Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2023 Nov 19.
Article in English | MEDLINE | ID: mdl-38014062

ABSTRACT

Human challenge experiments could greatly accelerate the development of a tuberculosis (TB) vaccine. Human challenge for tuberculosis requires a strain that can both replicate in the host and be reliably cleared. To accomplish this, we designed Mycobacterium tuberculosis (Mtb) strains featuring up to three orthogonal kill switches, tightly regulated by exogenous tetracyclines and trimethoprim. The resultant strains displayed immunogenicity and antibiotic susceptibility similar to wild-type Mtb under permissive conditions. In the absence of supplementary exogenous compounds, the strains were rapidly killed in axenic culture, mice and nonhuman primates. Notably, the strain that contained three kill switches had an escape rate of less than 10 -10 per genome per generation and displayed no relapse in a SCID mouse model. Collectively, these findings suggest that this engineered Mtb strain could be a safe and effective candidate for a human challenge model.

2.
Proc Natl Acad Sci U S A ; 119(15): e2201632119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35380903

ABSTRACT

Current chemotherapy against Mycobacterium tuberculosis (Mtb), an important human pathogen, requires a multidrug regimen lasting several months. While efforts have been made to optimize therapy by exploiting drug­drug synergies, testing new drug combinations in relevant host environments remains arduous. In particular, host environments profoundly affect the bacterial metabolic state and drug efficacy, limiting the accuracy of predictions based on in vitro assays alone. In this study, we utilized conditional Mtb knockdown mutants of essential genes as an experimentally tractable surrogate for drug treatment and probe the relationship between Mtb carbon metabolism and chemical­genetic interactions (CGIs). We examined the antitubercular drugs isoniazid, rifampicin, and moxifloxacin and found that CGIs are differentially responsive to the metabolic state, defining both environment-independent and -dependent interactions. Specifically, growth on the in vivo­relevant carbon source, cholesterol, reduced rifampicin efficacy by altering mycobacterial cell surface lipid composition. We report that a variety of perturbations in cell wall synthesis pathways restore rifampicin efficacy during growth on cholesterol, and that both environment-independent and cholesterol-dependent in vitro CGIs could be leveraged to enhance bacterial clearance in the mouse infection model. Our findings present an atlas of chemical­genetic­environmental interactions that can be used to optimize drug­drug interactions, as well as provide a framework for understanding in vitro correlates of in vivo efficacy.


Subject(s)
Antitubercular Agents , Carbon , Cell Wall , Drug Interactions , Gene-Environment Interaction , Mycobacterium tuberculosis , Antitubercular Agents/pharmacology , Carbon/metabolism , Cell Wall/ultrastructure , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/ultrastructure
3.
China CDC Wkly ; 3(12): 256-259, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-34594861
4.
Nat Commun ; 11(1): 1960, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327655

ABSTRACT

Gene rv3722c of Mycobacterium tuberculosis is essential for in vitro growth, and encodes a putative pyridoxal phosphate-binding protein of unknown function. Here we use metabolomic, genetic and structural approaches to show that Rv3722c is the primary aspartate aminotransferase of M. tuberculosis, and mediates an essential but underrecognized role in metabolism: nitrogen distribution. Rv3722c deficiency leads to virulence attenuation in macrophages and mice. Our results identify aspartate biosynthesis and nitrogen distribution as potential species-selective drug targets in M. tuberculosis.


Subject(s)
Aspartate Aminotransferases/metabolism , Aspartic Acid/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Nitrogen/metabolism , Animals , Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cells, Cultured , Female , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Protein Binding , Protein Conformation , Virulence/genetics
5.
Nature ; 571(7763): 72-78, 2019 07.
Article in English | MEDLINE | ID: mdl-31217586

ABSTRACT

New antibiotics are needed to combat rising levels of resistance, with new Mycobacterium tuberculosis (Mtb) drugs having the highest priority. However, conventional whole-cell and biochemical antibiotic screens have failed. Here we develop a strategy termed PROSPECT (primary screening of strains to prioritize expanded chemistry and targets), in which we screen compounds against pools of strains depleted of essential bacterial targets. We engineered strains that target 474 essential Mtb genes and screened pools of 100-150 strains against activity-enriched and unbiased compound libraries, probing more than 8.5 million chemical-genetic interactions. Primary screens identified over tenfold more hits than screening wild-type Mtb alone, with chemical-genetic interactions providing immediate, direct target insights. We identified over 40 compounds that target DNA gyrase, the cell wall, tryptophan, folate biosynthesis and RNA polymerase, as well as inhibitors that target EfpA. Chemical optimization yielded EfpA inhibitors with potent wild-type activity, thus demonstrating the ability of PROSPECT to yield inhibitors against targets that would have eluded conventional drug discovery.


Subject(s)
Antitubercular Agents/classification , Antitubercular Agents/isolation & purification , Drug Discovery/methods , Gene Deletion , Microbial Sensitivity Tests/methods , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Small Molecule Libraries/pharmacology , Antitubercular Agents/pharmacology , DNA Gyrase/metabolism , Drug Resistance, Microbial , Folic Acid/biosynthesis , Molecular Targeted Therapy , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Reproducibility of Results , Small Molecule Libraries/classification , Small Molecule Libraries/isolation & purification , Substrate Specificity , Topoisomerase II Inhibitors/isolation & purification , Topoisomerase II Inhibitors/pharmacology , Tryptophan/biosynthesis , Tuberculosis/drug therapy , Tuberculosis/microbiology
6.
mBio ; 8(6)2017 12 19.
Article in English | MEDLINE | ID: mdl-29259086

ABSTRACT

Tuberculosis is responsible for more deaths worldwide than any other infectious disease. For anyone looking to learn more about this persistent public health threat, this conversational "frequently asked questions" style review addresses a breadth of questions. It offers a brief, somewhat opinionated, review of what is and is not known, particularly in light of how findings in the lab do or do not help inform the understanding of human tuberculosis.


Subject(s)
Disease Management , Tuberculosis/diagnosis , Tuberculosis/drug therapy , Disease Transmission, Infectious/prevention & control , Humans , Tuberculosis/epidemiology , Tuberculosis/transmission
7.
mBio ; 5(1)2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24549842

ABSTRACT

UNLABELLED: Mycobacterium tuberculosis remains a major cause of death due to the lack of treatment accessibility, HIV coinfection, and drug resistance. Development of new drugs targeting previously unexplored pathways is essential to shorten treatment time and eliminate persistent M. tuberculosis. A promising biochemical pathway which may be targeted to kill both replicating and nonreplicating M. tuberculosis is the biosynthesis of NAD(H), an essential cofactor in multiple reactions crucial for respiration, redox balance, and biosynthesis of major building blocks. NaMN adenylyltransferase (NadD) and NAD synthetase (NadE), the key enzymes of NAD biosynthesis, were selected as promising candidate drug targets for M. tuberculosis. Here we report for the first time kinetic characterization of the recombinant purified NadD enzyme, setting the stage for its structural analysis and inhibitor development. A protein knockdown approach was applied to validate bothNadD and NadE as target enzymes. Induced degradation of either target enzyme showed a strong bactericidal effect which coincided with anticipated changes in relative levels of NaMN and NaAD intermediates (substrates of NadD and NadE, respectively) and ultimate depletion of the NAD(H) pool. A metabolic catastrophe predicted as a likely result of NAD(H) deprivation of cellular metabolism was confirmed by (13)C biosynthetic labeling followed by gas chromatography-mass spectrometry (GC-MS) analysis. A sharp suppression of metabolic flux was observed in multiple NAD(P)(H)-dependent pathways, including synthesis of many amino acids (serine, proline, aromatic amino acids) and fatty acids. Overall, these results provide strong validation of the essential NAD biosynthetic enzymes, NadD and NadE, as antimycobacterial drug targets. IMPORTANCE: To address the problems of M. tuberculosis drug resistance and persistence of tuberculosis, new classes of drug targets need to be explored. The biogenesis of NAD cofactors was selected for target validation because of their indispensable role in driving hundreds of biochemical transformations. We hypothesized that the disruption of NAD production in the cell via genetic suppression of the essential enzymes (NadD and NadE) involved in the last two steps of NAD biogenesis would lead to cell death, even under dormancy conditions. In this study, we confirmed the hypothesis using a protein knockdown approach in the model system of Mycobacterium smegmatis. We showed that induced proteolytic degradation of either target enzyme leads to depletion of the NAD cofactor pool, which suppresses metabolic flux through numerous NAD(P)-dependent pathways of central metabolism of carbon and energy production. Remarkably, bactericidal effect was observed even for nondividing bacteria cultivated under carbon starvation conditions.


Subject(s)
Amide Synthases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , NAD/biosynthesis , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Drug Discovery/methods , Gene Knockdown Techniques , Genes, Essential , Microbial Viability , NAD/antagonists & inhibitors
8.
PLoS One ; 8(9): e75245, 2013.
Article in English | MEDLINE | ID: mdl-24086479

ABSTRACT

Identification of new drug targets is vital for the advancement of drug discovery against Mycobacterium tuberculosis, especially given the increase of resistance worldwide to first- and second-line drugs. Because traditional target-based screening has largely proven unsuccessful for antibiotic discovery, we have developed a scalable platform for target identification in M. tuberculosis that is based on whole-cell screening, coupled with whole-genome sequencing of resistant mutants and recombineering to confirm. The method yields targets paired with whole-cell active compounds, which can serve as novel scaffolds for drug development, molecular tools for validation, and/or as ligands for co-crystallization. It may also reveal other information about mechanisms of action, such as activation or efflux. Using this method, we identified resistance-linked genes for eight compounds with anti-tubercular activity. Four of the genes have previously been shown to be essential: AspS, aspartyl-tRNA synthetase, Pks13, a polyketide synthase involved in mycolic acid biosynthesis, MmpL3, a membrane transporter, and EccB3, a component of the ESX-3 type VII secretion system. AspS and Pks13 represent novel targets in protein translation and cell-wall biosynthesis. Both MmpL3 and EccB3 are involved in membrane transport. Pks13, AspS, and EccB3 represent novel candidates not targeted by existing TB drugs, and the availability of whole-cell active inhibitors greatly increases their potential for drug discovery.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Drug Resistance, Bacterial/genetics , Mycobacterium tuberculosis/genetics , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Resistance, Bacterial/physiology , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/drug effects , Polyketide Synthases/chemistry , Polyketide Synthases/genetics , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL