scholarly journals The Mce3R stress-resistance pathway is vulnerable to small-molecule targeting that improves tuberculosis drug activities

2019 ◽  
Author(s):  
Xinxin Yang ◽  
Tianao Yuan ◽  
Rui Ma ◽  
Kieran Chacko ◽  
Melissa Smith ◽  
...  

<p>One-third of the world’s population carries <i>Mycobacterium tuberculosis </i>(<i>Mtb</i>), the infectious agent that causes tuberculosis (TB), and every 17 seconds someone dies of TB. After infection, <i>Mtb</i>can live dormant within macrophages for decades in a granuloma structure arising from the host immune response; and cholesterol is important for this persistence of <i>Mtb</i>. Current treatments require long-duration drug regimens with many associated toxicities, which are compounded by the high doses required. We phenotypically screened 35 6-azasteroid analogues against <i>Mtb</i>and found that at low micromolar concentrations, a subset of the <a>analogues sensitized <i>Mtb</i></a>to multiple TB drugs. Two analogues were selected for further study to characterize the bactericidal activity of bedaquiline and isoniazid under normoxic and low-oxygen conditions. These two 6-azasteroids <a>showed strong synergy with bedaquiline</a>(fractional inhibitory concentration index = 0.21, bedaquiline minimal inhibitory concentration = 16 nM at 1 μM 6-azasteroid). The rate at which spontaneous resistance to one of the 6-azasteroids arose in the presence of bedaquiline was approximately 10<sup>−9</sup>, and the 6-azasteroid-resistant mutants retained their isoniazid and bedaquiline sensitivity. Genes in the cholesterol-regulated Mce3R regulon were required for 6-azasteroid activity, whereas genes in the cholesterol catabolism pathway were not. Expression of a subset of Mce3R genes was down-regulated upon 6-azasteroid treatment. The Mce3R regulon is implicated in stress resistance and is absent in saprophytic mycobacteria. This regulon encodes a cholesterol-regulated stress-resistance pathway that we conclude is important for pathogenesis and contributes to drug tolerance, and that this pathway is vulnerable to small-molecule targeting in live mycobacteria.</p>

2019 ◽  
Author(s):  
Xinxin Yang ◽  
Tianao Yuan ◽  
Rui Ma ◽  
Kieran Chacko ◽  
Melissa Smith ◽  
...  

<p>One-third of the world’s population carries <i>Mycobacterium tuberculosis </i>(<i>Mtb</i>), the infectious agent that causes tuberculosis (TB), and every 17 seconds someone dies of TB. After infection, <i>Mtb</i>can live dormant within macrophages for decades in a granuloma structure arising from the host immune response; and cholesterol is important for this persistence of <i>Mtb</i>. Current treatments require long-duration drug regimens with many associated toxicities, which are compounded by the high doses required. We phenotypically screened 35 6-azasteroid analogues against <i>Mtb</i>and found that at low micromolar concentrations, a subset of the <a>analogues sensitized <i>Mtb</i></a>to multiple TB drugs. Two analogues were selected for further study to characterize the bactericidal activity of bedaquiline and isoniazid under normoxic and low-oxygen conditions. These two 6-azasteroids <a>showed strong synergy with bedaquiline</a>(fractional inhibitory concentration index = 0.21, bedaquiline minimal inhibitory concentration = 16 nM at 1 μM 6-azasteroid). The rate at which spontaneous resistance to one of the 6-azasteroids arose in the presence of bedaquiline was approximately 10<sup>−9</sup>, and the 6-azasteroid-resistant mutants retained their isoniazid and bedaquiline sensitivity. Genes in the cholesterol-regulated Mce3R regulon were required for 6-azasteroid activity, whereas genes in the cholesterol catabolism pathway were not. Expression of a subset of Mce3R genes was down-regulated upon 6-azasteroid treatment. The Mce3R regulon is implicated in stress resistance and is absent in saprophytic mycobacteria. This regulon encodes a cholesterol-regulated stress-resistance pathway that we conclude is important for pathogenesis and contributes to drug tolerance, and that this pathway is vulnerable to small-molecule targeting in live mycobacteria.</p>


2019 ◽  
Author(s):  
Xinxin Yang ◽  
Tianao Yuan ◽  
Rui Ma ◽  
Kieran Chacko ◽  
Melissa Smith ◽  
...  

<p>One-third of the world’s population carries <i>Mycobacterium tuberculosis </i>(<i>Mtb</i>), the infectious agent that causes tuberculosis (TB), and every 17 seconds someone dies of TB. After infection, <i>Mtb</i>can live dormant within macrophages for decades in a granuloma structure arising from the host immune response; and cholesterol is important for this persistence of <i>Mtb</i>. Current treatments require long-duration drug regimens with many associated toxicities, which are compounded by the high doses required. We phenotypically screened 35 6-azasteroid analogues against <i>Mtb</i>and found that at low micromolar concentrations, a subset of the <a>analogues sensitized <i>Mtb</i></a>to multiple TB drugs. Two analogues were selected for further study to characterize the bactericidal activity of bedaquiline and isoniazid under normoxic and low-oxygen conditions. These two 6-azasteroids <a>showed strong synergy with bedaquiline</a>(fractional inhibitory concentration index = 0.21, bedaquiline minimal inhibitory concentration = 16 nM at 1 μM 6-azasteroid). The rate at which spontaneous resistance to one of the 6-azasteroids arose in the presence of bedaquiline was approximately 10<sup>−9</sup>, and the 6-azasteroid-resistant mutants retained their isoniazid and bedaquiline sensitivity. Genes in the cholesterol-regulated Mce3R regulon were required for 6-azasteroid activity, whereas genes in the cholesterol catabolism pathway were not. Expression of a subset of Mce3R genes was down-regulated upon 6-azasteroid treatment. The Mce3R regulon is implicated in stress resistance and is absent in saprophytic mycobacteria. This regulon encodes a cholesterol-regulated stress-resistance pathway that we conclude is important for pathogenesis and contributes to drug tolerance, and that this pathway is vulnerable to small-molecule targeting in live mycobacteria.</p>


2019 ◽  
Vol 5 (7) ◽  
pp. 1239-1251 ◽  
Author(s):  
Xinxin Yang ◽  
Tianao Yuan ◽  
Rui Ma ◽  
Kieran I. Chacko ◽  
Melissa Smith ◽  
...  

Genes ◽  
2021 ◽  
Vol 12 (2) ◽  
pp. 225
Author(s):  
Lei Xuan ◽  
Jianfeng Hua ◽  
Fan Zhang ◽  
Zhiquan Wang ◽  
Xiaoxiao Pei ◽  
...  

The Taxodium hybrid ‘Zhongshanshan 406’ (T. hybrid ‘Zhongshanshan 406’) [Taxodium mucronatum Tenore × Taxodium distichum (L.). Rich] has an outstanding advantage in flooding tolerance and thus has been widely used in wetland afforestation in China. Alcohol dehydrogenase genes (ADHs) played key roles in ethanol metabolism to maintain energy supply for plants in low-oxygen conditions. Two ADH genes were isolated and characterized—ThADH1 and ThADH4 (GenBank ID: AWL83216 and AWL83217—basing on the transcriptome data of T. hybrid ‘Zhongshanshan 406’ grown under waterlogging stress. Then the functions of these two genes were investigated through transient expression and overexpression. The results showed that the ThADH1 and ThADH4 proteins both fall under ADH III subfamily. ThADH1 was localized in the cytoplasm and nucleus, whereas ThADH4 was only localized in the cytoplasm. The expression of the two genes was stimulated by waterlogging and the expression level in roots was significantly higher than those in stems and leaves. The respective overexpression of ThADH1 and ThADH4 in Populus caused the opposite phenotype, while waterlogging tolerance of the two transgenic Populus significantly improved. Collectively, these results indicated that genes ThADH1 and ThADH4 were involved in the tolerance and adaptation to anaerobic conditions in T. hybrid ‘Zhongshanshan 406’.


2015 ◽  
Vol 173 ◽  
pp. 827-837 ◽  
Author(s):  
Domien De Paepe ◽  
Katleen Coudijzer ◽  
Bart Noten ◽  
Dirk Valkenborg ◽  
Kelly Servaes ◽  
...  

BMC Cancer ◽  
2014 ◽  
Vol 14 (1) ◽  
Author(s):  
An Wouters ◽  
Bea Pauwels ◽  
Natalie Burrows ◽  
Marc Baay ◽  
Vanessa Deschoolmeester ◽  
...  

2017 ◽  
Vol 23 (6) ◽  
pp. 2321-2330 ◽  
Author(s):  
Marcelo E. Lagos ◽  
Diego R. Barneche ◽  
Craig R. White ◽  
Dustin J. Marshall

Sign in / Sign up

Export Citation Format

Share Document