Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Proc Natl Acad Sci U S A ; 114(16): 4231-4236, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28373551

ABSTRACT

The outer membranes (OMs) of members of the Corynebacteriales bacterial order, also called mycomembranes, harbor mycolic acids and unusual outer membrane proteins (OMPs), including those with α-helical structure. The signals that allow precursors of such proteins to be targeted to the mycomembrane remain uncharacterized. We report here the molecular features responsible for OMP targeting to the mycomembrane of Corynebacterium glutamicum, a nonpathogenic member of the Corynebacteriales order. To better understand the mechanisms by which OMP precursors were sorted in C. glutamicum, we first investigated the partitioning of endogenous and recombinant PorA, PorH, PorB, and PorC between bacterial compartments and showed that they were both imported into the mycomembrane and secreted into the extracellular medium. A detailed investigation of cell extracts and purified proteins by top-down MS, NMR spectroscopy, and site-directed mutagenesis revealed specific and well-conserved posttranslational modifications (PTMs), including O-mycoloylation, pyroglutamylation, and N-formylation, for mycomembrane-associated and -secreted OMPs. PTM site sequence analysis from C. glutamicum OMP and other O-acylated proteins in bacteria and eukaryotes revealed specific patterns. Furthermore, we found that such modifications were essential for targeting to the mycomembrane and sufficient for OMP assembly into mycolic acid-containing lipid bilayers. Collectively, it seems that these PTMs have evolved in the Corynebacteriales order and beyond to guide membrane proteins toward a specific cell compartment.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Cell Wall/metabolism , Corynebacterium glutamicum/metabolism , Lipid Bilayers/metabolism , Mycolic Acids/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Corynebacterium glutamicum/growth & development , Protein Transport , Sequence Homology
2.
J Biol Chem ; 293(14): 5172-5184, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29472294

ABSTRACT

Mycolic acids are the hallmark of the cell envelope in mycobacteria, which include the important human pathogens Mycobacterium tuberculosis and Mycobacterium leprae Mycolic acids are very long C60-C90 α-alkyl ß-hydroxy fatty acids having a variety of functional groups on their hydrocarbon chain that define several mycolate types. Mycobacteria also produce an unusually large number of putative epoxide hydrolases, but the physiological functions of these enzymes are still unclear. Here, we report that the mycobacterial epoxide hydrolase EphD is involved in mycolic acid metabolism. We found that orthologs of EphD from M. tuberculosis and M. smegmatis are functional epoxide hydrolases, cleaving a lipophilic substrate, 9,10-cis-epoxystearic acid, in vitro and forming a vicinal diol. The results of EphD overproduction in M. smegmatis and M. bovis BCG Δhma strains producing epoxymycolic acids indicated that EphD is involved in the metabolism of these forms of mycolates in both fast- and slow-growing mycobacteria. Moreover, using MALDI-TOF-MS and 1H NMR spectroscopy of mycolic acids and lipids isolated from EphD-overproducing M. smegmatis, we identified new oxygenated mycolic acid species that accumulated during epoxymycolate depletion. Disruption of the ephD gene in M. tuberculosis specifically impaired the synthesis of ketomycolates and caused accumulation of their precursor, hydroxymycolate, indicating either direct or indirect involvement of EphD in ketomycolate biosynthesis. Our results clearly indicate that EphD plays a role in metabolism of oxygenated mycolic acids in mycobacteria.


Subject(s)
Epoxide Hydrolases/metabolism , Mycolic Acids/metabolism , Cell Wall/metabolism , Fatty Acids/metabolism , Lipid Metabolism/physiology , Lipids/physiology , Mass Spectrometry/methods , Mycobacterium/metabolism , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/metabolism
3.
Mol Microbiol ; 105(4): 525-539, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28558126

ABSTRACT

Mycobacteria have a complex cell wall structure that includes many lipids; however, even within a single subspecies of Mycobacterium avium these lipids can differ. Total lipids from an M. avium subsp. paratuberculosis (Map) ovine strain (S-type) contained no identifiable glycopeptidolipids or lipopentapeptide (L5P), yet both lipids are present in other M. avium subspecies. We determined the genetic and phenotypic basis for this difference using sequence analysis as well as biochemical and physico-chemical approaches. This strategy showed that a nonribosomal peptide synthase, encoded by mps1, contains three amino acid specifying modules in ovine strains, compared to five modules in bovine strains (C-type). Sequence analysis predicted these modules would produce the tripeptide Phe-N-Methyl-Val-Ala with a lipid moiety, termed lipotripeptide (L3P). Comprehensive physico-chemical analysis of Map S397 extracts confirmed the structural formula of the native L3P as D-Phe-N-Methyl-L-Val-L-Ala-OMe attached in N-ter to a 20-carbon fatty acid chain. These data demonstrate that S-type strains, which are more adapted in sheep, produce a unique lipid. There is a dose-dependent effect observed for L3P on upregulation of CD25+ CD8 T cells from infected cows, while L5P effects were static. In contrast, L5P demonstrated a significantly stronger induction of CD25+ B cells from infected animals compared to L3P.


Subject(s)
Cell Wall/genetics , Membrane Lipids/genetics , Peptide Synthases/genetics , Amino Acid Sequence , Cell Wall/metabolism , Cell Wall/physiology , Membrane Lipids/chemistry , Mycobacterium avium/genetics , Mycobacterium avium/metabolism , Peptides/genetics , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
4.
Indian J Microbiol ; 58(3): 393-396, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30013284

ABSTRACT

Treatment of tuberculosis still represent a major public health issue. The emergence of multi-and extensively-drug resistant (MDR and XDR) Mycobacterium tuberculosis clinical strains further pinpoint the urgent need for new anti-tuberculous drugs. We previously showed that vancomycin can target mycobacteria lacking cell wall integrity, especially those lacking related phthiocerol and phthiodolone dimycocerosates, PDIM A and PDIM B, respectively. As aloe emodin was previously hypothesized to be able to target the synthesis of mycobacterial cell wall lipids, we tested its ability to potentiate glycopeptides antimycobacterial activity. The aloe emodin with the vancomycin induced a combination effect beyond simple addition, close to synergism, at a concentration lower to reported IC50 cytotoxic value, on M. bovis BCG and on H37Rv M. tuberculosis. Interestingly, out of six MDR and pre-XDR clinical strains, one showed a strong synergic susceptibility to the drug combination. Mycobacterial cell wall lipid analyses highlighted a selective reduction of PDIM B by aloe emodin.

5.
Mol Microbiol ; 99(4): 794-807, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26538472

ABSTRACT

Understanding the molecular strategies used by Mycobacterium tuberculosis to invade and persist within the host is of paramount importance to tackle the tuberculosis pandemic. Comparative genomic surveys have revealed that hadC, encoding a subunit of the HadBC dehydratase, is mutated in the avirulent M. tuberculosis H37Ra strain. We show here that mutation or deletion of hadC affects the biosynthesis of oxygenated mycolic acids, substantially reducing their production level. Additionally, it causes the loss of atypical extra-long mycolic acids, demonstrating the involvement of HadBC in the late elongation steps of mycolic acid biosynthesis. These events have an impact on the morphotype, cording capacity and biofilm growth of the bacilli as well as on their sensitivity to agents such as rifampicin. Furthermore, deletion of hadC leads to a dramatic loss of virulence: an almost 4-log drop of the bacterial load in the lungs and spleens of infected immunodeficient mice. Both its unique function and importance for M. tuberculosis virulence make HadBC an attractive therapeutic target for tuberculosis drug development.


Subject(s)
Bacterial Proteins/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Mycolic Acids/chemistry , Tuberculosis/microbiology , Animals , Antitubercular Agents/pharmacology , Bacterial Load , Bacterial Proteins/metabolism , Biofilms/growth & development , Gene Deletion , Lung/microbiology , Mice , Mutation , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Spleen/microbiology , Virulence/genetics
6.
Proc Natl Acad Sci U S A ; 111(31): 11491-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049399

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Biological Evolution , Mutation/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Alleles , Animals , Antigens, Bacterial , Bacterial Proteins/metabolism , Cattle , Conserved Sequence/genetics , Gene Deletion , Host-Pathogen Interactions , Humans , Mutagenesis, Insertional , Mycobacterium/genetics , Mycobacterium bovis/genetics , Mycobacterium bovis/pathogenicity , Phylogeny , Polymorphism, Single Nucleotide/genetics , Tuberculosis/genetics , Virulence/genetics
7.
Antimicrob Agents Chemother ; 60(10): 6193-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27503643

ABSTRACT

Tuberculosis is still a cause of major concern, partly due to the emergence of multidrug-resistant strains. New drugs are therefore needed. Vancomycin can target mycobacteria with cell envelope deficiency. In this study, we used a vancomycin susceptibility assay to detect drugs hampering lipid synthesis in Mycobacterium bovis BCG and in Mycobacterium tuberculosis We tested three drugs already used to treat human obesity: tetrahydrolipstatin (THL), simvastatin, and fenofibrate. Only vancomycin and THL were able to synergize on M. bovis BCG and on M. tuberculosis, although mycobacteria could also be inhibited by simvastatin alone. Lipid analysis allowed us to identify several lipid modifications in M. tuberculosis H37Rv treated with those drugs. THL treatment mainly reduced the phthiocerol dimycocerosate (PDIM) content in the mycobacterial cell wall, providing an explanation for the synergy, since PDIM deficiency has been related to vancomycin susceptibility. Proteomic analysis suggested that bacteria treated with THL, in contrast to bacteria treated with simvastatin, tried to recover, inducing, among other reactions, lipid synthesis. The combination of THL and vancomycin should be considered a promising solution in developing new strategies to treat multidrug-resistant tuberculosis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Hypolipidemic Agents/pharmacology , Mycobacterium bovis/drug effects , Mycobacterium tuberculosis/drug effects , Vancomycin/pharmacology , Antitubercular Agents/pharmacology , Drug Synergism , Fenofibrate/pharmacology , Lactones/pharmacology , Membrane Lipids/metabolism , Microbial Sensitivity Tests , Mycobacterium tuberculosis/metabolism , Orlistat , Simvastatin/pharmacology
8.
J Bacteriol ; 197(24): 3797-811, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416833

ABSTRACT

UNLABELLED: Mycobacterium tuberculosis, the etiological agent of tuberculosis, is a Gram-positive bacterium with a unique cell envelope composed of an essential outer membrane. Mycolic acids, which are very-long-chain (up to C100) fatty acids, are the major components of this mycomembrane. The enzymatic pathways involved in the biosynthesis and transport of mycolates are fairly well documented and are the targets of the major antituberculous drugs. In contrast, only fragmented information is available on the expression and regulation of the biosynthesis genes. In this study, we report that the hadA, hadB, and hadC genes, which code for the mycolate biosynthesis dehydratase enzymes, are coexpressed with three genes that encode proteins of the translational apparatus. Consistent with the well-established control of the translation potential by nutrient availability, starvation leads to downregulation of the hadABC genes along with most of the genes required for the synthesis, modification, and transport of mycolates. The downregulation of a subset of the biosynthesis genes is partially dependent on RelMtb, the key enzyme of the stringent response. We also report the phylogenetic evolution scenario that has shaped the current genetic organization, characterized by the coregulation of the hadABC operon with genes of the translational apparatus and with genes required for the modification of the mycolates. IMPORTANCE: Mycobacterium tuberculosis infects one-third of the human population worldwide, and despite the available therapeutic arsenal, it continues to kill millions of people each year. There is therefore an urgent need to identify new targets and develop a better understanding of how the bacterium is adapting itself to host defenses during infection. A prerequisite of this understanding is knowledge of how this adaptive skill has been implanted by evolution. Nutrient scarcity is an environmental condition the bacterium has to cope with during infection. In many bacteria, adaptation to starvation relies partly on the stringent response. M. tuberculosis's unique outer membrane layer, the mycomembrane, is crucial for its viability and virulence. Despite its being the target of the major antituberculosis drugs, only scattered information exists on how the genes required for biosynthesis of the mycomembrane are expressed and regulated during starvation. This work has addressed this issue as a step toward the identification of new targets in the fight against M. tuberculosis.


Subject(s)
Gene Expression Regulation, Bacterial/genetics , Hydro-Lyases/genetics , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/physiology , Down-Regulation , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/genetics , Hydro-Lyases/biosynthesis , Mycobacterium tuberculosis/genetics , Protein Biosynthesis/genetics , Starvation
9.
Antimicrob Agents Chemother ; 59(8): 5057-60, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26033733

ABSTRACT

Mycobacterium tuberculosis is wrapped in complex waxes, impermeable to most antibiotics. Comparing Mycobacterium bovis BCG and M. tuberculosis mutants that lack phthiocerol dimycocerosates (PDIM) and/or phenolic glycolipids with wild-type strains, we observed that glycopeptides strongly inhibited PDIM-deprived mycobacteria. Vancomycin together with a drug targeting lipid synthesis inhibited multidrug-resistant (MDR) and extensively drug-resistant (XDR) clinical isolates. Our study puts glycopeptides in the pipeline of potential antituberculosis (TB) agents and might provide a new antimycobacterial drug-screening strategy.


Subject(s)
Antitubercular Agents/pharmacology , Glycopeptides/pharmacology , Mycobacterium bovis/drug effects , Mycobacterium tuberculosis/drug effects , Vancomycin/pharmacology , Cell Wall/chemistry , Cell Wall/drug effects , Drug Resistance, Multiple, Bacterial , Humans , Lipids/biosynthesis , Microbial Sensitivity Tests , Tuberculosis, Pulmonary/drug therapy
10.
IUBMB Life ; 67(2): 129-38, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25865678

ABSTRACT

Bacteria living in a surface-attached community that contains a heterogeneous population, coated with an extracellular matrix, and showing drug tolerance (biofilms) are often linked to chronic infections. In mycobacteria, the pellicle mode of growth has been equated to an in vitro biofilm and meets several of the criteria mentioned above, while tuberculosis infection presents a chronic (latent) phase of infection. As mycobacteria lack most genes required to control biofilm production by other microorganisms, we deleted or expressed from the hsp60 strong promoter the only known c-di-GMP phosphodiesterase (PDE) gene in Mycobacterium bovis BCG. We found changes in pellicle production, cellular protein profiles, lipid production, resistance to nitrosative stress and maintenance in lungs and spleens of immunocompetent BALB/mice. Our results show that pellicle production and capacity to remain within the host are linked in BCG.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/genetics , Bacterial Proteins/genetics , Mycobacterium bovis/physiology , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Animals , Bacterial Proteins/metabolism , Biofilms/growth & development , Female , Gene Expression Regulation, Bacterial , Glycolipids/metabolism , Host-Pathogen Interactions , Lung/microbiology , Male , Mice, Inbred BALB C , Mycobacterium bovis/pathogenicity , Spleen/microbiology , Tuberculosis/microbiology , Tuberculosis/veterinary
11.
J Biol Chem ; 287(46): 38434-41, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23002234

ABSTRACT

Isoxyl (ISO) and thiacetazone (TAC), two prodrugs once used in the clinical treatment of tuberculosis, have long been thought to abolish Mycobacterium tuberculosis (M. tuberculosis) growth through the inhibition of mycolic acid biosynthesis, but their respective targets in this pathway have remained elusive. Here we show that treating M. tuberculosis with ISO or TAC results in both cases in the accumulation of 3-hydroxy C(18), C(20), and C(22) fatty acids, suggestive of an inhibition of the dehydratase step of the fatty-acid synthase type II elongation cycle. Consistently, overexpression of the essential hadABC genes encoding the (3R)-hydroxyacyl-acyl carrier protein dehydratases resulted in more than a 16- and 80-fold increase in the resistance of M. tuberculosis to ISO and TAC, respectively. A missense mutation in the hadA gene of spontaneous ISO- and TAC-resistant mutants was sufficient to confer upon M. tuberculosis high level resistance to both drugs. Other mutations found in hypersusceptible or resistant M. tuberculosis and Mycobacterium kansasii isolates mapped to hadC. Mutations affecting the non-essential mycolic acid methyltransferases MmaA4 and MmaA2 were also found in M. tuberculosis spontaneous ISO- and TAC-resistant mutants. That MmaA4, at least, participates in the activation of the two prodrugs as proposed earlier is not supported by our biochemical evidence. Instead and in light of the known interactions of both MmaA4 and MmaA2 with HadAB and HadBC, we propose that mutations affecting these enzymes may impact the binding of ISO and TAC to the dehydratases.


Subject(s)
Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/metabolism , Mycolic Acids/antagonists & inhibitors , Phenylthiourea/analogs & derivatives , Thioacetazone/pharmacology , Alleles , Antitubercular Agents/pharmacology , Cell Wall/metabolism , Chromatography, Liquid/methods , Fatty Acid Synthases/metabolism , Gas Chromatography-Mass Spectrometry/methods , Genome, Bacterial , Lipids/chemistry , Mass Spectrometry/methods , Models, Chemical , Phenylthiourea/pharmacology , Recombinant Proteins/chemistry , Sequence Analysis, DNA , Time Factors
12.
Microbiology (Reading) ; 159(Pt 1): 191-203, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23154972

ABSTRACT

Mycolic acids, very long-chain α-alkyl, ß-hydroxylated fatty acids, occur in the members of the order Corynebacteriales where their chain lengths (C(26)-C(88)) and structural features (oxygen functions, cis or trans double bonds, cyclopropane rings and methyl branches) are genus- and species-specific. The molecular composition and structures of the mycolic acids of two species belonging to the genus Segniliparus were determined by a combination of modern analytical chemical techniques, which include MS and NMR. They consist of mono-ethylenic C(62-)C(64) (α'), di-ethylenic C(77)-C(79) (α) and extremely long-chain mycolic acids (α(+)) ranging from 92 to 98 carbon atoms and containing three unsaturations, cis and/or trans double bonds and/or cyclopropanes. The double bonds in each class of mycolic acids were positioned by oxidative cleavage and exhibit locations similar to those of α- and α'-mycolic acids of mycobacteria. For the ultralong chain α-mycolic acids, the three double bonds were located at equally spaced carbon intervals (C(13)-C(16)), with the methyl branches adjacent to the proximal and distal trans double bonds. Examination of the Segniliparus rotundus genome compared with those of other members of the Corynebacteriales indicated two obvious differences in genes encoding the elongation fatty acid (FAS-II) enzymes involved in the biosynthesis of mycolic acids: the organization of 3-ketoacyl-ACP synthases (KasA and KasB) and (3R)-hydroxyacyl-ACP dehydratases (HadAB/BC), on one hand, and the presence of two copies of the hadB gene encoding the catalytic domain of the latter enzyme type, on the other. This observation is discussed in light of the most recent data accumulated on the biosynthesis of this hallmark of Corynebacteriales.


Subject(s)
Actinomycetales/chemistry , Actinomycetales/genetics , Biosynthetic Pathways/genetics , Mycolic Acids/chemistry , Mycolic Acids/metabolism , Actinomycetales/metabolism , Enzymes/genetics , Magnetic Resonance Spectroscopy , Mass Spectrometry
13.
Microbiology (Reading) ; 158(Pt 3): 843-855, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22194354

ABSTRACT

Corynebacterineae are characterized by the presence of long-chain lipids, notably mycolic acids (α-alkyl, ß-hydroxy fatty acids), the structures of which are genus-specific. Mycolic acids from two environmental strains, Amycolicicoccus subflavus and Hoyosella altamirensis, were isolated and their structures were established using a combination of mass spectrometry analysis, (1)H-NMR spectroscopy and chemical degradations. The C(2)-C(3) cleavage of these C(30)-C(36) acids led to the formation of two fragments: saturated C(9)-C(11) acids, and saturated and unsaturated C(20)-C(25) aldehydes. Surprisingly, the fatty acids at the origin of the two fragments making up these mycolic acids were present in only minute amounts in the fatty acid pool. Moreover, the double bond in the main C(24) aldehyde fragment was located at position ω16, whereas that found in the ethylenic fatty acids of the bacteria was at ω9. These data question the biosynthesis of these new mycolic acids in terms of the nature of the precursors, chain elongation and desaturation. Nevertheless, they are consistent with the occurrence of the key genes of mycolic acid biosynthesis, including those encoding proteins of the fatty acid synthase II system, identified in the genome of A. subflavus. Altogether, while the presence of mycolic acids and analysis of their 16S rDNA sequences would suggest that these strains belong to the Mycobacteriaceae family, the originality of their structures reinforces the recent description of the novel genera Amycolicicoccus and Hoyosella.


Subject(s)
Actinomycetales/chemistry , Actinomycetales/classification , Environmental Microbiology , Mycolic Acids/analysis , Actinomycetales/isolation & purification , Biosynthetic Pathways/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mycolic Acids/chemistry , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
14.
Mol Microbiol ; 78(4): 989-1003, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21062372

ABSTRACT

The MmpS family (mycobacterial membrane protein small) includes over 100 small membrane proteins specific to the genus Mycobacterium that have not yet been studied experimentally. The genes encoding MmpS proteins are often associated with mmpL genes, which are homologous to the RND (resistance nodulation cell division) genes of Gram-negative bacteria that encode proteins functioning as multidrug efflux system. We showed by molecular genetics and biochemical analysis that MmpS4 in Mycobacterium smegmatis is required for the production and export of large amounts of cell surface glycolipids, but is dispensable for biosynthesis per se. A new specific and sensitive method utilizing single-chain antibodies against the surface-exposed glycolipids was developed to confirm that MmpS4 was dispensable for transport to the surface. Orthologous complementation demonstrated that the MmpS4 proteins are exchangeable, thus not specific to a defined lipid species. MmpS4 function requires the formation of a protein complex at the pole of the bacillus, which requires the extracytosolic C-terminal domain of MmpS4. We suggest that MmpS proteins facilitate lipid biosynthesis by acting as a scaffold for coupled biosynthesis and transport machinery.


Subject(s)
Bacterial Proteins/metabolism , Glycolipids/metabolism , Glycopeptides/metabolism , Membrane Proteins/metabolism , Mycobacterium smegmatis/metabolism , Bacterial Proteins/genetics , Genetic Complementation Test , Membrane Proteins/genetics , Mycobacterium smegmatis/genetics , Single-Chain Antibodies/immunology
15.
Cell Surf ; 7: 100060, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34485766

ABSTRACT

Four serine/threonine kinases are present in all mycobacteria: PknA, PknB, PknG and PknL. PknA and PknB are essential for growth and replication, PknG regulates metabolism, but little is known about PknL. Inactivation of pknL and adjacent regulator MSMEG_4242 in rough colony M. smegmatis mc2155 produced both smooth and rough colonies. Upon restreaking rough colonies, smooth colonies appeared at a frequency of ~ 1/250. Smooth mutants did not form biofilms, showed increased sliding motility and anomalous lipids on thin-layer chromatography, identified by mass spectrometry as lipooligosaccharides and perhaps also glycopeptidolipids. RNA-seq and Sanger sequencing revealed that all smooth mutants had inactivated lsr2 genes due to mutations and different IS1096 insertions. When complemented with lsr2, the colonies became rough, anomalous lipids disappeared and sliding motility decreased. Smooth mutants showed increased expression of IS1096 transposase TnpA and MSMEG_4727, which encodes a protein similar to PKS5. When MSMEG_4727 was deleted, smooth pknL/MSMEG_4242/lsr2 mutants reverted to rough, formed good biofilms, their motility decreased slightly and their anomalous lipids disappeared. Rough delpknL/del4242 mutants formed poor biofilms and showed decreased, aberrant sliding motility and both phenotypes were complemented with the two deleted genes. Inactivation of lsr2 changes colony morphology from rough to smooth, augments sliding motility and increases expression of MSMEG_4727 and other enzymes synthesizing lipooligosaccharides, apparently preventing biofilm formation. Similar morphological phase changes occur in other mycobacteria, likely reflecting environmental adaptations. PknL and MSMEG_4242 regulate lipid components of the outer cell envelope and their absence selects for lsr2 inactivation. A regulatory, phosphorylation cascade model is proposed.

16.
J Bacteriol ; 192(18): 4776-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20601476

ABSTRACT

MSMEG_0220 from Mycobacterium smegmatis, the ortholog of the Rv0183 gene from M. tuberculosis, recently identified and characterized as encoding a monoacylglycerol lipase, was cloned and expressed in Escherichia coli. The recombinant protein (rMSMEG_0220), which exhibits 68% amino acid sequence identity with Rv0183, showed the same substrate specificity and similar patterns of pH-dependent activity and stability as the M. tuberculosis enzyme. rMSMEG_0220 was found to hydrolyze long-chain monoacylglycerol with a specific activity of 143 +/- 6 U mg(-1). Like Rv0183 in M. tuberculosis, MSMEG_0220 was found to be located in the cell wall. To assess the in vivo role of the homologous proteins, an MSMEG_0220 disrupted mutant of M. smegmatis (MsDelta0220) was produced. An intriguing change in the colony morphology and in the cell interaction, which were partly restored in the complemented mutant containing either an active (ComMsDelta0220) or an inactive (ComMsDelta0220S111A) enzyme, was observed. Growth studies performed in media supplemented with monoolein showed that the ability of both MsDelta0220 and ComMsDelta0220S111A to grow in the presence of this lipid was impaired. Moreover, studies of the antimicrobial susceptibility of the MsDelta0220 strain showed that this mutant is more sensitive to rifampin and more resistant to isoniazid than the wild-type strain, pointing to a critical structural role of this enzyme in mycobacterial physiology, in addition to its function in the hydrolysis of exogenous lipids.


Subject(s)
Monoacylglycerol Lipases/metabolism , Mycobacterium smegmatis/cytology , Mycobacterium smegmatis/enzymology , Anti-Bacterial Agents/pharmacology , Blotting, Western , Chloramphenicol/pharmacology , Electrophoresis, Polyacrylamide Gel , Genetic Complementation Test , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Monoacylglycerol Lipases/genetics , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Novobiocin/pharmacology , Rifampin/pharmacokinetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
17.
J Bacteriol ; 192(15): 4037-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20511508

ABSTRACT

The fatty acid synthase type II enzymatic complex of Mycobacterium tuberculosis (FAS-II(Mt)) catalyzes an essential metabolic pathway involved in the biosynthesis of major envelope lipids, mycolic acids. The partner proteins of this singular FAS-II system represent relevant targets for antituberculous drug design. Two heterodimers of the hydratase 2 protein family, HadAB and HadBC, were shown to be involved in the (3R)-hydroxyacyl-ACP dehydration (HAD) step of FAS-II(Mt) cycles. Recently, an additional member of this family, Rv0241c, was proposed to have the same function, based on the heterologous complementation of a HAD mutant of the yeast mitochondrial FAS-II system. In the present work, Rv0241c was able to complement a HAD mutant in the Escherichia coli model but not a dehydratase-isomerase deficient mutant. However, an enzymatic study of the purified protein demonstrated that Rv0241c possesses a broad chain length specificity for the substrate, unlike FAS-II(Mt) enzymes. Most importantly, Rv0241c exhibited a strict dependence on the coenzyme A (CoA) as opposed to AcpM, the natural acyl carrier protein bearing the chains elongated by FAS-II(Mt). The deletion of Rv0241c showed that this gene is not essential to M. tuberculosis survival in vitro. The resulting mutant did not display any change in the mycolic acid profile. This demonstrates that Rv0241c is a trans-2-enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydratase that does not belong to FAS-II(Mt). The relevance of a heterologous complementation strategy to identifying proteins of such a system is questioned.


Subject(s)
Fatty Acid Synthase, Type II/metabolism , Fatty Acids/metabolism , Mycobacterium tuberculosis/enzymology , Fatty Acid Synthase, Type II/classification , Fatty Acid Synthase, Type II/genetics , Gene Deletion , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/genetics , Mycolic Acids/metabolism , Substrate Specificity
18.
FASEB J ; 23(12): 4091-104, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19671666

ABSTRACT

Bacterial species utilize a vast repertoire of surface structures to interact with their surroundings and employ a number of strategies to reconfigure the cellular envelope according to specific stimuli. Gram-positive bacteria, exemplified by Streptomyces and Bacillus species, control production of some exposed molecules by importing oligopeptide signals via permeases (Opp). Such oligopeptides modulate intracellular signaling pathways. In this work, we functionally characterized an Opp of the human pathogen Mycobacterium tuberculosis (Mtb) and propose its reannotation. Using genome-wide transcriptional profiling, we found that Opp was required to modulate (fold-change ranging from -3.5 to 2.0) the expression of several genes, most of them encoding surface-exposed molecules. These included the virulence-associated lipids mycolic acids and phthiocerol dimycocerosates (PDIMs) as well as PE-family proteins. By thin-layer chromatography and MALDI-TOF-MS we confirmed changes in the lipid profile, including an altered accumulation of triacylglycerides and an affected ratio of mycolic acids to PDIMs. An Opp loss of function mutant showed no in vitro growth defect, but had diminished burden during chronic infection and produced a slightly delayed time to death of animals when compared to WT Mtb infection.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Animals , Bacterial Proteins/genetics , Biological Transport, Active , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Triglycerides/metabolism
19.
J Bacteriol ; 191(8): 2613-21, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19181796

ABSTRACT

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.


Subject(s)
Lipopolysaccharides/biosynthesis , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/metabolism , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Chromatography, Thin Layer , Gas Chromatography-Mass Spectrometry , Gene Deletion , Gene Order , Genes, Bacterial , Genetic Complementation Test , Metabolic Networks and Pathways/genetics , Multigene Family , Mutagenesis, Insertional , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
J Bacteriol ; 191(19): 5941-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19648248

ABSTRACT

Latent tuberculosis represents a high-risk burden for one-third of the world population. Previous analysis of murine tuberculosis identified a novel transcriptional regulator encoded by Rv0348 that could control the establishment of persistent tuberculosis. Disruption of the Rv0348 gene from the genome of the virulent H37Rv strain of Mycobacterium tuberculosis revealed a global impact on the transcriptional profiles of 163 genes, including induction of the mammalian cell entry (mce1) operon and the repression of a significant number of genes involved in hypoxia and starvation responses. Nonetheless, gel shift assays did not reveal direct binding between Rv0348 and a set of regulated promoters, suggesting an indirect regulatory role. However, when expressed in Mycobacterium smegmatis, the Rv0348 transcripts were significantly responsive to different levels of hypoxia and the encoded protein was shown to regulate genes involved in hypoxia [e.g., Rv3130c (tgs1)] and intracellular survival (e.g., mce1), among other genes. Interestingly, the colonization level of the DeltamosR mutant strain was significantly lower than that of the wild-type strain of M. tuberculosis, suggesting its attenuation in the murine model of tuberculosis. Taken together, our analyses indicated that the Rv0348 gene encodes a novel transcriptional factor that regulates several operons involved in mycobacterial survival, especially during hypoxia; hence, we propose that Rv0348 be renamed mosR for regulator of mycobacterial operons of survival.


Subject(s)
Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Transcription, Genetic/genetics , Virulence/genetics , Animals , Bacterial Proteins/genetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/physiology , Immunoblotting , Mice , Mice, Inbred BALB C , Mutation , Mycobacterium tuberculosis/pathogenicity , Operon/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Binding/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tuberculosis/genetics , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL