Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Bioorg Med Chem Lett ; 98: 129596, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38142914

RESUMO

To identify new compounds that can effectively inhibit Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), we screened, synthesized, and evaluated a series of novel aryl fluorosulfate derivatives for their in vitro inhibitory activity against Mtb. Compound 21b exhibited an in vitro minimum inhibitory concentration (MIC) of 0.06 µM against Mtb, no cytotoxicity against both HEK293T and HepG2 mammalian cell lines, and had good in vivo mouse plasma exposure and lung concentration with a 20 mg/kg oral dose, which supports advanced development as a new chemical entity for TB treatment.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Camundongos , Antituberculosos , Células HEK293 , Mamíferos , Testes de Sensibilidade Microbiana , Relação Estrutura-Atividade , Tuberculose/tratamento farmacológico , Ésteres do Ácido Sulfúrico/química , Ésteres do Ácido Sulfúrico/farmacologia
2.
Eur J Immunol ; 49(6): 894-910, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30912587

RESUMO

It is established that iNKT cells are a cell type that require strong TCR signal for their proper development and represent a model for thymic agonist selection. The nature of the signal perceived by iNKT cells promoting their specification is not well understood. To address this question, we analyzed iNKT cell development in relevant TCR Vα14-Jα18 alpha chain transgenic mice (Vα14Tg). In CD4-Vα14Tg mice, where the transgene is driven by CD4 promoter, we identified a block in iNKT cell development at early developmental stages due to a reduced expression of key transcription factors accompanied with a reduced TCR expression levels. This indicates that TCR signal strength control iNKT cell differentiation. Importantly, we found in WT mice that early precursors of iNKT cells express higher TCR levels compared to positively selected precursors of mainstream T cells showing that TCR levels could contribute to the strength of iNKT cell TCR signaling. Overall, our study highlights TCR signal strength associated with a higher TCR density as an important regulator of iNKT cell lineage specification.


Assuntos
Células T Matadoras Naturais/citologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Subpopulações de Linfócitos T/citologia , Animais , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Camundongos , Camundongos Transgênicos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
3.
PLoS Pathog ; 13(5): e1006399, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28545104

RESUMO

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


Assuntos
Aminoácidos/metabolismo , Proteínas de Bactérias/metabolismo , Metabolômica , Mycobacterium tuberculosis , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Ácido Aspártico/metabolismo , Proteínas de Bactérias/genética , Ácido Glutâmico/metabolismo , Homeostase , Macrófagos/microbiologia , Camundongos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Tuberculose/microbiologia , Virulência
4.
Cell Microbiol ; 16(2): 280-95, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24119166

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium marinum/metabolismo , Fatores de Virulência/metabolismo , Animais , Elementos de DNA Transponíveis , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Immunoblotting , Mutagênese Insercional , Infecções por Mycobacterium/microbiologia , Mycobacterium marinum/patogenicidade , Especificidade por Substrato , Peixe-Zebra
5.
Cell Microbiol ; 14(3): 356-67, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22050772

RESUMO

The role and function of PE_PGRS proteins of Mycobacterium tuberculosis (Mtb) remains elusive. In this study for the first time, Mtb isogenic mutants missing selected PE_PGRSs were used to investigate their role in the pathogenesis of tuberculosis (TB). We demonstrate that the MtbΔPE_PGRS30 mutant was impaired in its ability to colonize lung tissue and to cause tissue damage, specifically during the chronic steps of infection. Inactivation of PE_PGRS30 resulted in an attenuated phenotype in murine and human macrophages due to the inability of the Mtb mutant to inhibit phagosome-lysosome fusion. Using a series of functional deletion mutants of PE_PGRS30 to complement MtbΔPE_PGRS30, we show that the unique C-terminal domain of the protein is not required for the full virulence. Interestingly, when Mycobacterium smegmatis recombinant strain expressing PE_PGRS30 was used to infect macrophages or mice in vivo, we observed enhanced cytotoxicity and cell death, and this effect was dependent upon the PGRS domain of the protein.Taken together these results indicate that PE_PGRS30 is necessary for the full virulence of Mtb and sufficient to induce cell death in host cells by the otherwise non-pathogenic species M. smegmatis, clearly demonstrating that PE_PGRS30 is an Mtb virulence factor.


Assuntos
Proteínas de Bactérias/genética , Mycobacterium tuberculosis/patogenicidade , Tuberculose Pulmonar/microbiologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Células Cultivadas , Feminino , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagossomos/microbiologia , Estrutura Terciária de Proteína , Virulência
6.
J Biol Chem ; 286(21): 19024-34, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21471225

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Lipase/metabolismo , Mycobacterium marinum/enzimologia , Mycobacterium tuberculosis/enzimologia , Sinais Direcionadores de Proteínas/fisiologia , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Lipase/genética , Estrutura Terciária de Proteína , Especificidade da Espécie
7.
J Proteomics ; 244: 104276, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34044169

RESUMO

Mycobacterium tuberculosis, the etiological agent of tuberculosis, is among the deadliest human pathogens. One of M. tuberculosis's pathogenic hallmarks is its ability to persist in a dormant state in the host. Thus, this pathogen has developed mechanisms to withstand stressful conditions found in the human host. Particularly, the Ser/Thr-protein kinase PknG has gained relevance since it regulates nitrogen metabolism and facilitates bacterial survival inside macrophages. Nevertheless, the molecular mechanisms underlying these effects are far from being elucidated. To further investigate these issues, we performed quantitative proteomic analyses of protein extracts from M. tuberculosis H37Rv and a mutant lacking pknG. We found that in the absence of PknG the mycobacterial proteome was remodeled since 5.7% of the proteins encoded by M. tuberculosis presented significant changes in its relative abundance compared with the wild-type. The main biological processes affected by pknG deletion were cell envelope components biosynthesis and response to hypoxia. Thirteen DosR-regulated proteins were underrepresented in the pknG deletion mutant, including Hrp-1, which was 12.5-fold decreased according to Parallel Reaction Monitoring experiments. Altogether, our results allow us to postulate that PknG regulation of bacterial adaptation to stress conditions might be an important mechanism underlying its reported effect on intracellular bacterial survival. SIGNIFICANCE: PknG is a Ser/Thr kinase from Mycobacterium tuberculosis with key roles in bacterial metabolism and bacterial survival within the host. However, at present the molecular mechanisms underlying these functions remain largely unknown. In this work, we evaluate the effect of pknG deletion on M. tuberculosis proteome using different approaches. Our results clearly show that the global proteome was remodeled in the absence of PknG and shed light on new molecular mechanism underlying PknG role. Altogether, this work contributes to a better understanding of the molecular bases of the adaptation of M. tuberculosis, one of the most deadly human pathogens, to its host.


Assuntos
Fenômenos Biológicos , Mycobacterium tuberculosis , Proteínas de Bactérias/genética , Humanos , Hipóxia , Mycobacterium tuberculosis/genética , Proteínas Serina-Treonina Quinases/genética , Proteoma , Proteômica
8.
Infect Immun ; 78(12): 5202-13, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20921146

RESUMO

To improve the current vaccine against tuberculosis, a recombinant strain of Mycobacterium bovis bacillus Calmette-Guérin (rBCG) expressing a Mycobacterium tuberculosis vaccine candidate antigen (MPT64) in strong association with the mycobacterial cell wall was developed. To deliver the candidate antigen on the surface, we fused the mpt64 gene to the sequence encoding the PE domain of the PE_PGRS33 protein of M. tuberculosis (to create strain (H)PE-ΔMPT64-BCG), which we have previously shown to transport proteins to the bacterial surface. In a series of protection experiments in the mouse model of tuberculosis, we showed that (i) immunization of mice with (H)PE-ΔMPT64-BCG provides levels of protection significantly higher than those afforded by the parental BCG strain, as assessed by bacterial colonization in lungs and spleens and by lung involvement (at both 28 and 70 days postchallenge), (ii) rBCG strains expressing MPT64 provide better protection than the parental BCG strain only when this antigen is surface expressed, and (iii) the (H)PE-ΔMPT64-BCG-induced MPT64-specific T cell repertoire when characterized by ß chain variable region-ß chain joining region (BV-BJ) spectratyping indicates that protection is correlated with the ability to recruit gamma interferon (IFN-γ)-secreting T cells carrying the BV8.3-BJ1.5 (172 bp) shared rearrangement. These results demonstrate that (H)PE-ΔMPT64-BCG is one of the most effective new vaccines tested so far in the mouse model of tuberculosis and underscore the impact of antigen cellular localization on the induction of the specific immune response induced by rBCG.


Assuntos
Antígenos de Bactérias/imunologia , Vacina BCG/genética , Proteínas de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/prevenção & controle , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/uso terapêutico , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Fusão Gênica Artificial , Vacina BCG/imunologia , Vacina BCG/uso terapêutico , Proteínas de Bactérias/genética , Proteínas de Bactérias/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Feminino , Interferon gama/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/imunologia , Tuberculose/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
9.
Appl Environ Microbiol ; 76(15): 5312-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20543044

RESUMO

Genetic manipulation of mycobacteria still represents a serious challenge due to the lack of tools and selection markers. In this report, we describe the development of an intrinsically unstable excisable cassette for introduction of unmarked mutations in both Mycobacterium smegmatis and Mycobacterium tuberculosis.


Assuntos
Genética Microbiana/métodos , Mutagênese Insercional/métodos , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Recombinação Genética , Deleção de Sequência
10.
J Proteomics ; 192: 321-333, 2019 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-30267874

RESUMO

PknG from Mycobacterium tuberculosis is a multidomain Serine/Threonine protein kinase that regulates bacterial metabolism as well as the pathogen's ability to survive inside the host by still uncertain mechanisms. To uncover PknG interactome we developed an affinity purification-mass spectrometry strategy to stepwise recover PknG substrates and interactors; and to identify those involving PknG autophosphorylated docking sites. We report a confident list of 7 new putative substrates and 66 direct or indirect partners indicating that PknG regulates many physiological processes, such as nitrogen and energy metabolism, cell wall synthesis and protein translation. GarA and the 50S ribosomal protein L13, two previously reported substrates of PknG, were recovered in our interactome. Comparative proteome analyses of wild type and pknG null mutant M. tuberculosis strains provided evidence that two kinase interactors, the FHA-domain containing protein GarA and the enzyme glutamine synthetase, are indeed endogenous substrates of PknG, stressing the role of this kinase in the regulation of nitrogen metabolism. Interestingly, a second FHA protein was identified as a PknG substrate. Our results show that PknG phosphorylates specific residues in both glutamine synthetase and FhaA in vitro, and suggest that these proteins are phosphorylated by PknG in living mycobacteria.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Mutação , Mycobacterium tuberculosis/genética , Fosforilação , Domínios Proteicos , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Especificidade por Substrato
11.
J Med Chem ; 60(17): 7425-7433, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28846409

RESUMO

In this study, we aimed to decipher the natural resistance mechanisms of mycobacteria against novel compounds isolated by whole-cell-based high-throughput screening (HTS). We identified active compounds using Mycobacterium aurum. Further analyses were performed to determine the resistance mechanism of M. smegmatis against one hit, 3-bromo-N-(5-nitrothiazol-2-yl)-4-propoxybenzamide (3), which turned out to be an analog of the drug nitazoxanide (1). We found that the repression of the gene nfnB coding for the nitroreductase NfnB was responsible for the natural resistance of M. smegmatis against 3. The overexpression of nfnB resulted in sensitivity of M. smegmatis to 3. This compound must be metabolized into hydroxylamine intermediate for exhibiting antibacterial activity. Thus, we describe, for the first time, the activity of a mycobacterial nitroreductase against 1 analogs, highlighting the differences in the metabolism of nitro compounds among mycobacterial species and emphasizing the potential of nitro drugs as antibacterials in various bacterial species.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/enzimologia , Nitrorredutases/metabolismo , Tiazóis/química , Tiazóis/farmacologia , Regulação para Baixo , Farmacorresistência Bacteriana , Humanos , Mycobacterium/efeitos dos fármacos , Mycobacterium/enzimologia , Mycobacterium/genética , Infecções por Mycobacterium/tratamento farmacológico , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium smegmatis/genética , Nitrocompostos , Nitrorredutases/genética
12.
PLoS One ; 11(3): e0150800, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26978522

RESUMO

PE_PGRS represent a large family of proteins typical of pathogenic mycobacteria whose members are characterized by an N-terminal PE domain followed by a large Gly-Ala repeat-rich C-terminal domain. Despite the abundance of PE_PGRS-coding genes in the Mycobacterium tuberculosis (Mtb) genome their role and function in the biology and pathogenesis still remains elusive. In this study, we generated and characterized an Mtb H37Rv mutant (MtbΔ33) in which the structural gene of PE_PGRS33, a prototypical member of the protein family, was inactivated. We showed that this mutant entered macrophages with an efficiency up to ten times lower than parental or complemented strains, while its efficiency in infecting pneumocytes remained unaffected. Interestingly, the lack of PE_PGRS33 did not affect the intracellular growth of this mutant in macrophages. Using a series of functional deletion mutants of the PE_PGRS33 gene to complement the MtbΔ33 strain, we demonstrated that the PGRS domain is required to mediate cell entry into macrophages, with the key domain encompassing position 140-260 amino acids of PE_PGRS33. PE_PGRS33-mediated entry into macrophages was abolished in TLR2-deficient mice, as well as following treatment with wortmannin or an antibody against the complement receptor 3 (CR3), indicating that PE_PGRS33-mediated entry of Mtb in macrophages occurs through interaction with TLR2.


Assuntos
Proteínas de Bactérias/fisiologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/fisiologia , Receptor 2 Toll-Like/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL
13.
Nat Microbiol ; 1: 15019, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-27571976

RESUMO

Mycobacterium tuberculosis is a major, globally spread, aerosol-transmitted human pathogen, thought to have evolved by clonal expansion from a Mycobacterium canettii-like progenitor. In contrast, extant M. canettii strains are rare, genetically diverse, and geographically restricted mycobacteria of only marginal epidemiological importance. Here, we show that the contrasting evolutionary success of these two groups is linked to loss of lipooligosaccharide biosynthesis and subsequent morphotype changes. Spontaneous smooth-to-rough M. canettii variants were found to be mutated in the polyketide-synthase-encoding pks5 locus and deficient in lipooligosaccharide synthesis, a phenotype restored by complementation. Importantly, these rough variants showed an altered host-pathogen interaction and increased virulence in cellular- and animal-infection models. In one variant, lipooligosaccharide deficiency occurred via homologous recombination between two pks5 genes and removal of the intervening acyltransferase-encoding gene. The resulting single pks5 configuration is similar to that fixed in M. tuberculosis, which is known to lack lipooligosaccharides. Our results suggest that pks5-recombination-mediated bacterial surface remodelling increased virulence, driving evolution from putative generalist mycobacteria towards professional pathogens of mammalian hosts.


Assuntos
Vias Biossintéticas , Evolução Molecular , Lipopolissacarídeos/biossíntese , Mycobacterium/genética , Mycobacterium/patogenicidade , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo , Animais , Modelos Animais de Doenças , Deleção de Genes , Teste de Complementação Genética , Recombinação Homóloga , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/patologia , Virulência
14.
Sci Rep ; 5: 16918, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26603639

RESUMO

Mycobacterium tuberculosis, the agent of human tuberculosis has developed different virulence mechanisms and virulence-associated tools during its evolution to survive and multiply inside the host. Based on previous reports and by analogy with other bacteria, phospholipases C (PLC) of M. tuberculosis were thought to be among these tools. To get deeper insights into the function of PLCs, we investigated their putative involvement in the intracellular lifestyle of M. tuberculosis, with emphasis on phagosomal rupture and virulence, thereby re-visiting a research theme of longstanding interest. Through the construction and use of an M. tuberculosis H37Rv PLC-null mutant (ΔPLC) and control strains, we found that PLCs of M. tuberculosis were not required for induction of phagosomal rupture and only showed marginal, if any, impact on virulence of M. tuberculosis in the cellular and mouse infection models used in this study. In contrast, we found that PLC-encoding genes were strongly upregulated under phosphate starvation and that PLC-proficient M. tuberculosis strains survived better than ΔPLC mutants under conditions where phosphatidylcholine served as sole phosphate source, opening new perspectives for studies on the role of PLCs in the lifecycle of M. tuberculosis.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose/enzimologia , Fosfolipases Tipo C/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Colorimetria , Feminino , Humanos , Estágios do Ciclo de Vida , Pulmão/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Óperon/genética , Fagossomos/metabolismo , Fosfatidilcolinas/metabolismo , Baço/microbiologia , Tuberculose/microbiologia , Tuberculose/patologia , Fosfolipases Tipo C/deficiência , Fosfolipases Tipo C/genética , Virulência/genética
15.
Curr Pharm Des ; 20(27): 4346-56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24245757

RESUMO

The emergence of multi- and extensively-drug resistant strains of Mycobacterium tuberculosis makes the development of novel anti-tubercular compounds and the identification of alternative mycobacterial drugable targets urgent priorities. Recently, type VII secretion systems (T7SS) have been discovered in mycobacteria. The genome of M. tuberculosis encodes 5 of such systems (ESX-1 to -5), three of which have been characterized and shown to be essential for viability (ESX-3, ESX-5) or virulence (ESX-1, ESX-5). Because of their crucial role in host-pathogen interactions as well as their involvement in basic biological processes of tubercle bacilli, T7SS/ESX represent promising targets for novel anti-tuberculosis drugs. Here, we review the current knowledge of the T7SS/ESX and their impact on M. tuberculosis physiology and virulence. Finally, we discuss the possible approaches to develop T7SS/ESX inhibitors.


Assuntos
Antituberculosos , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/efeitos dos fármacos , Descoberta de Drogas/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Antituberculosos/química , Antituberculosos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/genética , Sequência de Bases , Sítios de Ligação , Parede Celular/metabolismo , Dados de Sequência Molecular , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Alinhamento de Sequência , Virulência
16.
Future Microbiol ; 9(8): 969-85, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25302954

RESUMO

Mycobacterium tuberculosis, the etiological agent of human TB, is the most important mycobacterial pathogen in terms of global patient numbers and gravity of disease. The molecular mechanisms by which M. tuberculosis causes disease are complex and the result of host-pathogen coevolution that might have started already in the time of its Mycobacterium canettii-like progenitors. Despite research progress, M. tuberculosis still holds many secrets of its successful strategy for circumventing host defences, persisting in the host and developing resistance, which makes anti-TB treatment regimens extremely long and often inefficient. Here, we discuss what we have learned from recent studies on the evolution of the pathogen and its putative new drug targets that are essential for mycobacterial growth under in vitro or in vivo conditions.


Assuntos
Antituberculosos/farmacologia , Desenho de Fármacos , Evolução Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Animais , Antituberculosos/classificação , Genes Bacterianos , Genes Essenciais , Humanos , Camundongos , Terapia de Alvo Molecular , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Tuberculose/tratamento farmacológico , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
17.
PLoS One ; 8(3): e57517, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469198

RESUMO

PPE represent a peculiar family of mycobacterial proteins characterized by a 180 aminoacids conserved N-terminal domain. Several PPE genes are co-transcribed with a gene encoding for a protein belonging to another family of mycobacterial specific proteins named PE. Only one PE-PPE couple has been extensively characterized so far (PE25-PPE41) and it was shown that these two proteins form a heterodimer and that this interaction is essential for PPE41 stability and translocation through the mycobacterial cell wall. In this study we characterize the PE11-PPE17 couple. In contrast with what was found for PE25-PPE41, we show that PPE17 is not secreted but surface exposed. Moreover, we demonstrate that the presence of PE11 is not necessary for PPE17 stability or for its localization on the mycobacterial surface. Finally, we show that the PPE domain of PPE17 targets the mycobacterial cell wall and that this domain can be used as a fusion partner to expose heterologous proteins on the mycobacterial surface.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Parede Celular/genética , Mycobacterium bovis/genética , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Parede Celular/metabolismo , Expressão Gênica , Mycobacterium bovis/metabolismo , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/metabolismo , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Transporte Proteico
18.
Pathog Dis ; 69(3): 232-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24106104

RESUMO

PE_PGRSs are a large family of proteins identified in Mycobacterium tuberculosis complex and in few other pathogenic mycobacteria. The PE domain of PE_PGRS33 mediates localization of the protein on the mycobacterial cell surface, where the PGRS domain is available to interact with host components. In this study, PE_PGRS33 and its functional deletion mutants were expressed in M. smegmatis, and in vitro and in vivo assays were used to dissect the protein domains involved in the immunomodulatory properties of the protein. We demonstrate that PE_PGRS33-mediated secretion of TNF-α by macrophages occurs by extracellular interaction with TLR2. Our results also show that while the PGRS domain of the protein is required for triggering TNF-α secretion, mutation in the PE domain affects the pro-inflammatory properties of the protein. These results indicate that PE_PGRS33 is a protein with immunomodulatory activity and that protein stability and localization on the mycobacterial surface can affect these properties.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Mycobacterium tuberculosis/imunologia , Domínios e Motivos de Interação entre Proteínas , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Morte Celular/genética , Espaço Extracelular , Feminino , Expressão Gênica , Imunomodulação , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutação , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
19.
Nat Genet ; 45(2): 172-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23291586

RESUMO

Global spread and limited genetic variation are hallmarks of M. tuberculosis, the agent of human tuberculosis. In contrast, Mycobacterium canettii and related tubercle bacilli that also cause human tuberculosis and exhibit unusual smooth colony morphology are restricted to East Africa. Here, we sequenced and analyzed the whole genomes of five representative strains of smooth tubercle bacilli (STB) using Sanger (4-5× coverage), 454/Roche (13-18× coverage) and/or Illumina DNA sequencing (45-105× coverage). We show that STB isolates are highly recombinogenic and evolutionarily early branching, with larger genome sizes, higher rates of genetic variation, fewer molecular scars and distinct CRISPR-Cas systems relative to M. tuberculosis. Despite the differences, all tuberculosis-causing mycobacteria share a highly conserved core genome. Mouse infection experiments showed that STB strains are less persistent and virulent than M. tuberculosis. We conclude that M. tuberculosis emerged from an ancestral STB-like pool of mycobacteria by gain of persistence and virulence mechanisms, and we provide insights into the molecular events involved.


Assuntos
Adaptação Biológica/genética , Adaptação Biológica/imunologia , Evolução Molecular , Variação Genética , Genoma Bacteriano/genética , Mycobacterium tuberculosis/genética , Filogenia , Adaptação Biológica/fisiologia , Animais , Sequência de Bases , Análise por Conglomerados , Genômica , Sequências Repetidas Invertidas/genética , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mycobacterium tuberculosis/classificação , Mycobacterium tuberculosis/patogenicidade , Polimorfismo de Nucleotídeo Único/genética , Análise de Sequência de DNA , Especificidade da Espécie , Baço/virologia , Virulência
20.
PLoS One ; 6(11): e27713, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110736

RESUMO

PE are peculiar exported mycobacterial proteins over-represented in pathogenic mycobacterial species. They are characterized by an N-terminal domain of about 110 amino acids (PE domain) which has been demonstrated to be responsible for their export and localization. In this paper, we characterize the PE domain of PE_PGRS33 (PE(Rv1818c)), one of the best characterized PE proteins. We constructed several mutated proteins in which portions of the PE domain were deleted or subjected to defined mutations. These proteins were expressed in different mycobacterial species and their localization was characterized. We confirmed that the PE domain is essential for PE_PGRS33 surface localization, and demonstrated that a PE domain lacking its first 30 amino acids loses its function. However, single amino acid substitutions in two regions extremely well conserved within the N-terminal domain of all PE proteins had some effect on the stability of PE_PGRS33, but not on its localization. Using Mycobacterium marinum we could show that the type VII secretion system ESX-5 is essential for PE_PGRS33 export. Moreover, in M. marinum, but not in Mycobacterium bovis BCG and in Mycobacterium tuberculosis, the PE domain of PE_PGRS33 is processed and secreted into the culture medium when expressed in the absence of the PGRS domain. Finally, using chimeric proteins in which different portions of the PE(Rv1818c) domain were fused to the N-terminus of the green fluorescent protein, we could hypothesize that the first 30 amino acids of the PE domain contain a sequence that allows protein translocation.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Mycobacterium/citologia , Mycobacterium/metabolismo , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Espaço Intracelular/metabolismo , Mutação , Estabilidade Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA