Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
mLife ; 3(1): 87-100, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38827510

ABSTRACT

Insertion sequences (ISs) exist widely in bacterial genomes, but their roles in the evolution of bacterial antiphage defense remain to be clarified. Here, we report that, under the pressure of phage infection, the IS1096 transposition of Mycobacterium smegmatis into the lsr2 gene can occur at high frequencies, which endows the mutant mycobacterium with a broad-spectrum antiphage ability. Lsr2 functions as a negative regulator and directly silences expression of a gene island composed of 11 lipid metabolism-related genes. The complete or partial loss of the gene island leads to a significant decrease of bacteriophage adsorption to the mycobacterium, thus defending against phage infection. Strikingly, a phage that has evolved mutations in two tail-filament genes can re-escape from the lsr2 inactivation-triggered host defense. This study uncovered a new signaling pathway for activating antimycobacteriophage immunity by IS transposition and provided insight into the natural evolution of bacterial antiphage defense.

2.
Microbiol Spectr ; : e0106923, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37668384

ABSTRACT

Zinc excess is toxic to bacteria and, thus, represents an important innate defense mechanism of host cells, especially against mycobacterial infections. However, the signaling pathway triggered by zinc excess and its relationship with iron homeostasis remain poorly understood in mycobacteria. Here, we characterize a novel Zur-IdeR-iron homeostasis signaling pathway that modulates the growth of Mycobacterium bovis under zinc toxicity. We found that the regulator Zur interacts with the iron-homeostasis regulator IdeR, enhancing the DNA-binding ability of IdeR. Excess zinc disrupts this interaction and represses ideR transcription through Zur, which promotes the expression of iron uptake genes and leads to the accumulation of intracellular iron in M. bovis. The elevated iron levels lower the bacterial survival ability under excess zinc stress. Consistently, deleting zur hinders intracellular iron accumulation of M. bovis and enhances bacterial growth under stress, while silencing ideR impairs the growth of the wild-type and zur-deleted strains under the same conditions. Interestingly, both Zur and IdeR are conserved in bacteria facing zinc toxicity. Overall, our work uncovers a novel antimicrobial signal pathway whereby zinc excess disrupts iron homeostasis, which may deepen our understanding of the crosstalk mechanism between iron and zinc homeostasis in bacteria.IMPORTANCEAs a catalytic and structural cofactor of proteins, zinc is essential for almost all living organisms. However, zinc excess is toxic and represents a vital innate immunity strategy of macrophages to combat intracellular pathogens, especially against mycobacterial pathogens such as Mycobacterium tuberculosis, the causative agent of tuberculosis. Here, we first characterize an antibacterial signaling pathway of zinc excess and its relationship with iron homeostasis in M. bovis. We found that excess zinc inhibits the transcription of ideR and its DNA-binding activity through Zur, which, in turn, promotes the expression of iron uptake genes, causes intracellular iron accumulation, and finally impairs the bacterial growth. This study reveals the existence of the Zur-IdeR-iron homeostasis pathway triggered by zinc excess in M. bovis, which will shed light on the crosstalk mechanisms between zinc and iron homeostasis in bacteria and the antimicrobial mechanisms of host-mediated zinc toxicity.

3.
Cell Host Microbe ; 31(9): 1469-1480.e4, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37567169

ABSTRACT

In eukaryotic cells, serine/threonine protein kinases (StpKs) play important roles in limiting viral infections. StpKs are commonly activated upon infections, inhibiting the expression of genes central for viral replication. Here, we report that a eukaryotic-like StpK7 encoded by MSMEG_1200 in M. smegmatis is required for mycobacteriophage TM4 to escape bacterial defense. stpK7 is located within a gene island, MSMEG_1191-MSMEG_1200, containing multiple anti-phage genes resembling the BREX (bacteriophage exclusion) phage-resistance system. StpK7 negatively regulates the expression of this gene island. Following phage TM4 infection, StpK7 is induced, directly phosphorylating the transcriptional regulator MSMEG_1198 and inhibiting its positive regulatory activity, thus reducing the expression of multiple downstream genes in the BREX-like gene island. Further analysis showed that genes within this anti-phage island critically regulate mycobacterial lipid hemostasis and phage adsorption. Collectively, this work characterizes a regulatory network driven by StpK7, which is utilized by phage TM4 to escape from the host defense against mycobacteria.


Subject(s)
Bacteriophages , Mycobacterium , Bacteriophages/genetics , Bacteriophages/metabolism , Eukaryota , Protein Kinases , Eukaryotic Cells/metabolism , Mycobacterium/metabolism , Protein Serine-Threonine Kinases/genetics , Bacterial Proteins/metabolism
4.
Front Cell Infect Microbiol ; 12: 877265, 2022.
Article in English | MEDLINE | ID: mdl-35444960

ABSTRACT

The normal operation of the endoplasmic reticulum (ER) is critical for cells and organisms. However, ER stress, caused by imbalanced protein folding, occurs frequently, which perturbs the function of the ER and even results in cell apoptosis eventually. Many insults can induce ER stress; pathogen infection is one of them. Most of the genes involved in ER stress have been reported to be upregulated in Mycobacterium tuberculosis (Mtb) granulomas of humans and mice, implicating that infection with Mtb can induce ER stress. However, little is known about the molecular mechanism of Mtb induction of ER stress. Here, we reveal that Mycobacterium protein CDP-diglyceride hydrolase of Mycobacteriumn (CdhM) could target the ER and cause abnormal ER morphology and cell death. RNA-seq analysis suggests that most of the ER stress-involved genes were modulated by CdhM. Further assessed by biochemical experiments, the transcription and protein levels of ER stress markers BiP and CHOP, as well as the levels of XBP1 splicing and eIF2α phosphorylation, were significantly increased by CdhM, confirming that CdhM could induce ER stress alone or during infection. A single conserved amino acid mutant of CdhM, including L44A, G96A, H150A, and W175A, was incapable of inducing ER stress, which indicates that induction of ER stress by CdhM is specific and functional. Furthermore, CdhM-induced ER stress could also promote apoptosis of macrophages during Mtb infection. Overexpression of CdhM conferred a significant benefit for Mtb replication by releasing Mtb into extracellular during infection of macrophage in vitro, as presented in CFU assays. Overall, our study identified a novel Mtb effector protein CdhM which may promote Mtb dissemination and proliferation by induction of ER stress and apoptosis and provided new insight into the physiological significance of induction of ER stress in tuberculosis (TB) granulomas.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Apoptosis , Endoplasmic Reticulum Stress , Granuloma , Macrophages/microbiology , Mice , Mycobacterium tuberculosis/metabolism , Pyrophosphatases , Tuberculosis/microbiology
5.
Microbiol Spectr ; 10(2): e0145621, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35377187

ABSTRACT

Zinc is a microelement essential for the growth of almost all organisms, but it is toxic at high concentrations and represents an antimicrobial strategy for macrophages. Mycobacterium tuberculosis and Mycobacterium bovis are two well-known intracellular pathogens with strong environmental adaptability, including zinc toxicity. However, the signaling pathway and molecular mechanisms on sensing and resistance to zinc toxicity remains unclear in mycobacteria. Here, we first report that P1B-type ATPase CtpG acts as a zinc efflux transporter and characterize a novel CmtR-CtpG-Zn2+ regulatory pathway that enhances mycobacterial resistance to zinc toxicity. We found that zinc upregulates ctpG expression via transcription factor CmtR and stimulates the ATPase activity of CtpG. The APC residues in TM6 is essential for CtpG to export zinc and enhance M. bovis BCG resistance to zinc toxicity. During infection, CtpG inhibits zinc accumulation in the mycobacteria, and aids bacterial survival in THP-1 macrophage and mice with elevated inflammatory responses. Our findings revealed the existence of a novel regulatory pathway on mycobacteria responding to and adapting to host-mediated zinc toxicity. IMPORTANCE Tuberculosis is caused by the bacillus Mycobacterium tuberculosis and is one of the major sources of mortality. M. tuberculosis has developed unique mechanisms to adapt to host environments, including zinc deficiency and toxicity, during infection. However, the molecular mechanism by which mycobacteria promote detoxification of zinc, and the associated signaling pathways remains largely unclear. In this study, we first report that P1B-type ATPase CtpG acts as a zinc efflux transporter and characterize a novel CmtR-CtpG-Zn2+ regulatory pathway that enhances mycobacterial resistance to zinc toxicity in M. bovis. Our findings reveal the existence of a novel excess zinc-triggered signaling circuit, provide new insights into mycobacterial adaptation to the host environment during infection, and might be useful targets for the treatment of tuberculosis.


Subject(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculosis , Adenosine Triphosphatases/metabolism , Animals , Mice , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Zinc/metabolism , Zinc/toxicity
6.
Environ Microbiol ; 24(9): 4382-4400, 2022 09.
Article in English | MEDLINE | ID: mdl-35348272

ABSTRACT

During infection, intracellular pathogens inevitably face the pressure of hypoxia. Mycobacterium tuberculosis and Mycobacterium bovis represent two typical intracellular bacteria, but the signalling pathway of their adaptation to hypoxia remains unclear. Here, we report a new mechanism of the hypoxic adaptation in M. bovis driven by the second messenger molecule c-di-GMP. We found that c-di-GMP was significantly accumulated in bacterial cells under hypoxic stress and blocked the inhibitory activity of ArgR, an arginine metabolism gene cluster regulator, which increased arginine synthesis and slowed tricarboxylic acid cycle (TCA cycle) and aerobic respiration. Meanwhile, c-di-GMP relieved the self-inhibition of argR expression, and ArgR could interact with the nitrite metabolic gene regulator Cmr, promoting the positive regulation of Cmr and, thereafter, the nitrite respiration. Consistently, c-di-GMP significantly induced the expression of arginine and nitrite metabolism gene clusters and increased the mycobacterial survival ability under hypoxia. Therefore, we found a new function of the second messenger molecule c-di-GMP and characterized ArgR as a metabolic switching regulator that can coordinate the c-di-GMP signal to trigger hypoxic adaptation in mycobacteria. Our findings provide a potential new target for blocking the life cycle of M. tuberculosis infection.


Subject(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Humans , Hypoxia/genetics , Mycobacterium bovis/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Nitrites/metabolism
7.
Environ Microbiol ; 23(2): 1096-1114, 2021 02.
Article in English | MEDLINE | ID: mdl-32985741

ABSTRACT

An unusually high lipid content and a complex lipid profile are the most distinctive features of the mycobacterial cell envelope. However, our understanding of the regulatory mechanism underlying mycobacterial lipid metabolism is limited, and the major regulators responsible for lipid homeostasis remain to be characterized. Here, we identified MmbR as a novel master regulator that is essential for maintaining lipid homeostasis in Mycolicibacterium smegmatis. We found that MmbR controls fatty acid ß-oxidation and modulates biofilm formation in Mycolicibacterium smegmatis. Although MmbR possesses the properties of nucleoid-associated proteins, it acts as a TetR-like transcription factor, directly regulating and intensively repressing the expression of a group of core genes involved in fatty acid ß-oxidation. Furthermore, both long-chain acyl-Coenzyme A and fatty acids appear to regulate the signal molecules modulated by MmbR. The deletion of mmbR led to a significant reduction in intracellular fatty acid content and a decrease in the relative lipid composition of the biofilm. The lack of mmbR led to morphological changes in the mycobacterial colony, defects in biofilm formation and enhanced sensitivity to anti-tuberculosis drugs. Our study is the first to establish a link between the transcriptional regulation of fatty acid ß-oxidation genes and lipid homeostasis in mycobacteria.


Subject(s)
Bacterial Proteins/metabolism , Fatty Acids/metabolism , Lipid Metabolism/genetics , Mycobacterium smegmatis/physiology , Transcription Factors/metabolism , Acyl Coenzyme A/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Biofilms/growth & development , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Transcription Factors/genetics
8.
J Biol Chem ; 295(50): 17083-17099, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33033071

ABSTRACT

Reactive oxygen species (ROS) are an unavoidable host environmental cue for intracellular pathogens such as Mycobacterium tuberculosis and Mycobacterium bovis; however, the signaling pathway in mycobacteria for sensing and responding to environmental stress remains largely unclear. Here, we characterize a novel CmtR-Zur-ESX3-Zn2+ regulatory pathway in M. bovis that aids mycobacterial survival under oxidative stress. We demonstrate that CmtR functions as a novel redox sensor and that its expression can be significantly induced under H2O2 stress. CmtR can physically interact with the negative regulator Zur and de-represses the expression of the esx-3 operon, which leads to Zn2+ accumulation and promotion of reactive oxygen species detoxication in mycobacterial cells. Zn2+ can also act as an effector molecule of the CmtR regulator, using which the latter can de-repress its own expression for further inducing bacterial antioxidant adaptation. Consistently, CmtR can induce the expression of EsxH, a component of esx-3 operon involved in Zn2+ transportation that has been reported earlier, and inhibit phagosome maturation in macrophages. Lastly, CmtR significantly contributes to bacterial survival in macrophages and in the lungs of infected mice. Our findings reveal the existence of an antioxidant regulatory pathway in mycobacteria and provide novel information on stress-triggered gene regulation and its association with host-pathogen interaction.


Subject(s)
Bacterial Proteins/metabolism , Microbial Viability , Mycobacterium bovis/metabolism , Oxidative Stress , Transcription Factors/metabolism , Zinc/metabolism , Bacterial Proteins/genetics , Mycobacterium bovis/genetics , Transcription Factors/genetics
9.
PLoS One ; 15(4): e0230786, 2020.
Article in English | MEDLINE | ID: mdl-32302313

ABSTRACT

Mycobacterium tuberculosis is the causative agent of tuberculosis and has evolved an ability to survive in hostile host environments. M. tuberculosis is thought to utilize the rTCA cycle to sustain its latent growth during infection, but the enzymatic characteristics and physiological function for the key citrate lyase of the rTCA cycle, MtbCitE, in the important pathogen remain unclear. In this study, we investigated the function of MtbCitE based on its structural properties and sequence comparisons with other bacterial citrate lyase subunits. We showed that several amino acid residues were important for the citrate cleavage activity of MtbCitE. Strikingly, the citrate cleavage activity of MtbCitE was inhibited by ATP, indicating that energy metabolism might couple with the regulation of MtbCitE activity, which differed from other CitEs. More interestingly, deletion of citE from Mycobacterium bovis BCG decreased the mycobacterial survival rate under hypoxic conditions, whereas complementation with citE restored the phenotype to wild-type levels. Consistently, three key rTCA cycle enzymes were positively regulated under hypoxic conditions in mycobacteria. Therefore, we characterized a unique citrate lyase MtbCitE from M. tuberculosis and found that the CitE protein significantly contributed to mycobacterial survival under hypoxic conditions.


Subject(s)
Bacterial Proteins/metabolism , Hypoxia/metabolism , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Oxo-Acid-Lyases/metabolism , Tuberculosis/microbiology , Amino Acid Sequence , Animals , Cell Line , Mice , Mycobacterium bovis/metabolism , Mycobacterium bovis/pathogenicity , RAW 264.7 Cells , Survival Rate , Virulence/physiology
10.
J Biol Chem ; 294(34): 12729-12742, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31270210

ABSTRACT

Cyclic di-GMP (c-di-GMP) is an important second messenger in bacteria, and its regulatory network has been extensively studied. However, information regarding the activation mechanisms of its receptors remains limited. In this study, we characterized the two-component regulator DevR as a new c-di-GMP receptor and further uncovered a novel co-activation mechanism for effective regulation of DevR in mycobacteria. We show that high c-di-GMP levels induce the expression of the devR operon in Mycobacterium smegmatis and increase mycobacterial survival under oxidative stress. The deletion of either DevR or its two-component kinase DevS significantly weakened the stimulating effect of c-di-GMP on oxidative-stress tolerance of mycobacteria. We also found that DevR senses the c-di-GMP signal through its C-terminal structure and that c-di-GMP alone does not directly affect the DNA-binding activity of DevR. Strikingly, c-di-GMP stimulated DevR phosphorylation by the kinase DevS, thereby activating DevR's DNA-binding affinity. In summary, our results indicated that c-di-GMP triggers a phosphorylation-dependent mechanism that co-activates DevR's transcriptional activity. Our findings suggest a novel paradigm for the cross-talk between c-di-GMP signaling and two-component regulatory systems that activates transcription of stress-response genes in bacteria.


Subject(s)
Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Mycobacterium smegmatis/metabolism , Oxidative Stress , Bacterial Proteins/genetics , Cyclic GMP/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
PLoS One ; 14(6): e0218449, 2019.
Article in English | MEDLINE | ID: mdl-31199855

ABSTRACT

Cordycepin is an efficient component of Cordyceps spp, a traditional Chinese medicine widely used for healthcare in China, and has been recently acted as a strong anticancer agent for clinic. However, whether and how it may play a role in combating tuberculosis, caused by Mycobacterium tuberculosis, remains unknown. Here we report that cordycepin can kill Mycobacterium by hijacking the bacterial adenosine kinase (AdoK), a purine salvage enzyme responsible for the phosphorylation of adenosine (Ado) to adenosine monophosphate (AMP). We show that cordycepin is a poor AdoK substrate but it competitively inhibits the catalytic activity of AdoK for adenosine phosphorylation. Cordycepin does not affect the activity of the human adenosine kinase (hAdoK), whereas hAdoK phosphorylates cordycepin to produce a new monophosphate derivative. Co-use of cordycepin and deoxycoformycin, an inhibitor of adenosine deaminase (ADD), more efficiently kills M. bovis and M. tuberculosis. The add-deleted mycobacterium is more sensitive to cordycepin. This study characterized cordycepin as a new mycobactericidal compound and also uncovered a potential anti-mycobacterial mechanism.


Subject(s)
Adenosine Kinase/antagonists & inhibitors , Antitubercular Agents/pharmacology , Deoxyadenosines/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Antitubercular Agents/chemistry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Deoxyadenosines/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Mutation , Mycobacterium tuberculosis/genetics , Polymorphism, Single Nucleotide , Tandem Mass Spectrometry
12.
Commun Biol ; 2: 65, 2019.
Article in English | MEDLINE | ID: mdl-30793043

ABSTRACT

Hostile environmental cues cause Mycobacterium tuberculosis to enter a state of slow growth for survival. However, the underlying regulatory mechanism remains unclear. DnaA is essential for DNA replication initiation and represents an efficient target for growth regulation in bacteria. Here, we show that the nucleoid-associated protein NapM is a DnaA antagonist, protecting M. tuberculosis from stress-mediated killing. NapM can be induced by diverse stressful signals. It binds to DnaA to inhibit both its DNA replication origin-binding and ATP hydrolysis activity. As a DnaA antagonist, NapM inhibits the mycobacterial DNA synthesis in vitro and in vivo in M. tuberculosis. Furthermore, we show that NapM contributes to the survival of M. tuberculosis under stress and within macrophages during infection. Our findings provide a previously unidentified mechanism of mycobacterial survival under stress and also suggest NapM as a potential drug target for tuberculosis control.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , Stress, Physiological/physiology , Tuberculosis/metabolism , Bacterial Proteins/genetics , DNA Replication/genetics , DNA, Bacterial/genetics , Macrophages/microbiology , Microbial Viability/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology
13.
J Genet Genomics ; 46(12): 575-589, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31974004

ABSTRACT

Mycobacterium tuberculosis possesses unique cellular envelope components that contribute to bacterial escape from host immune surveillance. Phosphatidylinositol mannosides (PIMs) and their higher derivatives are important molecules implicated in host-pathogen interactions in the course of tuberculosis. However, the biosynthetic regulation of these specific lipids and its effect on the bacterial fate in the infected host remain unclear. Here, we show that a hypothetical M. tuberculosis transcriptional factor designated as MpbR negatively regulates two transporter genes and affects mycobacterial PIM biosynthesis and biofilm formation. MpbR inhibits the accumulation of acylated PIM lipids and triggers the mycobacterium to reduce the production of reactive oxygen species and NO during infection, which enhances the survival of M. tuberculosis in macrophages. MpbR deletion reduces M. tuberculosis lung burdens and inflammation of infected mice. These findings provide new insights into the regulation of mycobacterial lipid metabolism and its correlation with pathogenesis of M. tuberculosis.


Subject(s)
Bacterial Proteins/metabolism , Immunity, Innate/physiology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Line , Female , Host-Pathogen Interactions , Humans , Lipid Metabolism/physiology , Macrophages/metabolism , Mice , Phosphatidylinositols/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Transcription Factors/genetics
14.
J Innate Immun ; 11(3): 242-248, 2019.
Article in English | MEDLINE | ID: mdl-30253386

ABSTRACT

Cyclic dimeric guanosine monophosphate (c-di-GMP) is a universally conserved second messenger that contributes to the pathogenicity of numerous bacterial species. In recent years, growing evidence has shown that bacterial extracellular c-di-GMP can interact with the innate immune system and regulate host immune responses. This review summarizes our current understanding on the dual roles of bacterial c-di-GMP in pathogen-host interaction: activation of the antibacterial innate immune response through the cytosolic surveillance pathway and inhibition of innate immune defense for iron restriction.


Subject(s)
Cyclic GMP/analogs & derivatives , Host-Pathogen Interactions , Immunity, Innate , Cyclic GMP/physiology , DEAD-box RNA Helicases/physiology , Humans , Lipocalin-2/physiology , Membrane Proteins/physiology
15.
J Genet Genomics ; 45(9): 477-488, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30279094

ABSTRACT

Biofilm formation has been implicated to be tightly regulated in bacteria. Mycobacterial species possess a unique cell-wall structure; however, the underlying regulation mechanism for their biofilm formation remains largely unclear. In this study, we characterized a hypothetical mannitol metabolism and transportation gene cluster (Ms5571-Ms5576), designated as mmt operon, whose expression significantly contributes to the biofilm formation in Mycobacterium smegmatis. We showed that in the operon the Ms5575 gene encodes a GntR-like transcriptional repressor and the Ms5576 gene encodes a mannitol 2-dehydrogenase which can produce D-mannitol from D-mannose. Strikingly, the D-mannitol molecule can derepress the negative regulation of Ms5575 on the mmt operon to stimulate the operon's expression. Consistently, addition of D-mannitol into the medium can obviously induce mycobacterial biofilm formation. Furthermore, we found that Ms0179 positively regulates the mmt operon through its downstream regulator Ms0180. Ms0180 directly binds the mmt operon to positively regulate its expression. Both Ms0179 and Ms0180 significantly affect the mycobacterial biofilm formation. Taken together, we explored a regulatory pathway for the mannitol metabolism and its coordination with the biofilm formation in M. smegmatis. This finding provides novel insights into the unique mechanism of biofilm formation regulation in mycobacteria.


Subject(s)
Bacterial Proteins/genetics , Mannitol/metabolism , Mycobacterium smegmatis/physiology , Bacterial Proteins/metabolism , Biofilms/growth & development , Gene Expression Regulation, Bacterial , Multigene Family , Operon , Transcription Factors/genetics , Transcription Factors/metabolism
16.
J Biol Chem ; 293(43): 16741-16750, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30185616

ABSTRACT

Isoniazid (INH) and ethambutol (EMB) are two major first-line drugs for managing tuberculosis (TB), caused by the microbe Mycobacterium tuberculosis Although co-use of these two drugs is common in clinical practice, the mechanism for the potential synergistic interplay between them remains unclear. Here, we present first evidence that INH and EMB act synergistically through a transcriptional repressor of the inhA gene, the target gene of INH encoding an enoyl-acyl carrier protein reductase of the fatty acid synthase type II system required for bacterial cell wall integrity. We report that EMB binds a hypothetical transcription factor encoded by the Rv0273c gene, designated here as EtbR. Using DNA footprinting, we found that EtbR specifically recognizes a motif sequence in the upstream region of the inhA gene. Using isothermal titration calorimetry and surface plasmon resonance assays, we observed that EMB binds EtbR in a 1:1 ratio and thereby stimulates its DNA-binding activity. When a nonlethal dose of EMB was delivered in combination with INH, EMB increased the INH susceptibility of cultured M. tuberculosis cells. In summary, EMB induces EtbR-mediated repression of inhA and thereby enhances the mycobactericidal effect of INH. Our findings uncover a molecular mechanism for the synergistic activity of two important anti-TB drugs.


Subject(s)
Antitubercular Agents/pharmacology , Drug Synergism , Ethambutol/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Therapy, Combination , Humans , Mycobacterium tuberculosis/physiology , Tuberculosis/metabolism , Tuberculosis/microbiology
17.
Nucleic Acids Res ; 46(14): 7270-7283, 2018 08 21.
Article in English | MEDLINE | ID: mdl-29982829

ABSTRACT

Cyclic diguanylate monophosphate (c-di-GMP) is a global signaling molecule that modulates diverse cellular processes through its downstream receptors. However, no study has fully clarified the mechanisms by which c-di-GMP organizes functionally divergent regulators to drive the gene expression for coping with environmental stress. Here, we reported that c-di-GMP can integrate two functionally opposite receptor transcription factors, namely, LtmA and HpoR, into a pathway to regulate the antioxidant processes in Mycobacterium smegmatis. In contrast to HpoR, LtmA is an activator that positively regulates the expression of redox gene clusters and the mycobacterial H2O2 resistance. LtmA can physically interact with HpoR. A high level of c-di-GMP stimulates the positive regulation of LtmA and boosts the physical interaction between the two regulators, further enhancing the DNA-binding ability of LtmA and reducing the inhibitory activity of HpoR. Therefore, upon exposure to oxidative stress, c-di-GMP can orchestrate functionally divergent transcription factors to trigger antioxidant defense in mycobacteria. This finding presents a noteworthy example of how a bacterium remodels its transcriptional network via c-di-GMP in response to environmental stress.


Subject(s)
Antioxidants/metabolism , Bacterial Proteins/genetics , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial , Signal Transduction/genetics , Transcription Factors/metabolism , Bacterial Proteins/metabolism , Cyclic GMP/metabolism , Hydrogen Peroxide/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Oxidants/metabolism , Protein Binding
18.
Nucleic Acids Res ; 46(7): 3595-3611, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29490073

ABSTRACT

Cyclic di-GMP (c-di-GMP) is a global signaling molecule that widely modulates diverse cellular processes. However, whether or not the c-di-GMP signal participates in regulation of bacterial antioxidant defense is unclear, and the involved regulators remain to be explored. In this study, we characterized HpoR as a novel c-di-GMP effective transcription factor and found a link between the c-di-GMP signal and the antioxidant regulation in Mycobacterium smegmatis. H2O2 stress induces c-di-GMP accumulation in M. smegmatis. High level of c-di-GMP triggers expression of a redox gene cluster, designated as hpoR operon, which is required for the mycobacterial H2O2 resistance. HpoR acts as an inhibitor of the hpoR operon and recognizes a 12-bp motif sequence within the upstream regulatory region of the operon. c-di-GMP specifically binds with HpoR at a ratio of 1:1. Low concentrations of c-di-GMP stimulate the DNA-binding activity of HpoR, whereas high concentrations of the signal molecule inhibit the activity. Strikingly, high level of c-di-GMP de-represses the intracellular association of HpoR with the regulatory region of the hpoR operon in M. smegmatis and enhances the mycobacterial H2O2 resistance. Therefore, we report a novel c-di-GMP effective regulator in mycobacteria, which extends the second messenger's function to bacterial antioxidant defense.


Subject(s)
Antioxidants/pharmacology , Biofilms/drug effects , Mycobacterium smegmatis/genetics , Second Messenger Systems/genetics , Cyclic GMP/analogs & derivatives , Cyclic GMP/chemistry , Cyclic GMP/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Bacterial/drug effects , Hydrogen Peroxide/pharmacology , Mycobacterium smegmatis/drug effects , Operon/genetics , Promoter Regions, Genetic , Signal Transduction/drug effects
19.
Sci Rep ; 6: 27489, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27271013

ABSTRACT

Transport-related genes significantly affect bacterial antibiotic resistance. However, the effects of these genes and their regulation of bacterial drug resistance in several mycobacterial species, including the fast-growing Mycobacterium smegmatis, the pathogen M. tuberculosis and M. avium have not been clearly characterized. We identified Ms4022 (MSMEG_4022) as a novel TetR family regulator that activates the expression of seven transport-related genes and affects drug resistance in M. smegmatis. Overexpression of Ms4022 inhibited M. smegmatis growth and enhanced mycobacterial resistance to the anti-tuberculosis drug rifampicin (RIF). By contrast, the Ms4022-deleted mycobacterial strain has shown sensitive to RIF. Ms4022 recognized three 19 bp non-palindromic motifs containing a 9 bp conserved region at their 5' end and it directly regulated seven transport-related genes, which affects mycobacterial resistance to RIF. Overexpression of three of seven transport-related genes (Ms1448, Ms1613, and Ms5278) inhibited the growth of M. smegmatis. This study improves our understanding of the function of mycobacterial transport-related genes and their regulation of bacterial drug resistance.


Subject(s)
Bacterial Proteins/physiology , Drug Resistance, Bacterial/physiology , Gene Expression Regulation, Bacterial/physiology , Genes, Bacterial , Mycobacterium smegmatis/drug effects , Rifampin/pharmacology , Binding Sites , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Mycobacterium smegmatis/genetics , Operon , Real-Time Polymerase Chain Reaction
20.
Mol Microbiol ; 101(1): 167-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27010232

ABSTRACT

Nucleoid-associated proteins (NAPs) play important roles in the global organization of bacterial chromosomes. However, potential NAPs and their functions are barely characterized in mycobacteria. In this study, NapM, an alkaline protein, functions as a new NAP. NapM is conserved in all of the sequenced mycobacterial genomes, and can recognize DNA in a length-dependent but sequence-independent manner. It prefers AT-rich DNA and binds to the major groove. NapM possesses a clear DNA-bridging function, and can protect DNA from DNase I digestion. NapM globally regulates the expression of more than 150 genes and the resistance of Mycobacterium smegmatis to two anti-tuberculosis drugs, namely, rifampicin and ethambutol. An ABC transporter operon was found to be specifically responsible for the napM-dependent ethambutol resistance of M. smegmatis. NapM also presents a similar regulation of anti-tuberculosis drug resistance in M. tuberculosis. These results suggest that NapM is a new member of the mycobacterial NAP family. Our findings expand the range of identified NAPs and improve the understanding on the relationship between NAPs with antibiotic resistance in mycobacteria.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium/genetics , ATP-Binding Cassette Transporters/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/genetics , Drug Resistance, Microbial , Gene Expression , Mycobacterium/drug effects , Mycobacterium smegmatis/genetics , Mycobacterium tuberculosis/genetics , Operon/genetics , Tuberculosis/drug therapy , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...