scholarly journals An outer membrane channel protein of Mycobacterium tuberculosis with exotoxin activity

2014 ◽  
Vol 111 (18) ◽  
pp. 6750-6755 ◽  
Author(s):  
O. Danilchanka ◽  
J. Sun ◽  
M. Pavlenok ◽  
C. Maueroder ◽  
A. Speer ◽  
...  
2015 ◽  
Vol 59 (4) ◽  
pp. 2328-2336 ◽  
Author(s):  
Olga Danilchanka ◽  
David Pires ◽  
Elsa Anes ◽  
Michael Niederweis

ABSTRACTMycobacterium tuberculosis, the causative agent of tuberculosis, is protected from toxic solutes by an effective outer membrane permeability barrier. Recently, we showed that the outer membrane channel protein CpnT is required for efficient nutrient uptake byM. tuberculosisandMycobacterium bovisBCG. In this study, we found that thecpnTmutant ofM. bovisBCG is more resistant than the wild type to a large number of drugs and antibiotics, including rifampin, ethambutol, clarithromycin, tetracycline, and ampicillin, by 8- to 32-fold. Furthermore, thecpnTmutant ofM. bovisBCG was 100-fold more resistant to nitric oxide, a major bactericidal agent required to controlM. tuberculosisinfections in mice. Thus, CpnT constitutes the first outer membrane susceptibility factor in slow-growing mycobacteria. The dual functions of CpnT in uptake of nutrients and mediating susceptibility to toxic molecules are reflected in macrophage infection experiments: while loss of CpnT was detrimental forM. bovisBCG in macrophages that enable bacterial replication, presumably due to inadequate nutrient uptake, it conferred a survival advantage in macrophages that mount a strong bactericidal response. Importantly, thecpnTgene showed a significantly higher density of nonsynonymous mutations in drug-resistant clinicalM. tuberculosisstrains, indicating that CpnT is under selective pressure in human tuberculosis and/or during chemotherapy. Our results indicate that the CpnT channel constitutes an outer membrane gateway controlling the influx of nutrients and toxic molecules into slow-growing mycobacteria. This study revealed that reducing protein-mediated outer membrane permeability might constitute a new drug resistance mechanism in slow-growing mycobacteria.


2003 ◽  
Vol 43 (supplement) ◽  
pp. S49
Author(s):  
K. Seki ◽  
A. Suenaga ◽  
T. Narumi ◽  
M. Taiji ◽  
C. Danelon ◽  
...  

2019 ◽  
Vol 55 (38) ◽  
pp. 5431-5434 ◽  
Author(s):  
Deepak Anand ◽  
Gaurao V. Dhoke ◽  
Julia Gehrmann ◽  
Tayebeh M. Garakani ◽  
Mehdi D. Davari ◽  
...  

Development of a novel whole cell system for chiral separation of arginine enantiomers through an engineered outer membrane channel protein ferric hydroxamate uptake protein A (FhuA).


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Uday Tak ◽  
Terje Dokland ◽  
Michael Niederweis

AbstractMycobacterium tuberculosis secretes the tuberculosis necrotizing toxin (TNT) to kill host cells. Here, we show that the WXG100 proteins EsxE and EsxF are essential for TNT secretion. EsxE and EsxF form a water-soluble heterodimer (EsxEF) that assembles into oligomers and long filaments, binds to membranes, and forms stable membrane-spanning channels. Electron microscopy of EsxEF reveals mainly pentameric structures with a central pore. Mutations of both WXG motifs and of a GXW motif do not affect dimerization, but abolish pore formation, membrane deformation and TNT secretion. The WXG/GXW mutants are locked in conformations with altered thermostability and solvent exposure, indicating that the WXG/GXW motifs are molecular switches controlling membrane interaction and pore formation. EsxF is accessible on the bacterial cell surface, suggesting that EsxEF form an outer membrane channel for toxin export. Thus, our study reveals a protein secretion mechanism in bacteria that relies on pore formation by small WXG proteins.


Sign in / Sign up

Export Citation Format

Share Document