mechanism for Buruli ulcer pathogenesis. Our findings should allow for the rationale design of competitive inhibitors of mycolactone binding to N-WASP, with anti-Buruli ulcer therapeutic potential.


Assuntos
Toxinas Bacterianas/farmacologia , Úlcera de Buruli/metabolismo , Macrolídeos/farmacologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/química , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Úlcera de Buruli/microbiologia , Úlcera de Buruli/patologia , Carbazóis/farmacologia , Adesão Celular , Movimento Celular , Núcleo Celular/metabolismo , Epiderme/efeitos dos fármacos , Epiderme/patologia , Células HeLa , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mycobacterium ulcerans , Propanolaminas/farmacologia , Multimerização Proteica , Transporte Proteico , Família de Proteínas da Síndrome de Wiskott-Aldrich/antagonistas & inibidores , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/antagonistas & inibidores
18.
Tuberculosis (Edinb) ; 91(6): 544-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21802366

RESUMO

Small membrane proteins emerge as a novel class of regulatory molecules in bacteria. Experiments carried out in Mycobacterium bovis BCG indicate that the ompATb gene (Rv0899), encoding a major outer membrane protein, is organized in operon with Rv0900 and Rv0901, encoding two small proteins with a predicted transmembrane domain. Fractioning experiment confirmed the association of Rv0901 with the membrane fraction. To investigate the role of Rv0900 and Rv0901 in M. bovis BCG, we have constructed a strain deleted for the whole operon as well as complemented strains carrying a deletion of Rv0900 or a frameshift mutation in either Rv0900 or Rv0901. Importantly, mutations in Rv0900 and/or Rv0901 strongly altered OmpATb expression, demonstrating that Rv0900 and Rv0901 play a regulatory role, which appears to occur at a post-transcriptional level.


Assuntos
Proteínas de Bactérias/genética , Mutação da Fase de Leitura , Regulação Bacteriana da Expressão Gênica , Mycobacterium bovis/genética , Porinas/genética , Deleção de Sequência , Vacina BCG/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Western Blotting , Humanos , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mycobacterium bovis/metabolismo , Óperon , Fenótipo , Porinas/metabolismo
19.
PLoS One ; 6(12): e29564, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22216317

RESUMO

BACKGROUND: The human pathogen Mycobacterium tuberculosis (Mtb) has the originality of possessing a multifunctional mega-enzyme FAS-I (Fatty Acid Synthase-I), together with a multi-protein FAS-II system, to carry out the biosynthesis of common and of specific long chain fatty acids: the mycolic acids (MA). MA are the main constituents of the external mycomembrane that represents a tight permeability barrier involved in the pathogenicity of Mtb. The MA biosynthesis pathway is essential and contains targets for efficient antibiotics. We have demonstrated previously that proteins of FAS-II interact specifically to form specialized and interconnected complexes. This finding suggested that the organization of FAS-II resemble to the architecture of multifunctional mega-enzyme like the mammalian mFAS-I, which is devoted to the fatty acid biosynthesis. PRINCIPAL FINDINGS: Based on conventional and reliable studies using yeast-two hybrid, yeast-three-hybrid and in vitro Co-immunoprecipitation, we completed here the analysis of the composition and architecture of the interactome between the known components of the Mtb FAS-II complexes. We showed that the recently identified dehydratases HadAB and HadBC are part of the FAS-II elongation complexes and may represent a specific link between the core of FAS-II and the condensing enzymes of the system. By testing four additional methyltransferases involved in the biosynthesis of mycolic acids, we demonstrated that they display specific interactions with each type of complexes suggesting their coordinated action during MA elongation. SIGNIFICANCE: These results provide a global update of the architecture and organization of a FAS-II system. The FAS-II system of Mtb is organized in specialized interconnected complexes and the specificity of each elongation complex is given by preferential interactions between condensing enzymes and dehydratase heterodimers. This study will probably allow defining essential and specific interactions that correspond to promising targets for Mtb FAS-II inhibitors.


Assuntos
Ácido Graxo Sintase Tipo II/metabolismo , Hidroliases/metabolismo , Mycobacterium tuberculosis/enzimologia , Ácidos Micólicos/metabolismo , Dimerização , Ácido Graxo Sintase Tipo II/química , Hidroliases/química , Imunoprecipitação
20.
PLoS Negl Trop Dis ; 5(7): e1237, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21811642

RESUMO

BACKGROUND: Mycobacterium ulcerans, the causative agent of Buruli ulcer (BU), is unique among human pathogens in its capacity to produce a polyketide-derived macrolide called mycolactone, making this molecule an attractive candidate target for diagnosis and disease monitoring. Whether mycolactone diffuses from ulcerated lesions in clinically accessible samples and is modulated by antibiotic therapy remained to be established. METHODOLOGY/PRINCIPAL FINDING: Peripheral blood and ulcer exudates were sampled from patients at various stages of antibiotic therapy in Ghana and Ivory Coast. Total lipids were extracted from serum, white cell pellets and ulcer exudates with organic solvents. The presence of mycolactone in these extracts was then analyzed by a recently published, field-friendly method using thin layer chromatography and fluorescence detection. This approach did not allow us to detect mycolactone accurately, because of a high background due to co-extracted human lipids. We thus used a previously established approach based on high performance liquid chromatography coupled to mass spectrometry. By this means, we could identify structurally intact mycolactone in ulcer exudates and serum of patients, and evaluate the impact of antibiotic treatment on the concentration of mycolactone. CONCLUSIONS/SIGNIFICANCE: Our study provides the proof of concept that assays based on mycolactone detection in serum and ulcer exudates can form the basis of BU diagnostic tests. However, the identification of mycolactone required a technology that is not compatible with field conditions and point-of-care assays for mycolactone detection remain to be worked out. Notably, we found mycolactone in ulcer exudates harvested at the end of antibiotic therapy, suggesting that the toxin is eliminated by BU patients at a slow rate. Our results also indicated that mycolactone titres in the serum may reflect a positive response to antibiotics, a possibility that it will be interesting to examine further through longitudinal studies.


Assuntos
Toxinas Bacterianas/sangue , Úlcera de Buruli/sangue , Úlcera de Buruli/microbiologia , Mycobacterium ulcerans/isolamento & purificação , Adolescente , Adulto , Antibacterianos/uso terapêutico , Toxinas Bacterianas/análise , Biomarcadores/análise , Biomarcadores/sangue , Úlcera de Buruli/diagnóstico , Úlcera de Buruli/tratamento farmacológico , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Exsudatos e Transudatos/química , Exsudatos e Transudatos/microbiologia , Feminino , Humanos , Leucócitos Mononucleares/química , Leucócitos Mononucleares/microbiologia , Macrolídeos , Masculino , Espectrometria de Massas , Mycobacterium ulcerans/química , Ferimentos e Lesões/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA