Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
EMBO J ; 40(14): e106111, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34018220

ABSTRACT

Mycobacterium tuberculosis (Mtb) has evolved diverse cellular processes in response to the multiple stresses it encounters within the infected host. We explored available TnSeq datasets to identify transcription factors (TFs) that are essential for Mtb survival inside the host. The analysis identified a single TF, Rv1332 (AosR), conserved across actinomycetes with a so-far uncharacterized function. AosR mitigates phagocyte-derived oxidative and nitrosative stress, thus promoting mycobacterial growth in the murine lungs and spleen. Oxidative stress induces formation of a single intrasubunit disulphide bond in AosR, which in turn facilitates AosR interaction with an extracytoplasmic-function sigma factor, SigH. This leads to the specific upregulation of the CysM-dependent non-canonical cysteine biosynthesis pathway through an auxiliary intragenic stress-responsive promoter, an axis critical in detoxifying host-derived oxidative and nitrosative radicals. Failure to upregulate AosR-dependent cysteine biosynthesis during the redox stress causes differential expression of 6% of Mtb genes. Our study shows that the AosR-SigH pathway is critical for detoxifying host-derived oxidative and nitrosative radicals to enhance Mtb survival in the hostile intracellular environment.


Subject(s)
Actinobacteria/genetics , Homeostasis/genetics , Mycobacterium tuberculosis/genetics , Transcription Factors/genetics , Animals , Bacterial Proteins/genetics , Female , Gene Expression Regulation, Bacterial/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress/genetics , Promoter Regions, Genetic/genetics , Sigma Factor/genetics , Transcription, Genetic/genetics
2.
J Biol Chem ; 299(3): 102933, 2023 03.
Article in English | MEDLINE | ID: mdl-36690275

ABSTRACT

SigA (σA) is an essential protein and the primary sigma factor in Mycobacterium tuberculosis (Mtb). However, due to the absence of genetic tools, our understanding of the role and regulation of σA activity and its molecular attributes that help modulate Mtb survival is scant. Here, we generated a conditional gene replacement of σA in Mtb and showed that its depletion results in a severe survival defect in vitro, ex vivo, and in vivo in a murine infection model. Our RNA-seq analysis suggests that σA either directly or indirectly regulates ∼57% of the Mtb transcriptome, including ∼28% of essential genes. Surprisingly, we note that despite having ∼64% similarity with σA, overexpression of the primary-like σ factor SigB (σB) fails to compensate for the absence of σA, suggesting minimal functional redundancy. RNA-seq analysis of the Mtb σB deletion mutant revealed that 433 genes are regulated by σB, of which 283 overlap with the σA transcriptome. Additionally, surface plasmon resonance, in vitro transcription, and functional complementation experiments reveal that σA residues between 132-179 that are disordered and missing from all experimentally determined σA-RNAP structural models are imperative for σA function. Moreover, phosphorylation of σA in the intrinsically disordered N-terminal region plays a regulatory role in modulating its activity. Collectively, these observations and analysis provide a rationale for the centrality of σA for the survival and pathogenicity of this bacillus.


Subject(s)
Bacterial Proteins , Microbial Viability , Mycobacterium tuberculosis , Sigma Factor , Sigma Factor/genetics , Sigma Factor/metabolism , Animals , Mice , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Transcriptome , Tuberculosis/microbiology , Sequence Deletion , Microbial Viability/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics
3.
J Biol Chem ; 299(12): 105364, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865319

ABSTRACT

Nucleoid-associated proteins (NAPs) regulate multiple cellular processes such as gene expression, virulence, and dormancy throughout bacterial species. NAPs help in the survival and adaptation of Mycobacterium tuberculosis (Mtb) within the host. Fourteen NAPs have been identified in Escherichia coli; however, only seven NAPs are documented in Mtb. Given its complex lifestyle, it is reasonable to assume that Mtb would encode for more NAPs. Using bioinformatics tools and biochemical experiments, we have identified the heparin-binding hemagglutinin (HbhA) protein of Mtb as a novel sequence-independent DNA-binding protein which has previously been characterized as an adhesion molecule required for extrapulmonary dissemination. Deleting the carboxy-terminal domain of HbhA resulted in a complete loss of its DNA-binding activity. Atomic force microscopy showed HbhA-mediated architectural modulations in the DNA, which may play a regulatory role in transcription and genome organization. Our results showed that HbhA colocalizes with the nucleoid region of Mtb. Transcriptomics analyses of a hbhA KO strain revealed that it regulates the expression of ∼36% of total and ∼29% of essential genes. Deletion of hbhA resulted in the upregulation of ∼73% of all differentially expressed genes, belonging to multiple pathways suggesting it to be a global repressor. The results show that HbhA is a nonessential NAP regulating gene expression globally and acting as a plausible transcriptional repressor.


Subject(s)
Bacterial Proteins , Hemagglutinins , Mycobacterium tuberculosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA/chemistry , DNA/metabolism , Hemagglutinins/genetics , Hemagglutinins/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Gene Expression Regulation, Bacterial/genetics , Gene Deletion , DNA-Binding Proteins/genetics , Protein Domains/genetics , Microscopy, Atomic Force
4.
PLoS Pathog ; 17(3): e1009452, 2021 03.
Article in English | MEDLINE | ID: mdl-33740020

ABSTRACT

Tuberculosis caused by Mycobacterium tuberculosis (Mtb) is a significant public health concern, exacerbated by the emergence of drug-resistant TB. To combat the host's dynamic environment, Mtb encodes multiple DNA repair enzymes that play a critical role in maintaining genomic integrity. Mtb possesses a GC-rich genome, rendering it highly susceptible to cytosine deaminations, resulting in the occurrence of uracils in the DNA. UDGs encoded by ung and udgB initiate the repair; hence we investigated the biological impact of deleting UDGs in the adaptation of pathogen. We generated gene replacement mutants of uracil DNA glycosylases, individually (RvΔung, RvΔudgB) or together (RvΔdKO). The double KO mutant, RvΔdKO exhibited remarkably higher spontaneous mutation rate, in the presence of antibiotics. Interestingly, RvΔdKO showed higher survival rates in guinea pigs and accumulated large number of SNPs as revealed by whole-genome sequence analysis. Competition assays revealed the superior fitness of RvΔdKO over Rv, both in ex vivo and in vivo conditions. We propose that compromised DNA repair results in the accumulation of mutations, and a subset of these drives adaptation in the host. Importantly, this property allowed us to utilize RvΔdKO for the facile identification of drug targets.


Subject(s)
Adaptation, Physiological/genetics , DNA Repair/physiology , Host Specificity/genetics , Mycobacterium tuberculosis/genetics , Animals , Guinea Pigs , Mice
5.
Antimicrob Agents Chemother ; 66(3): e0177321, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35156855

ABSTRACT

Applying antibiotics to susceptible bacterial cultures generates a minor population of persisters that remain susceptible to antibiotics but can endure them for extended periods. Recent reports suggest that antibiotic persisters (APs) of mycobacteria experience oxidative stress and develop resistance upon treatment with lethal doses of ciprofloxacin or rifampicin. However, the mechanisms driving the de novo emergence of resistance remained unclear. Here, we show that mycobacterial APs activate the SOS response, resulting in the upregulation of the error-prone DNA polymerase DnaE2. The sustained expression of dnaE2 in APs led to mutagenesis across the genome and resulted in the rapid evolution of resistance to antibiotics. Inhibition of RecA by suramin, an anti-Trypanosoma drug, reduced the rate of conversion of persisters to resistors in a diverse group of bacteria. Our study highlights suramin's novel application as a broad-spectrum agent in combating the development of drug resistance.


Subject(s)
Drug Resistance, Bacterial , Mycobacterium tuberculosis , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial/genetics , Mycobacterium tuberculosis/genetics , Rifampin/pharmacology
6.
Biochem J ; 478(11): 2081-2099, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33955473

ABSTRACT

N-acetyl glucosamine-1-phosphate uridyltransferase (GlmU) is a bifunctional enzyme involved in the biosynthesis of Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc). UDP-GlcNAc is a critical precursor for the synthesis of peptidoglycan and other cell wall components. The absence of a homolog in eukaryotes makes GlmU an attractive target for therapeutic intervention. Mycobacterium tuberculosis GlmU (GlmUMt) has features, such as a C-terminal extension, that are not present in GlmUorthologs from other bacteria. Here, we set out to determine the uniqueness of GlmUMt by performing in vivo complementation experiments using RvΔglmU mutant. We find that any deletion of the carboxy-terminal extension region of GlmUMt abolishes its ability to complement the function of GlmUMt. Results show orthologs of GlmU, including its closest ortholog, from Mycobacterium smegmatis, cannot complement the function of GlmUMt. Furthermore, the co-expression of GlmUMt domain deletion mutants with either acetyl or uridyltransferase activities failed to rescue the function. However, co-expression of GlmUMt point mutants with either acetyl or uridyltransferase activities successfully restored the biological function of GlmUMt, likely due to the formation of heterotrimers. Based on the interactome experiments, we speculate that GlmUMt participates in unique interactions essential for its in vivo function.


Subject(s)
Acetyltransferases/metabolism , Bacterial Proteins/metabolism , Multienzyme Complexes/metabolism , Mutation , Mycobacterium tuberculosis/growth & development , Tuberculosis/microbiology , UDPglucose-Hexose-1-Phosphate Uridylyltransferase/metabolism , Acetyltransferases/chemistry , Acetyltransferases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Protein Conformation , Protein Domains , UDPglucose-Hexose-1-Phosphate Uridylyltransferase/chemistry , UDPglucose-Hexose-1-Phosphate Uridylyltransferase/genetics
7.
J Infect Dis ; 224(8): 1383-1393, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33580239

ABSTRACT

Attenuated intracellular survival of Mycobacterium tuberculosis (Mtb) secretory gene mutants exemplifies their role as virulence factors. Mtb peptidyl prolyl isomerase A (PPiA) assists in protein folding through cis/trans isomerization of prolyl bonds. Here, we show that PPiA abets Mtb survival and aids in disease progression by exploiting host-associated factors. While the deletion of PPiA has no discernable effect on bacillary survival in a murine infection model, it compromises the formation of granuloma-like lesions and promotes host cell death through ferroptosis. Overexpression of PPiA enhances the bacillary load and exacerbates pathology in mice lungs. Importantly, PPiA interacts with the integrin α5ß1 receptor through a conserved surface-exposed RGD motif. The secretion of PPiA as well as interaction with integrin contributes to disease progression by upregulating multiple host matrix metalloproteinases. Collectively, we identified a novel nonchaperone role of PPiA that is critical in facilitating host-pathogen interaction and ensuing disease progression.


Subject(s)
Host-Pathogen Interactions , Mycobacterium tuberculosis/enzymology , Peptidylprolyl Isomerase/metabolism , Animals , Disease Progression , Integrins , Mice
8.
Article in English | MEDLINE | ID: mdl-33468473

ABSTRACT

Eradication of tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), has been a challenge due to its uncanny ability to survive in a dormant state inside host granulomas for decades. Mtb rewires its metabolic and redox regulatory networks to survive in the hostile hypoxic and nutrient-limiting environment, facilitating the formation of drug-tolerant persisters. Previously, we showed that protein kinase G (PknG), a virulence factor required for lysosomal escape, aids in metabolic adaptation, thereby promoting the survival of nonreplicating mycobacteria. Here, we sought to investigate the therapeutic potential of PknG against latent mycobacterium. We show that inhibition of PknG by AX20017 reduces mycobacterial survival in in vitro latency models such as hypoxia, persisters, and nutrient starvation. Targeting PknG enhances the bactericidal activity of the frontline anti-TB drugs in peritoneal macrophages. Deletion of pknG resulted in 5- to 15-fold-reduced survival of Mtb in chronically infected mice treated with anti-TB drugs. Importantly, in the Cornell mouse model of latent TB, the deletion of pknG drastically attenuated Mtb's ability to resuscitate after antibiotic treatment compared with wild-type and complemented strains. This is the first study to investigate the sterilizing activity of pknG deletion and inhibition for adjunct therapy against latent TB in a preclinical model. Collectively, these results suggest that PknG may be a promising drug target for adjunct therapy to shorten the treatment duration and reduce disease relapse.


Subject(s)
Latent Tuberculosis , Mycobacterium tuberculosis , Tuberculosis , Animals , Antitubercular Agents/pharmacology , Latent Tuberculosis/drug therapy , Mice , Mycobacterium tuberculosis/genetics , Temefos , Tuberculosis/drug therapy
9.
J Immunol ; 203(5): 1218-1229, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31375544

ABSTRACT

Mycobacterium tuberculosis employs defense mechanisms to protect itself from reactive oxygen species (ROS)-mediated cytotoxicity inside macrophages. In the current study, we found that a secretory protein of M. tuberculosis PPE2 disrupted the assembly of NADPH oxidase complex. PPE2 inhibited NADPH oxidase-mediated ROS generation in RAW 264.7 macrophages and peritoneal macrophages from BALB/c mice. PPE2 interacted with the cytosolic subunit of NADPH oxidase, p67phox, and prevented translocation of p67phox and p47phox to the membrane, resulting in decreased NADPH oxidase activity. Trp236 residue present in the SH3-like domain of PPE2 was found to be critical for its interaction with p67phox Trp236Ala mutant of PPE2 did not interact with p67phox and thereby did not affect ROS generation. M. tuberculosis expressing PPE2 and PPE2-null mutants complemented with PPE2 survived better than PPE2-null mutants in infected RAW 264.7 macrophages. Altogether, this study suggests that PPE2 inhibits NADPH oxidase-mediated ROS production to favor M. tuberculosis survival in macrophages. The findings that M. tuberculosis PPE2 protein is involved in the modulation of oxidative response in macrophages will help us in improving our knowledge of host-pathogen interactions and the application of better therapeutics against tuberculosis.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Phosphoproteins/physiology , Reactive Oxygen Species/metabolism , Animals , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Host-Pathogen Interactions , Macrophages/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Tetradecanoylphorbol Acetate/pharmacology , src Homology Domains
10.
J Bacteriol ; 201(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30617240

ABSTRACT

Extracytoplasmic function σ factors that are stress inducible are often sequestered in an inactive complex with a membrane-associated anti-σ factor. Mycobacterium tuberculosis membrane-associated anti-σ factors have a small, stable RNA gene A (ssrA)-like degron for targeted proteolysis. Interaction between the unfoldase, ClpX, and a substrate with an accessible degron initiates energy-dependent proteolysis. Four anti-σ factors with a mutation in the degron provided a set of natural substrates to evaluate the influence of the degron on degradation strength in ClpX-substrate processivity. We note that a point mutation in the degron (X-Ala-Ala) leads to an order-of-magnitude difference in the dwell time of the substrate on ClpX. Differences in ClpX/anti-σ interactions were correlated with changes in unfoldase activities. Green fluorescent protein (GFP) chimeras or polypeptides with a length identical to that of the anti-σ factor degron also demonstrate degron-dependent variation in ClpX activities. We show that degron-dependent ClpX activity leads to differences in anti-σ degradation, thereby regulating the release of free σ from the σ/anti-σ complex. M. tuberculosis ClpX activity thus influences changes in gene expression by modulating the cellular abundance of ECF σ factors.IMPORTANCE The ability of Mycobacterium tuberculosis to quickly adapt to changing environmental stimuli occurs by maintaining protein homeostasis. Extracytoplasmic function (ECF) σ factors play a significant role in coordinating the transcription profile to changes in environmental conditions. Release of the σ factor from the anti-σ is governed by the ClpXP2P1 assembly. M. tuberculosis ECF anti-σ factors have an ssrA-like degron for targeted degradation. A point mutation in the degron leads to differences in ClpX-mediated proteolysis and affects the cellular abundance of ECF σ factors. ClpX activity thus synchronizes changes in gene expression with environmental stimuli affecting M. tuberculosis physiology.


Subject(s)
Endopeptidase Clp/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Repressor Proteins/metabolism , Sigma Factor/metabolism , DNA Mutational Analysis , Point Mutation , Proteolysis , Repressor Proteins/genetics
11.
J Biol Chem ; 293(17): 6497-6516, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29530985

ABSTRACT

The cell wall of Mycobacterium tuberculosis (Mtb) is a complex structure that protects the pathogen in hostile environments. Peptidoglycan (PG), which helps determine the morphology of the cell envelope, undergoes substantial remodeling under stress. This meshwork of linear chains of sugars, cross-linked through attached peptides, is generated through the sequential action of enzymes termed transglycosylases and transpeptidases. The Mtb genome encodes two classical transglycosylases and four transpeptidases, the functions of which are not fully elucidated. Here, we present work on the yet uncharacterized transpeptidase PbpA and a nonclassical transglycosylase RodA. We elucidate their roles in regulating in vitro growth and in vivo survival of pathogenic mycobacteria. We find that RodA and PbpA are required for regulating cell length, but do not affect mycobacterial growth. Biochemical analyses show PbpA to be a classical transpeptidase, whereas RodA is identified to be a member of an emerging class of noncanonical transglycosylases. Phosphorylation of RodA at Thr-463 modulates its biological function. In a guinea pig infection model, RodA and PbpA are found to be required for both bacterial survival and formation of granuloma structures, thus underscoring the importance of these proteins in mediating mycobacterial virulence in the host. Our results emphasize the fact that whereas redundant enzymes probably compensate for the absence of RodA or PbpA during in vitro growth, the two proteins play critical roles for the survival of the pathogen inside its host.


Subject(s)
Bacterial Proteins , Glycosyltransferases , Granuloma, Respiratory Tract , Microbial Viability , Mycobacterium tuberculosis , Peptidyl Transferases , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Genome, Bacterial , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Granuloma, Respiratory Tract/enzymology , Granuloma, Respiratory Tract/genetics , Granuloma, Respiratory Tract/pathology , Guinea Pigs , Male , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Peptidyl Transferases/genetics , Peptidyl Transferases/metabolism , Tuberculosis/enzymology , Tuberculosis/genetics , Tuberculosis/pathology
12.
J Biol Chem ; 293(32): 12331-12349, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29903917

ABSTRACT

Identifying and characterizing the individual contributors to bacterial cellular elongation and division will improve our understanding of their impact on cell growth and division. Here, we delineated the role of ftsQ, a terminal gene of the highly conserved division cell wall (dcw) operon, in growth, survival, and cell length maintenance in the human pathogen Mycobacterium tuberculosis (Mtb). We found that FtsQ overexpression significantly increases the cell length and number of multiseptate cells. FtsQ depletion in Mtb resulted in cells that were shorter than WT cells during the initial growth stages (4 days after FtsQ depletion) but were longer than WT cells at later stages (10 days after FtsQ depletion) and compromised the survival in vitro and in differentiated THP1 macrophages. Overexpression of N- and C-terminal FtsQ regions altered the cell length, and the C-terminal domain alone complemented the FtsQ depletion phenotype. MS analyses suggested robust FtsQ phosphorylation on Thr-24, and although phosphoablative and -mimetic mutants rescued the FtsQ depletion-associated cell viability defects, they failed to complement the cell length defects. MS and coimmunoprecipitation experiments identified 63 FtsQ-interacting partners, and we show that the interaction of FtsQ with the recently identified cell division protein SepIVA is independent of FtsQ phosphorylation and suggests a role of FtsQ in modulating cell division. FtsQ exhibited predominantly septal localization in both the presence and absence of SepIVA. Our results suggest a role for FtsQ in modulating the length, division, and survival of Mtb cells both in vitro and in the host.


Subject(s)
Bacterial Proteins/metabolism , Cell Division , Macrophages/cytology , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Bacterial Proteins/genetics , Cells, Cultured , Humans , Macrophages/microbiology , Mutation , Protein Binding
14.
Bioconjug Chem ; 30(3): 721-732, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30669829

ABSTRACT

A major impediment to developing effective antimicrobials against Gram-negative bacteria like Salmonella is the ability of the bacteria to develop resistance against existing antibiotics and the inability of the antimicrobials to clear the intracellular bacteria residing in the gastrointestinal tract. As the critical balance of charge and hydrophobicity is required for effective membrane-targeting antimicrobials without causing any toxicity to mammalian cells, herein we report the synthesis and antibacterial properties of cholic acid-derived amphiphiles conjugated with alkyl chains of varied hydrophobicity. Relative to other hydrophobic counterparts, a compound with hexyl chain (6) acted as an effective antimicrobial against different Gram-negative bacteria. Apart from its ability to permeate the outer and inner membranes of bacteria; compound 6 can cross the cellular and lysosomal barriers of epithelial cells and macrophages and kill the facultative intracellular bacteria without disrupting the mammalian cell membranes. Oral delivery of compound 6 was able to clear the Salmonella-mediated gut infection and inflammation, and was able to combat persistent, stationary, and multi-drug-resistant clinical strains. Therefore, our study reveals the ability of cholic acid-derived amphiphiles to clear intracellular bacteria and Salmonella-mediated gut infection and inflammation.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cholic Acid/administration & dosage , Inflammation/prevention & control , Intestinal Diseases/prevention & control , Salmonella Infections/prevention & control , Administration, Oral , Animals , Drug Resistance, Multiple, Bacterial , Intestinal Diseases/microbiology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Salmonella/isolation & purification , Salmonella/pathogenicity
15.
J Biol Chem ; 292(39): 16093-16108, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28821621

ABSTRACT

Protein kinase G (PknG), a thioredoxin-fold-containing eukaryotic-like serine/threonine protein kinase, is a virulence factor in Mycobacterium tuberculosis, required for inhibition of phagolysosomal fusion. Here, we unraveled novel functional facets of PknG during latency-like conditions. We found that PknG mediates persistence under stressful conditions like hypoxia and abets drug tolerance. PknG mutant displayed minimal growth in nutrient-limited conditions, suggesting its role in modulating cellular metabolism. Intracellular metabolic profiling revealed that PknG is necessary for efficient metabolic adaptation during hypoxia. Notably, the PknG mutant exhibited a reductive shift in mycothiol redox potential and compromised stress response. Exposure to antibiotics and hypoxic environment resulted in higher oxidative shift in mycothiol redox potential of PknG mutant compared with the wild type. Persistence during latency-like conditions required kinase activity and thioredoxin motifs of PknG and is mediated through phosphorylation of a central metabolic regulator GarA. Finally, using a guinea pig model of infection, we assessed the in vivo role of PknG in manifestation of disease pathology and established a role for PknG in the formation of stable granuloma, hallmark structures of latent tuberculosis. Taken together, PknG-mediated GarA phosphorylation is important for maintenance of both mycobacterial physiology and redox poise, an axis that is dispensable for survival under normoxic conditions but is critical for non-replicating persistence of mycobacteria. In conclusion, we propose that PknG probably acts as a modulator of latency-associated signals.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Granuloma/etiology , Latent Tuberculosis/microbiology , Mycobacterium tuberculosis/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acid Substitution , Animals , Antibiotics, Antitubercular/pharmacology , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Female , Gene Deletion , Granuloma/metabolism , Granuloma/microbiology , Guinea Pigs , Isoniazid/pharmacology , Kinetics , Latent Tuberculosis/metabolism , Latent Tuberculosis/physiopathology , Metabolomics/methods , Microbial Viability/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/physiology , Phosphorylation/drug effects , Point Mutation , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Stress, Physiological
16.
J Biol Chem ; 292(17): 6855-6868, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28209712

ABSTRACT

Mycobacterium tuberculosis is known to modulate the host immune responses to facilitate its persistence inside the host cells. One of the key mechanisms includes repression of class-II transactivator (CIITA) and MHC-II expression in infected macrophages. However, the precise mechanism of CIITA and MHC-II down-regulation is not well studied. M. tuberculosis 6-kDa early secretory antigenic target (ESAT-6) is a known potent virulence and antigenic determinant. The M. tuberculosis genome encodes 23 such ESAT-6 family proteins. We herein report that M. tuberculosis and M. bovis bacillus Calmette-Guérin infection down-regulated the expression of CIITA/MHC-II by inducing hypermethylation in histone H3 lysine 9 (H3K9me2/3). Further, we showed that M. tuberculosis ESAT-6 family protein EsxL, encoded by Rv1198, is responsible for the down-regulation of CIITA/MHC-II by inducing H3K9me2/3. We further report that M. tuberculosis esxL induced the expression of nitric-oxide synthase, NO production, and p38 MAPK pathway, which in turn was responsible for the increased H3K9me2/3 in CIITA via up-regulation of euchromatic histone-lysine N-methyltransferase 2 (G9a). In contrast, inhibition of nitric-oxide synthase, p38 MAPK, and G9a abrogated H3K9me2/3, resulting in increased CIITA expression. A chromatin immunoprecipitation assay confirmed that hypermethylation at the promoter IV region of CIITA is mainly responsible for CIITA down-regulation and subsequent antigen presentation. We found that co-culture of macrophages infected with esxL-expressing M. smegmatis and mouse splenocytes led to down-regulation of IL-2, a key cytokine involved in T-cell proliferation. In summary, we demonstrate that M. tuberculosis EsxL inhibits antigen presentation by enhancing H3K9me2/3 at the CIITA promoter, thereby repressing its expression through NO and p38 MAPK activation.


Subject(s)
Bacterial Proteins/physiology , DNA Methylation , Macrophages/metabolism , Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/metabolism , Nuclear Proteins/genetics , Trans-Activators/genetics , Animals , Antigen Presentation , Antigens, Bacterial/metabolism , Cell Line, Tumor , Cell Proliferation , Genome, Bacterial , Histones/metabolism , Humans , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System , Mice , Mutation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Signal Transduction , Spleen/cytology , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
IUBMB Life ; 70(9): 889-904, 2018 09.
Article in English | MEDLINE | ID: mdl-29934969

ABSTRACT

The emergence of increasingly drug-resistant Mycobacterium tuberculosis strains has become a crucial public health concern. In order to effectively treat tuberculosis, it is imperative to find newer drug targets, which are important for the in vivo bacterial survival and persistence. Phosphorylation based signaling cascades modulated by eukaryotic-like serine/threonine protein kinases and phosphatase in M. tuberculosis, transduce extracellular stimuli to a cellular response ensuing pathogen's growth, persistence and pathogenesis. Of the 11 STPKs that M. tuberculosis genome encodes, three kinases, namely PknA, PknB and PknG and the sole serine/threonine phosphatase PstP are crucial for the intracellular survival of the bacteria. PknA and PknB regulates cell growth, cell wall synthesis and morphological changes during bacterial cell division; while PknG modulates metabolic changes in response to stress and aids in bacterial survival during latency like conditions. PstP functions to dephosphorylate STPKs and their substrates and hence is important at nearly all stages of infection. Here, we review the current knowledge on PstP, PknA, PknB and PknG based on the genetic, biochemical, and functional studies in M. tuberculosis physiopathology. We further explore the potential of these molecules as targets for therapeutic intervention and discuss the advancement made in the development of inhibitors against these targets. © 2018 IUBMB Life, 70(9):889-904, 2018.


Subject(s)
Antitubercular Agents/pharmacology , Drug Development , Gene Expression Regulation, Enzymologic/drug effects , Mycobacterium tuberculosis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tuberculosis/drug therapy , Animals , Humans , Mycobacterium tuberculosis/enzymology , Phosphorylation , Tuberculosis/enzymology , Tuberculosis/microbiology
18.
J Biol Chem ; 291(46): 24215-24230, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27758870

ABSTRACT

Protein phosphatases play vital roles in phosphorylation-mediated cellular signaling. Although there are 11 serine/threonine protein kinases in Mycobacterium tuberculosis, only one serine/threonine phosphatase, PstP, has been identified. Although PstP has been biochemically characterized and multiple in vitro substrates have been identified, its physiological role has not yet been elucidated. In this study, we have investigated the impact of PstP on cell growth and survival of the pathogen in the host. Overexpression of PstP led to elongated cells and partially compromised survival. We find that depletion of PstP is detrimental to cell survival, eventually leading to cell death. PstP depletion results in elongated multiseptate cells, suggesting a role for PstP in regulating cell division events. Complementation experiments performed with PstP deletion mutants revealed marginally compromised survival, suggesting that all of the domains, including the extracellular domain, are necessary for complete rescue. On the other hand, the catalytic activity of PstP is absolutely essential for the in vitro growth. Mice infection experiments establish a definitive role for PstP in pathogen survival within the host. Depletion of PstP from established infections causes pathogen clearance, indicating that the continued presence of PstP is necessary for pathogen survival. Taken together, our data suggest an important role for PstP in establishing and maintaining infection, possibly via the modulation of cell division events.


Subject(s)
Bacterial Proteins/metabolism , Cell Division/physiology , Microbial Viability , Mycobacterium tuberculosis/enzymology , Phosphoprotein Phosphatases/metabolism , Tuberculosis/enzymology , Animals , Bacterial Proteins/genetics , Gene Deletion , Mice , Mycobacterium tuberculosis/genetics , Phosphoprotein Phosphatases/genetics , Tuberculosis/genetics
19.
J Biol Chem ; 291(27): 14257-14273, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27151218

ABSTRACT

Mycobacterium tuberculosis is an adaptable intracellular pathogen, existing in both dormant as well as active disease-causing states. Here, we report systematic proteomic analyses of four strains, H37Ra, H37Rv, and clinical isolates BND and JAL, to determine the differences in protein expression patterns that contribute to their virulence and drug resistance. Resolution of lysates of the four strains by liquid chromatography, coupled to mass spectrometry analysis, identified a total of 2161 protein groups covering ∼54% of the predicted M. tuberculosis proteome. Label-free quantification analysis of the data revealed 257 differentially expressed protein groups. The differentially expressed protein groups could be classified into seven K-means cluster bins, which broadly delineated strain-specific variations. Analysis of the data for possible mechanisms responsible for drug resistance phenotype of JAL suggested that it could be due to a combination of overexpression of proteins implicated in drug resistance and the other factors. Expression pattern analyses of transcription factors and their downstream targets demonstrated substantial differential modulation in JAL, suggesting a complex regulatory mechanism. Results showed distinct variations in the protein expression patterns of Esx and mce1 operon proteins in JAL and BND strains, respectively. Abrogating higher levels of ESAT6, an important Esx protein known to be critical for virulence, in the JAL strain diminished its virulence, although it had marginal impact on the other strains. Taken together, this study reveals that strain-specific variations in protein expression patterns have a meaningful impact on the biology of the pathogen.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Proteomics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Species Specificity , Virulence
20.
PLoS Pathog ; 11(10): e1005235, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26489015

ABSTRACT

M. tuberculosis N-acetyl-glucosamine-1-phosphate uridyltransferase (GlmUMtb) is a bi-functional enzyme engaged in the synthesis of two metabolic intermediates N-acetylglucosamine-1-phosphate (GlcNAc-1-P) and UDP-GlcNAc, catalyzed by the C- and N-terminal domains respectively. UDP-GlcNAc is a key metabolite essential for the synthesis of peptidoglycan, disaccharide linker, arabinogalactan and mycothiols. While glmUMtb was predicted to be an essential gene, till date the role of GlmUMtb in modulating the in vitro growth of Mtb or its role in survival of pathogen ex vivo / in vivo have not been deciphered. Here we present the results of a comprehensive study dissecting the role of GlmUMtb in arbitrating the survival of the pathogen both in vitro and in vivo. We find that absence of GlmUMtb leads to extensive perturbation of bacterial morphology and substantial reduction in cell wall thickness under normoxic as well as hypoxic conditions. Complementation studies show that the acetyl- and uridyl- transferase activities of GlmUMtb are independently essential for bacterial survival in vitro, and GlmUMtb is also found to be essential for mycobacterial survival in THP-1 cells as well as in guinea pigs. Depletion of GlmUMtb from infected murine lungs, four weeks post infection, led to significant reduction in the bacillary load. The administration of Oxa33, a novel oxazolidine derivative that specifically inhibits GlmUMtb, to infected mice resulted in significant decrease in the bacillary load. Thus our study establishes GlmUMtb as a strong candidate for intervention measures against established tuberculosis infections.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Multienzyme Complexes/antagonists & inhibitors , Mycobacterium tuberculosis , Tuberculosis/metabolism , Animals , Benzylidene Compounds/pharmacology , Blotting, Western , Disease Models, Animal , Guinea Pigs , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Naphthalenes/pharmacology , Oxazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL