Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
PLoS Pathog ; 19(7): e1011437, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37450466

RESUMEN

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


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Humanos , Virulencia/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mutación , Tuberculosis/microbiología , Mamíferos
2.
PLoS Pathog ; 17(11): e1010020, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34724002

RESUMEN

Mycobacterium tuberculosis, the main causative agent of human tuberculosis, is transmitted from person to person via small droplets containing very few bacteria. Optimizing the chance to seed in the lungs is therefore a major adaptation to favor survival and dissemination in the human population. Here we used TnSeq to identify genes important for the early events leading to bacterial seeding in the lungs. Beside several genes encoding known virulence factors, we found three new candidates not previously described: rv0180c, rv1779c and rv1592c. We focused on the gene, rv0180c, of unknown function. First, we found that deletion of rv0180c in M. tuberculosis substantially reduced the initiation of infection in the lungs of mice. Next, we established that Rv0180c enhances entry into macrophages through the use of complement-receptor 3 (CR3), a major phagocytic receptor for M. tuberculosis. Silencing CR3 or blocking the CR3 lectin site abolished the difference in entry between the wild-type parental strain and the Δrv0180c::km mutant. However, we detected no difference in the production of both CR3-known carbohydrate ligands (glucan, arabinomannan, mannan), CR3-modulating lipids (phthiocerol dimycocerosate), or proteins in the capsule of the Δrv0180c::km mutant in comparison to the wild-type or complemented strains. By contrast, we established that Rv0180c contributes to the functionality of the bacterial cell envelope regarding resistance to toxic molecule attack and cell shape. This alteration of bacterial shape could impair the engagement of membrane receptors that M. tuberculosis uses to invade host cells, and open a new perspective on the modulation of bacterial infectivity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Forma de la Célula , Pared Celular/química , Macrófagos/microbiología , Metaloproteinasas de la Matriz/metabolismo , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Animales , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Femenino , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Macrófagos/metabolismo , Macrófagos/patología , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Polisacáridos/metabolismo , Tuberculosis/metabolismo , Tuberculosis/patología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
3.
BMC Biol ; 20(1): 147, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729566

RESUMEN

BACKGROUND: Type I polyketide synthases (PKSs) are multifunctional enzymes responsible for the biosynthesis of a group of diverse natural compounds with biotechnological and pharmaceutical interest called polyketides. The diversity of polyketides is impressive despite the limited set of catalytic domains used by PKSs for biosynthesis, leading to considerable interest in deciphering their structure-function relationships, which is challenging due to high intrinsic flexibility. Among nineteen polyketide synthases encoded by the genome of Mycobacterium tuberculosis, Pks13 is the condensase required for the final condensation step of two long acyl chains in the biosynthetic pathway of mycolic acids, essential components of the cell envelope of Corynebacterineae species. It has been validated as a promising druggable target and knowledge of its structure is essential to speed up drug discovery to fight against tuberculosis. RESULTS: We report here a quasi-atomic model of Pks13 obtained using small-angle X-ray scattering of the entire protein and various molecular subspecies combined with known high-resolution structures of Pks13 domains or structural homologues. As a comparison, the low-resolution structures of two other mycobacterial polyketide synthases, Mas and PpsA from Mycobacterium bovis BCG, are also presented. This study highlights a monomeric and elongated state of the enzyme with the apo- and holo-forms being identical at the resolution probed. Catalytic domains are segregated into two parts, which correspond to the condensation reaction per se and to the release of the product, a pivot for the enzyme flexibility being at the interface. The two acyl carrier protein domains are found at opposite sides of the ketosynthase domain and display distinct characteristics in terms of flexibility. CONCLUSIONS: The Pks13 model reported here provides the first structural information on the molecular mechanism of this complex enzyme and opens up new perspectives to develop inhibitors that target the interactions with its enzymatic partners or between catalytic domains within Pks13 itself.


Asunto(s)
Mycobacterium tuberculosis , Policétidos , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/genética , Ácidos Micólicos/química , Ácidos Micólicos/metabolismo , Sintasas Poliquetidas/química , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Policétidos/metabolismo
4.
Cell Microbiol ; 19(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28095608

RESUMEN

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


Asunto(s)
Antígenos Bacterianos/metabolismo , Apoptosis/fisiología , Proteínas Bacterianas/metabolismo , Lípidos/fisiología , Macrófagos/metabolismo , Mycobacterium bovis/patogenicidad , Mycobacterium tuberculosis/patogenicidad , Fagosomas/metabolismo , Línea Celular Tumoral , Membrana Celular/patología , Humanos , Macrófagos/microbiología , Fagosomas/microbiología , Células THP-1 , Factores de Virulencia
5.
Proc Natl Acad Sci U S A ; 111(31): 11491-6, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049399

RESUMEN

Although the bovine tuberculosis (TB) agent, Mycobacterium bovis, may infect humans and cause disease, long-term epidemiological data indicate that humans represent a spill-over host in which infection with M. bovis is not self-maintaining. Indeed, human-to-human transmission of M. bovis strains and other members of the animal lineage of the tubercle bacilli is very rare. Here, we report on three mutations affecting the two-component virulence regulation system PhoP/PhoR (PhoPR) in M. bovis and in the closely linked Mycobacterium africanum lineage 6 (L6) that likely account for this discrepancy. Genetic transfer of these mutations into the human TB agent, Mycobacterium tuberculosis, resulted in down-regulation of the PhoP regulon, with loss of biologically active lipids, reduced secretion of the 6-kDa early antigenic target (ESAT-6), and lower virulence. Remarkably, the deleterious effects of the phoPR mutations were partly compensated by a deletion, specific to the animal-adapted and M. africanum L6 lineages, that restores ESAT-6 secretion by a PhoPR-independent mechanism. Similarly, we also observed that insertion of an IS6110 element upstream of the phoPR locus may completely revert the phoPR-bovis-associated fitness loss, which is the case for an exceptional M. bovis human outbreak strain from Spain. Our findings ultimately explain the long-term epidemiological data, suggesting that M. bovis and related phoPR-mutated strains pose a lower risk for progression to overt human TB, with major impact on the evolutionary history of TB.


Asunto(s)
Proteínas Bacterianas/genética , Evolución Biológica , Mutación/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Alelos , Animales , Antígenos Bacterianos , Proteínas Bacterianas/metabolismo , Bovinos , Secuencia Conservada/genética , Eliminación de Gen , Interacciones Huésped-Patógeno , Humanos , Mutagénesis Insercional , Mycobacterium/genética , Mycobacterium bovis/genética , Mycobacterium bovis/patogenicidad , Filogenia , Polimorfismo de Nucleótido Simple/genética , Tuberculosis/genética , Virulencia/genética
6.
PLoS Pathog ; 10(5): e1004183, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24874799

RESUMEN

The PhoPR two-component system is essential for virulence in Mycobacterium tuberculosis where it controls expression of approximately 2% of the genes, including those for the ESX-1 secretion apparatus, a major virulence determinant. Mutations in phoP lead to compromised production of pathogen-specific cell wall components and attenuation both ex vivo and in vivo. Using antibodies against the native protein in ChIP-seq experiments (chromatin immunoprecipitation followed by high-throughput sequencing) we demonstrated that PhoP binds to at least 35 loci on the M. tuberculosis genome. The PhoP regulon comprises several transcriptional regulators as well as genes for polyketide synthases and PE/PPE proteins. Integration of ChIP-seq results with high-resolution transcriptomic analysis (RNA-seq) revealed that PhoP controls 30 genes directly, whilst regulatory cascades are responsible for signal amplification and downstream effects through proteins like EspR, which controls Esx1 function, via regulation of the espACD operon. The most prominent site of PhoP regulation was located in the intergenic region between rv2395 and PE_PGRS41, where the mcr7 gene codes for a small non-coding RNA (ncRNA). Northern blot experiments confirmed the absence of Mcr7 in an M. tuberculosis phoP mutant as well as low-level expression of the ncRNA in M. tuberculosis complex members other than M. tuberculosis. By means of genetic and proteomic analyses we demonstrated that Mcr7 modulates translation of the tatC mRNA thereby impacting the activity of the Twin Arginine Translocation (Tat) protein secretion apparatus. As a result, secretion of the immunodominant Ag85 complex and the beta-lactamase BlaC is affected, among others. Mcr7, the first ncRNA of M. tuberculosis whose function has been established, therefore represents a missing link between the PhoPR two-component system and the downstream functions necessary for successful infection of the host.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Transporte de Membrana/biosíntesis , Mycobacterium tuberculosis/metabolismo , ARN no Traducido/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Productos del Gen tat/metabolismo , Humanos , Ratones Endogámicos C57BL , Mutación/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Operón/genética , Proteómica/métodos , ARN no Traducido/genética , Virulencia , beta-Lactamasas/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(16): 6560-5, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23550160

RESUMEN

A posttranslational protein O-mannosylation process resembling that found in fungi and animals has been reported in the major human pathogen Mycobacterium tuberculosis (Mtb) and related actinobacteria. However, the role and incidence of this process, which is essential in eukaryotes, have never been explored in Mtb. We thus analyzed the impact of interrupting O-mannosylation in the nonpathogenic saprophyte Mycobacterium smegmatis and in the human pathogen Mtb by inactivating the respective putative protein mannosyl transferase genes Msmeg_5447 and Rv1002c. Loss of protein O-mannosylation in both mutant strains was unambiguously demonstrated by efficient mass spectrometry-based glycoproteomics analysis. Unexpectedly, although the M. smegmatis phenotype was unaffected by the lack of manno-proteins, the Mtb mutant had severely impacted growth in vitro and in cellulo associated with a strong attenuation of its pathogenicity in immunocompromised mice. These data are unique in providing evidence of the biological significance of protein O-mannosylation in mycobacteria and demonstrate the crucial contribution of this protein posttranslational modification to Mtb virulence in the host.


Asunto(s)
Manosa/metabolismo , Manosiltransferasas/metabolismo , Mycobacterium smegmatis/enzimología , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/patogenicidad , Procesamiento Proteico-Postraduccional/fisiología , Animales , Silenciador del Gen , Manosiltransferasas/genética , Espectrometría de Masas , Ratones , Mycobacterium tuberculosis/crecimiento & desarrollo , Proteómica/métodos , Especificidad de la Especie , Virulencia
8.
Cell Microbiol ; 16(2): 195-213, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24028583

RESUMEN

Several specific lipids of the cell envelope are implicated in the pathogenesis of M. tuberculosis (Mtb), including phthiocerol dimycocerosates (DIM) that have clearly been identified as virulence factors. Others, such as trehalose-derived lipids, sulfolipids (SL), diacyltrehaloses (DAT) and polyacyltrehaloses (PAT), are believed to be essential for Mtb virulence, but the details of their role remain unclear. We therefore investigated the respective contribution of DIM, DAT/PAT and SL to tuberculosis by studying a collection of mutants, each with impaired production of one or several lipids. We confirmed that among those with a single lipid deficiency, only strains lacking DIM were affected in their replication in lungs and spleen of mice in comparison to the WT Mtb strain. We found also that the additional loss of DAT/PAT, and to a lesser extent of SL, increased the attenuated phenotype of the DIM-less mutant. Importantly, the loss of DAT/PAT and SL in a DIM-less background also affected Mtb growth in human monocyte-derived macrophages (hMDMs). Fluorescence microscopy revealed that mutants lacking DIM or DAT/PAT were localized in an acid compartment and that bafilomycin A1, an inhibitor of phagosome acidification, rescued the growth defect of these mutants. These findings provide evidence for DIM being dominant virulence factors that mask the functions of lipids of other families, notably DAT/PAT and to a lesser extent of SL, which we showed for the first time to contribute to Mtb virulence.


Asunto(s)
Interacciones Huésped-Patógeno , Metabolismo de los Lípidos , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/fisiología , Sintasas Poliquetidas/metabolismo , Tuberculosis/microbiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Sintasas Poliquetidas/genética , Bazo/microbiología , Virulencia
9.
Infect Immun ; 82(2): 476-90, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478064

RESUMEN

During the dormant phase of tuberculosis, Mycobacterium tuberculosis persists in lung granulomas by residing in foamy macrophages (FM) that contain abundant lipid bodies (LB) in their cytoplasm, allowing bacilli to accumulate lipids as intracytoplasmic lipid inclusions (ILI). An experimental model of FM is presented where bone marrow-derived mouse macrophages are infected with M. avium and exposed to very-low-density lipoprotein (VLDL) as a lipid source. Quantitative analysis of detailed electron microscope observations showed the following results. (i) Macrophages became foamy, and mycobacteria formed ILI, for which host triacylglycerides, rather than cholesterol, was essential. (ii) Lipid transfer occurred via mycobacterium-induced fusion between LB and phagosomes. (iii) Mycobacteria showed a thinned cell wall and became elongated but did not divide. (iv) Upon removal of VLDL, LB and ILI declined within hours, and simultaneous resumption of mycobacterial division restored the number of mycobacteria to the same level as that found in untreated control macrophages. This showed that the presence of ILI resulted in a reversible block of division without causing a change in the mycobacterial replication rate. Fluctuation between ILI either partially or fully extending throughout the mycobacterial cytoplasm was suggestive of bacterial cell cycle events. We propose that VLDL-driven FM constitute a well-defined cellular system in which to study changed metabolic states of intracellular mycobacteria that may relate to persistence and reactivation of tuberculosis.


Asunto(s)
Metabolismo de los Lípidos , Lipoproteínas VLDL/metabolismo , Macrófagos/microbiología , Mycobacterium avium/crecimiento & desarrollo , Mycobacterium avium/metabolismo , Animales , División Celular , Células Cultivadas , Femenino , Cuerpos de Inclusión/microbiología , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mycobacterium avium/ultraestructura
10.
J Biol Chem ; 287(40): 33675-90, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22825853

RESUMEN

Pks13 is a type I polyketide synthase involved in the final biosynthesis step of mycolic acids, virulence factors, and essential components of the Mycobacterium tuberculosis envelope. Here, we report the biochemical and structural characterization of a 52-kDa fragment containing the acyltransferase domain of Pks13. This fragment retains the ability to load atypical extender units, unusually long chain acyl-CoA with a predilection for carboxylated substrates. High resolution crystal structures were determined for the apo, palmitoylated, and carboxypalmitoylated forms. Structural conservation with type I polyketide synthases and related fatty-acid synthases also extends to the interdomain connections. Subtle changes could be identified both in the active site and in the upstream and downstream linkers in line with the organization displayed by this singular polyketide synthase. More importantly, the crystallographic analysis illustrated for the first time how a long saturated chain can fit in the core structure of an acyltransferase domain through a dedicated channel. The structures also revealed the unexpected binding of a 12-mer peptide that might provide insight into domain-domain interaction.


Asunto(s)
Proteínas Bacterianas/química , Sintasas Poliquetidas/química , Secuencia de Aminoácidos , Antibacterianos/síntesis química , Proteínas Bacterianas/metabolismo , Unión Competitiva , Dominio Catalítico , Química Farmacéutica/métodos , Clonación Molecular , Cristalografía por Rayos X/métodos , Diseño de Fármacos , Ligandos , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Mycobacterium tuberculosis/metabolismo , Ácidos Micólicos/metabolismo , Sintasas Poliquetidas/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
11.
PLoS Pathog ; 6(10): e1001159, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20975946

RESUMEN

The species-specific phenolic glycolipid 1 (PGL-1) is suspected to play a critical role in the pathogenesis of leprosy, a chronic disease of the skin and peripheral nerves caused by Mycobacterium leprae. Based on studies using the purified compound, PGL-1 was proposed to mediate the tropism of M. leprae for the nervous system and to modulate host immune responses. However, deciphering the biological function of this glycolipid has been hampered by the inability to grow M. leprae in vitro and to genetically engineer this bacterium. Here, we identified the M. leprae genes required for the biosynthesis of the species-specific saccharidic domain of PGL-1 and reprogrammed seven enzymatic steps in M. bovis BCG to make it synthesize and display PGL-1 in the context of an M. leprae-like cell envelope. This recombinant strain provides us with a unique tool to address the key questions of the contribution of PGL-1 in the infection process and to study the underlying molecular mechanisms. We found that PGL-1 production endowed recombinant BCG with an increased capacity to exploit complement receptor 3 (CR3) for efficient invasion of human macrophages and evasion of inflammatory responses. PGL-1 production also promoted bacterial uptake by human dendritic cells and dampened their infection-induced maturation. Our results therefore suggest that M. leprae produces PGL-1 for immune-silent invasion of host phagocytic cells.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/fisiología , Glucolípidos/genética , Glucolípidos/fisiología , Mycobacterium bovis/genética , Fagocitos/inmunología , Fagocitos/metabolismo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/fisiología , Antígenos Bacterianos/metabolismo , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Glucolípidos/metabolismo , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Modelos Biológicos , Mycobacterium bovis/metabolismo , Mycobacterium leprae/genética , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Tiempo
12.
PLoS Pathog ; 5(2): e1000289, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19197369

RESUMEN

Phthiocerol dimycocerosates (DIM) are major virulence factors of Mycobacterium tuberculosis (Mtb), in particular during the early step of infection when bacilli encounter their host macrophages. However, their cellular and molecular mechanisms of action remain unknown. Using Mtb mutants deleted for genes involved in DIM biosynthesis, we demonstrated that DIM participate both in the receptor-dependent phagocytosis of Mtb and the prevention of phagosomal acidification. The effects of DIM required a state of the membrane fluidity as demonstrated by experiments conducted with cholesterol-depleting drugs that abolished the differences in phagocytosis efficiency and phagosome acidification observed between wild-type and mutant strains. The insertion of a new cholesterol-pyrene probe in living cells demonstrated that the polarity of the membrane hydrophobic core changed upon contact with Mtb whereas the lateral diffusion of cholesterol was unaffected. This effect was dependent on DIM and was consistent with the effect observed following DIM insertion in model membrane. Therefore, we propose that DIM control the invasion of macrophages by Mtb by targeting lipid organisation in the host membrane, thereby modifying its biophysical properties. The DIM-induced changes in lipid ordering favour the efficiency of receptor-mediated phagocytosis of Mtb and contribute to the control of phagosomal pH driving bacilli in a protective niche.


Asunto(s)
Membrana Celular/metabolismo , Lípidos/fisiología , Macrófagos/metabolismo , Lípidos de la Membrana/metabolismo , Mycobacterium tuberculosis/metabolismo , Membrana Celular/microbiología , Colesterol/metabolismo , Técnicas de Inactivación de Genes , Humanos , Concentración de Iones de Hidrógeno , Luz , Lípidos/genética , Macrófagos/microbiología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Fagocitosis , Fagosomas/metabolismo , Fagosomas/microbiología , Dispersión de Radiación , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
13.
Nat Microbiol ; 6(8): 1082-1093, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34294904

RESUMEN

Pathogenomic evidence suggests that Mycobacterium tuberculosis (MTB) evolved from an environmental ancestor similar to Mycobacterium canettii, a rare human pathogen. Although the adaptations responsible for this transition are poorly characterized, the ability to persist in humans seems to be important. We set out to identify the adaptations contributing to the evolution of persistence in MTB. We performed an experimental evolution of eight M. canettii populations in mice; four populations were derived from the isolate STB-K (phylogenomically furthest from MTB) and four from STB-D (closest to MTB), which were monitored for 15 and 6 cycles, respectively. We selected M. canettii mutants with enhanced persistence in vivo compared with the parental strains, which were phenotypically closer to MTB. Genome sequencing of 140 mutants and complementation analysis revealed that mutations in two loci were responsible for enhanced persistence. Most of the tested mutants were more resistant than their parental strains to nitric oxide, an important effector of immunity. Modern MTB were similarly more resistant to nitric oxide than M. canettii. Our findings demonstrate phenotypic convergence during experimental evolution of M. canettii, which mirrors natural evolution of MTB. Furthermore, they indicate that the ability to withstand host-induced stresses was key for the emergence of persistent MTB.


Asunto(s)
Evolución Biológica , Mycobacterium tuberculosis/fisiología , Mycobacterium/fisiología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Mutación , Mycobacterium/genética , Mycobacterium tuberculosis/genética , Estrés Fisiológico , Tuberculosis/microbiología
14.
Elife ; 102021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-34951591

RESUMEN

Successful control of Mycobacterium tuberculosis (Mtb) infection by macrophages relies on immunometabolic reprogramming, where the role of fatty acids (FAs) remains poorly understood. Recent studies unraveled Mtb's capacity to acquire saturated and monounsaturated FAs via the Mce1 importer. However, upon activation, macrophages produce polyunsaturated fatty acids (PUFAs), mammal-specific FAs mediating the generation of immunomodulatory eicosanoids. Here, we asked how Mtb modulates de novo synthesis of PUFAs in primary mouse macrophages and whether this benefits host or pathogen. Quantitative lipidomics revealed that Mtb infection selectively activates the biosynthesis of ω6 PUFAs upstream of the eicosanoid precursor arachidonic acid (AA) via transcriptional activation of Fads2. Inhibiting FADS2 in infected macrophages impaired their inflammatory and antimicrobial responses but had no effect on Mtb growth in host cells nor mice. Using a click-chemistry approach, we found that Mtb efficiently imports ω6 PUFAs via Mce1 in axenic culture, including AA. Further, Mtb preferentially internalized AA over all other FAs within infected macrophages by mechanisms partially depending on Mce1 and supporting intracellular persistence. Notably, IFNγ repressed de novo synthesis of AA by infected mouse macrophages and restricted AA import by intracellular Mtb. Together, these findings identify AA as a major FA substrate for intracellular Mtb, whose mobilization by innate immune responses is opportunistically hijacked by the pathogen and downregulated by IFNγ.


Asunto(s)
Ácidos Grasos Insaturados/farmacología , Factores Inmunológicos/farmacología , Mycobacterium tuberculosis/fisiología , Animales , Línea Celular , Ácidos Grasos Insaturados/metabolismo , Femenino , Humanos , Inmunidad Innata , Factores Inmunológicos/metabolismo , Masculino , Ratones , Mycobacterium tuberculosis/metabolismo , Nutrientes/metabolismo
15.
J Biol Chem ; 284(40): 27101-13, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19648652

RESUMEN

The Mycobacterium tuberculosis Beijing strains are a family highly prevalent in Asia and have recently spread worldwide, causing a number of epidemics, suggesting that they express virulence factors not found in other M. tuberculosis strains. Accordingly, we looked for putative characteristic compounds by comparing the lipid profiles of several Beijing and non-Beijing strains. All the Beijing strains analyzed were found to synthesize structural variants of two well known characteristic lipids of the tubercle bacillus, namely phthiocerol dimycocerosates (DIM) and eventually phenolglycolipids (PGL). These variants were not found in non-Beijing M. tuberculosis isolates. Structural elucidation of these variants showed that they consist of phthiotriol and glycosylated phenolphthiotriol dimycocerosates, eventually acylated with 1 mol of palmitic acid, in addition to the conventional acylation of the beta-diol by mycocerosic acids. We demonstrated that this unusual lipid profile resulted from a single point mutation in the Rv2952 gene, which encodes the S-adenosylmethionine-dependent methyltransferase participating to the O-methylation of the third hydroxyl of the phthiotriol and phenolphthiotriol in the biosynthetic pathway of DIM and PGL. Consistently, the mutated enzyme exhibited in vitro a much lower O-methyltransferase activity than did the wild-type Rv2952. We finally demonstrated that the structural variants of DIM and PGL fulfill the same function in the cell envelope and virulence than their conventional counterparts.


Asunto(s)
Glucolípidos/química , Glucolípidos/metabolismo , Lípidos/química , Mutación , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Genes Bacterianos/genética , Genotipo , Glucolípidos/biosíntesis , Lípidos/biosíntesis , Metiltransferasas/genética , Metiltransferasas/metabolismo , Mycobacterium tuberculosis/citología , Mycobacterium tuberculosis/patogenicidad , Mutación Puntual , S-Adenosilmetionina/metabolismo , Especificidad de la Especie , Virulencia
16.
Tuberculosis (Edinb) ; 123: 101956, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32741533

RESUMEN

The ability to perform genetic manipulation of mycobacteria is important for characterization of gene function. Homologous recombination-based protocols are frequently used for reverse genetics studies with mycobacteria. It is known that Mycobacteriumbovis BCG Russia, closely related to M. bovis BCG Moreau, is a natural recA deficient strain and is non-permissive to homologous recombination assays. In this work we show that M. bovis BCG Moreau is also deficient in homologous recombination, shown by a specialized transduction assay, but this phenotype can be reverted by complementation with heterologous recombinases, using a recombineering protocol. Sequence analysis of the genes known to be involved in homologous recombination annotated in the genome of BCG Moreau detected no differences compared to the genome of BCG Pasteur. Further studies are needed in order to determine the exact mechanism underlying this deficiency in BCG Moreau.


Asunto(s)
Proteínas Bacterianas/genética , Recombinación Homóloga , Mycobacterium bovis/genética , Rec A Recombinasas/genética , Proteínas Bacterianas/metabolismo , Genotipo , Mycobacterium bovis/enzimología , Fenotipo , Rec A Recombinasas/metabolismo
17.
Res Microbiol ; 171(5-6): 203-210, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32283218

RESUMEN

Targeted gene disruption by homologous recombination, has been widely used in mycobacterium species to understand the genetic basis of virulence and persistence in the host and to develop efficacious potential live vaccines. However, in slow growing pathogenic mycobacteria as Mycobacterium avium subsp paratuberculosis (MAP), these methods have been inefficient, in part due to the low frequency of legitimate homologous recombination. Another feature of mycobacteria is the low efficiency of transformation; therefore, some years ago, a phage-mediated transduction process was developed to introduce DNA into mycobacteria. This strategy is very efficient, due to the high rate of infection of the phage. This report describes a genetic method for the generation of targeted deletion mutations in MAP by allelic exchange using in vitro-generated specialized transducing mycobacteriophages, which does not require the critical packaging step and that could also be applied to other mycobacteria. We provide a detailed gene deletion methodology and demonstrate the use of this genetic system by deleting the mce4 operon of MAP. Finally, our results showed that the deletion of mce4 in MAP induces triacylglycerol accumulation; alter morphology and aggregation in liquid culture.


Asunto(s)
Eliminación de Gen , Recombinación Homóloga , Micobacteriófagos/genética , Mycobacterium avium subsp. paratuberculosis/genética , Alelos , Proteínas Bacterianas/genética , Genes Bacterianos , Técnicas Genéticas , Mutación , Mycobacterium avium subsp. paratuberculosis/citología , Operón , Transducción Genética , Triglicéridos/metabolismo
18.
Front Microbiol ; 11: 624121, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33510737

RESUMEN

Tuberculosis is a world widespread disease, caused by Mycobacterium tuberculosis (M.tb). Although considered an obligate aerobe, this organism can resist life-limiting conditions such as microaerophily mainly due to its set of enzymes responsible for energy production and coenzyme restoration under these conditions. One of these enzymes is fumarate reductase, an heterotetrameric complex composed of a catalytic (FrdA), an iron-sulfur cluster (FrdB) and two transmembrane (FrdC and FrdD) subunits involved in anaerobic respiration and important for the maintenance of membrane potential. In this work, aiming to further characterize this enzyme function in mycobacteria, we analyzed the expression of FrdB-containing proteins in M.tb and Mycobacterium bovis Bacillus Calmette-Guérin (BCG) Moreau, the Brazilian vaccine strain against tuberculosis. We identified three isoforms in both mycobacteria, two of them corresponding to the predicted encoded polypeptides of M.tb (27 kDa) and BCG Moreau (40 kDa) frd sequences, as due to an insertion on the latter's operon a fused FrdBC protein is expected. The third 52 kDa band can be explained by a transcriptional slippage event, typically occurring when mutation arises in a repetitive region within a coding sequence, thought to reduce its impact allowing the production of both native and variant forms. Comparative modeling of the M.tb and BCG Moreau predicted protein complexes allowed the detection of subtle overall differences, showing a high degree of structure and maybe functional resemblance among them. Axenic growth and macrophage infection assays show that the frd locus is important for proper bacterial development in both scenarios, and that both M.tb's and BCG Moreau's alleles can partially revert the hampered phenotype of the knockout strain. Altogether, our results show that the frdABCD operon of Mycobacteria may have evolved to possess other yet non-described functions, such as those necessary during aerobic logarithmic growth and early stage steps of infection.

19.
Vaccine ; 38(6): 1416-1423, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31862194

RESUMEN

Tuberculosis still claims more lives than any other pathogen, and a vaccine better than BCG is urgently needed. One of the challenges for novel TB vaccines is to protect against all Mycobacterium tuberculosis lineages, including the most virulent ones, such as the Beijing lineage. Here we developed a live attenuated M. tuberculosis mutant derived from GC1237, a Beijing strain responsible for tuberculosis outbreaks in the Canary Islands. The mutant strain is inactivated both in the Rv1503c gene, responsible for surface glycolipid synthesis, and in the two-component global regulator PhoPR. This double mutant is as safe as BCG in immunodeficient SCID mice. In immune-competent mice and guinea pigs, the mutant is as protective as BCG against M. tuberculosis strains of common lineage 4 (Euro-American). By contrast, in mice the vaccine is protective against a M. tuberculosis strain of lineage 2 (East-Asian, Beijing), while BCG is not. These results highlight differences in protection efficacy of live attenuated M. tuberculosis-derived vaccine candidates depending on their genetic background, and provide insights for the development of novel live vaccines against TB, especially in East-Asian countries where M. tuberculosis strains of the Beijing family are highly dominant.


Asunto(s)
Vacunas contra la Tuberculosis/inmunología , Tuberculosis , Animales , Vacuna BCG , Cobayas , Ratones , Ratones SCID , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunas Atenuadas/inmunología
20.
J Bacteriol ; 191(8): 2613-21, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19181796

RESUMEN

Lipooligosaccharides (LOS) are highly antigenic glycolipids produced by a number of Mycobacterium species, which include "M. canettii," a member of the M. tuberculosis complex, and the opportunistic pathogens M. marinum and M. kansasii. The various LOS share a core composed of trehalose esterified by at least 1 mole of polymethyl-branched fatty acid (PMB-FA) and differ from one another by their oligosaccharide extensions. In this study, we identified a cluster of genes, MSMEG_4727 through MSMEG_4741, likely involved in the synthesis of LOS in M. smegmatis. Disruption of MSMEG_4727 (the ortholog of pks5 of M. tuberculosis), which encodes a putative polyketide synthase, resulted in the concomitant abrogation of the production of both PMB-FA and LOS in the mutant strain. Complementation of the mutant with the wild-type gene fully restored the phenotype. We also showed that, in contrast to the case for "M. canettii" and M. marinum, LOS are located in deeper compartments of the cell envelope of M. smegmatis. The availability of two mycobacterial strains differing only in LOS production should help in defining the biological role(s) of this important glycolipid.


Asunto(s)
Lipopolisacáridos/biosíntesis , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/metabolismo , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Cromatografía en Capa Delgada , Cromatografía de Gases y Espectrometría de Masas , Eliminación de Gen , Orden Génico , Genes Bacterianos , Prueba de Complementación Genética , Redes y Vías Metabólicas/genética , Familia de Multigenes , Mutagénesis Insercional , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA