Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Microbiol ; 11: 572419, 2020.
Article in English | MEDLINE | ID: mdl-33224116

ABSTRACT

One of the most important stress responses in bacteria is the stringent response. The main player in this response is the signal molecule (p)ppGpp, which is synthesized by a Rel family protein. In Escherichia coli, RelA is the main synthetase of (p)ppGpp in response to amino acid starvation. Although the synthetic activity of RelA is well-understood, its regulation is not yet fully characterized. The C-terminus domain (CTD) of the E. coli RelA is responsible for the regulation of the protein and for its complete dependency on wild-type (WT) ribosome. The CTD contains three Cysteine residues, positioned in a very conserved order. Together with our previous results, we show in vitro the negative dominant effect of a part of the WT CTD (AA 564-744) named YG4 on RelA synthetic activity. This effect is abolished using mutated YG4 (YG4-638). In vitro and mass spectrometry (MS)-MS analysis of the native RelA and the mutated RelA in Cys-638 (Rel638) in the presence of the native and mutated YG4 (YG4-638) reveals that RelA forms a homodimer via its CTD by the formation of a disulfide bond between the two Cys-638 residues. This supports our previous data which showed, using a two-hybrid system, interactions between RelA proteins via the CTD. Finally, we show in vitro that excess of the native YG4 inhibited RelA synthetic activity but did not affect the amount of RelA bound to the ribosome. Our results suggest that the regulatory mechanism of RelA is by the dimerization of the protein via disulfide bonds in the CTD. Upon amino-acid starvation, the dimer changes its conformation, thus activating the stringent response in the cell.

2.
Nat Commun ; 4: 3001, 2013.
Article in English | MEDLINE | ID: mdl-24343429

ABSTRACT

Bacterial persistence has been shown to be an underlying factor in the failure of antibiotic treatments. Although many pathways, among them the stringent response and toxin-antitoxin modules, have been linked to antibiotic persistence, a clear molecular mechanism for the growth arrest that characterizes persistent bacteria remained elusive. Here, we screened an expression library for putative targets of HipA, the first toxin linked to persistence, and a serine/threonine kinase. We found that the expression of GltX, the glutamyl-tRNA-synthetase, reverses the toxicity of HipA and prevents persister formation. We show that upon HipA expression, GltX undergoes phosphorylation at Ser239, its ATP-binding site. This phosphorylation leads to accumulation of uncharged tRNA(Glu) in the cell, which results in the activation of the stringent response. Our findings demonstrate a mechanism for persister formation by the hipBA toxin-antitoxin module and provide an explanation for the long-observed connection between persistence and the stringent response.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Glutamate-tRNA Ligase/metabolism , Adenosine Triphosphate/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Escherichia coli/drug effects , Gene Library , Phenotype , Phosphorylation , Serine/metabolism , Time Factors
3.
Eur J Med Chem ; 70: 497-504, 2013.
Article in English | MEDLINE | ID: mdl-24189495

ABSTRACT

Rel proteins in bacteria synthesize the signal molecules (p)ppGpp that trigger the Stringent Response, responsible for bacterial survival. Inhibiting the activity of such enzymes prevents the Stringent Response, resulting in the inactivation of long-term bacterial survival strategies, leading to bacterial cell death. Herein, we describe a series of deoxyguanosine-based analogs of the Relacin molecule that inhibit in vitro the synthetic activity of Rel proteins from Gram positive and Gram negative bacteria, providing a deeper insight on the SAR for a better understanding of their potential interactions and inhibitory activity. Among the inhibitors evaluated, compound 2d was found to be more effective and potent than our previously reported Relacin.


Subject(s)
Deoxyguanosine/analogs & derivatives , Dipeptides/pharmacology , Drug Design , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Deinococcus/chemistry , Deoxyguanosine/chemical synthesis , Deoxyguanosine/chemistry , Deoxyguanosine/pharmacology , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dose-Response Relationship, Drug , Escherichia coli/chemistry , Molecular Structure , Proto-Oncogene Proteins c-rel/isolation & purification , Structure-Activity Relationship
4.
PLoS Pathog ; 8(9): e1002925, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028324

ABSTRACT

Finding bacterial cellular targets for developing novel antibiotics has become a major challenge in fighting resistant pathogenic bacteria. We present a novel compound, Relacin, designed to inhibit (p)ppGpp production by the ubiquitous bacterial enzyme RelA that triggers the Stringent Response. Relacin inhibits RelA in vitro and reduces (p)ppGpp production in vivo. Moreover, Relacin affects entry into stationary phase in Gram positive bacteria, leading to a dramatic reduction in cell viability. When Relacin is added to sporulating Bacillus subtilis cells, it strongly perturbs spore formation regardless of the time of addition. Spore formation is also impeded in the pathogenic bacterium Bacillus anthracis that causes the acute anthrax disease. Finally, the formation of multicellular biofilms is markedly disrupted by Relacin. Thus, we establish that Relacin, a novel ppGpp analogue, interferes with bacterial long term survival strategies, placing it as an attractive new antibacterial agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Bacillus subtilis/drug effects , Deoxyguanosine/analogs & derivatives , Dipeptides/pharmacology , Guanosine Tetraphosphate/metabolism , Ligases/antagonists & inhibitors , Bacillus anthracis/metabolism , Bacillus anthracis/physiology , Bacillus subtilis/metabolism , Bacillus subtilis/physiology , Biofilms/drug effects , Deoxyguanosine/chemistry , Deoxyguanosine/pharmacology , Dipeptides/chemistry , Gene Expression Regulation, Bacterial/drug effects , Ligases/metabolism , Microbial Sensitivity Tests , Spores, Bacterial/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL