Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(24): E4832-E4840, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28559332

ABSTRACT

Mycobacterium tuberculosis (Mtb) encounters stresses during the pathogenesis and treatment of tuberculosis (TB) that can suppress replication of the bacteria and render them phenotypically tolerant to most available drugs. Where studied, the majority of Mtb in the sputum of most untreated subjects with active TB have been found to be nonreplicating by the criterion that they do not grow as colony-forming units (cfus) when plated on agar. However, these cells are viable because they grow when diluted in liquid media. A method for generating such "differentially detectable" (DD) Mtb in vitro would aid studies of the biology and drug susceptibility of this population, but lack of independent confirmation of reported methods has contributed to skepticism about their existence. Here, we identified confounding artifacts that, when avoided, allowed development of a reliable method of producing cultures of ≥90% DD Mtb in starved cells. We then characterized several drugs according to whether they contribute to the generation of DD Mtb or kill them. Of the agents tested, rifamycins led to DD Mtb generation, an effect lacking in a rifampin-resistant strain with a mutation in rpoB, which encodes the canonical rifampin target, the ß subunit of RNA polymerase. In contrast, thioridazine did not generate DD Mtb from starved cells but killed those generated by rifampin.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Bacterial Proteins/antagonists & inhibitors , DNA-Directed RNA Polymerases/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Rifamycins/pharmacology , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial/genetics , Humans , Isoniazid/pharmacology , Mutation , Mycobacterium tuberculosis/genetics , Thioridazine/pharmacology , Tuberculosis/drug therapy , Tuberculosis/microbiology
2.
Proc Natl Acad Sci U S A ; 113(31): E4523-30, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27432954

ABSTRACT

The rising incidence of antimicrobial resistance (AMR) makes it imperative to understand the underlying mechanisms. Mycobacterium tuberculosis (Mtb) is the single leading cause of death from a bacterial pathogen and estimated to be the leading cause of death from AMR. A pyrido-benzimidazole, 14, was reported to have potent bactericidal activity against Mtb. Here, we isolated multiple Mtb clones resistant to 14. Each had mutations in the putative DNA-binding and dimerization domains of rv2887, a gene encoding a transcriptional repressor of the MarR family. The mutations in Rv2887 led to markedly increased expression of rv0560c. We characterized Rv0560c as an S-adenosyl-L-methionine-dependent methyltransferase that N-methylates 14, abolishing its mycobactericidal activity. An Mtb strain lacking rv0560c became resistant to 14 by mutating decaprenylphosphoryl-ß-d-ribose 2-oxidase (DprE1), an essential enzyme in arabinogalactan synthesis; 14 proved to be a nanomolar inhibitor of DprE1, and methylation of 14 by Rv0560c abrogated this activity. Thus, 14 joins a growing list of DprE1 inhibitors that are potently mycobactericidal. Bacterial methylation of an antibacterial agent, 14, catalyzed by Rv0560c of Mtb, is a previously unreported mechanism of AMR.


Subject(s)
Antitubercular Agents/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Gene Expression Regulation, Bacterial , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Models, Molecular , Molecular Structure , Mutation , Mycobacterium tuberculosis/genetics , Protein Domains , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , S-Adenosylmethionine/metabolism
3.
Environ Microbiol ; 19(1): 174-184, 2017 01.
Article in English | MEDLINE | ID: mdl-27696646

ABSTRACT

Dietary changes are known to affect gut community structure, but questions remain about the mechanisms by which diet induces shifts in microbiome membership. Here, we addressed these questions in the rumen microbiome ecosystem - a complex microbial community that resides in the upper digestive tract of ruminant animals and is responsible for the degradation of the ingested plant material. Our dietary intervention experiments revealed that diet affects the most abundant taxa within the microbiome and that a specific group of methanogenic archaea of the order Methanomicrobiales is highly sensitive to its changes. Using metabolomic analyses together with in vitro microbiology approaches and whole-genome sequencing of Methanomicrobium mobile, a key species within this group, we identified that redox potential changes with diet and is the main factor that causes these dietary induced alternations in this taxa's abundance. Our genomic analysis suggests that the redox potential effect stems from a reduced number of anti-reactive oxygen species proteins coded in this taxon's genome. Our study highlights redox potential as a pivotal factor that could serve as a sculpturing force of community assembly within anaerobic gut microbial communities.


Subject(s)
Archaea/isolation & purification , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Rumen/microbiology , Animal Feed/analysis , Animals , Archaea/classification , Archaea/genetics , Cattle , Diet , Oxidation-Reduction
4.
Antimicrob Agents Chemother ; 59(10): 6521-38, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26239979

ABSTRACT

The search for drugs that can kill replicating and nonreplicating Mycobacterium tuberculosis faces practical bottlenecks. Measurement of CFU and discrimination of bacteriostatic from bactericidal activity are costly in compounds, supplies, labor, and time. Testing compounds against M. tuberculosis under conditions that prevent the replication of M. tuberculosis often involves a second phase of the test in which conditions are altered to permit the replication of bacteria that survived the first phase. False-positive determinations of activity against nonreplicating M. tuberculosis may arise from carryover of compounds from the nonreplicating stage of the assay that act in the replicating stage. We mitigate these problems by carrying out a 96-well microplate liquid MIC assay and then transferring an aliquot of each well to a second set of plates in which each well contains agar supplemented with activated charcoal. After 7 to 10 days-about 2 weeks sooner than required to count CFU-fluorometry reveals whether M. tuberculosis bacilli in each well have replicated extensively enough to reduce a resazurin dye added for the final hour. This charcoal agar resazurin assay (CARA) distinguishes between bacterial biomasses in any two wells that differ by 2 to 3 log10 CFU. The CARA thus serves as a pretest and semiquantitative surrogate for longer, more laborious, and expensive CFU-based assays, helps distinguish bactericidal from bacteriostatic activity, and identifies compounds that are active under replicating conditions, nonreplicating conditions, or both. Results for 14 antimycobacterial compounds, including tuberculosis (TB) drugs, revealed that PA-824 (pretomanid) and TMC207 (bedaquiline) are largely bacteriostatic.


Subject(s)
Antitubercular Agents/pharmacology , Biological Assay , Colony Count, Microbial/methods , Mycobacterium tuberculosis/drug effects , Agar , Antitubercular Agents/classification , Charcoal/chemistry , Colony Count, Microbial/instrumentation , Coloring Agents/chemistry , Diarylquinolines/pharmacology , Fluorometry , Microbial Sensitivity Tests , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Nitroimidazoles/pharmacology , Oxazines/chemistry , Xanthenes/chemistry
5.
Proc Natl Acad Sci U S A ; 109(40): 16004-11, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-23012453

ABSTRACT

Existing drugs are slow to eradicate Mycobacterium tuberculosis (Mtb) in patients and have failed to control tuberculosis globally. One reason may be that host conditions impair Mtb's replication, reducing its sensitivity to most antiinfectives. We devised a high-throughput screen for compounds that kill Mtb when its replication has been halted by reactive nitrogen intermediates (RNIs), acid, hypoxia, and a fatty acid carbon source. At concentrations routinely achieved in human blood, oxyphenbutazone (OPB), an inexpensive anti-inflammatory drug, was selectively mycobactericidal to nonreplicating (NR) Mtb. Its cidal activity depended on mild acid and was augmented by RNIs and fatty acid. Acid and RNIs fostered OPB's 4-hydroxylation. The resultant 4-butyl-4-hydroxy-1-(4-hydroxyphenyl)-2-phenylpyrazolidine-3,5-dione (4-OH-OPB) killed both replicating and NR Mtb, including Mtb resistant to standard drugs. 4-OH-OPB depleted flavins and formed covalent adducts with N-acetyl-cysteine and mycothiol. 4-OH-OPB killed Mtb synergistically with oxidants and several antituberculosis drugs. Thus, conditions that block Mtb's replication modify OPB and enhance its cidal action. Modified OPB kills both replicating and NR Mtb and sensitizes both to host-derived and medicinal antimycobacterial agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Resistance, Microbial/drug effects , High-Throughput Screening Assays/methods , Mycobacterium tuberculosis/drug effects , Oxyphenbutazone/pharmacology , Animals , Chromatography, High Pressure Liquid , Drug Resistance, Microbial/physiology , Fatty Acids/metabolism , Female , Hydroxylation , Magnetic Resonance Spectroscopy , Mice , Microbial Sensitivity Tests , Mycobacterium tuberculosis/physiology , Oxyphenbutazone/metabolism , Oxyphenbutazone/pharmacokinetics , Reactive Nitrogen Species/metabolism
6.
Phys Rev Lett ; 113(3): 031301, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-25083631

ABSTRACT

The recent BICEP2 measurement of B-mode polarization in the cosmic microwave background (r = 0.2(-0.05)(+0.07)), a possible indication of primordial gravity waves, appears to be in tension with the upper limit from WMAP (r < 0.13 at 95% C.L.) and Planck (r < 0.11 at 95% C.L.). We carefully quantify the level of tension and show that it is very significant (around 0.1% unlikely) when the observed deficit of large-scale temperature power is taken into account. We show that measurements of TE and EE power spectra in the near future will discriminate between the hypotheses that this tension is either a statistical fluke or a sign of new physics. We also discuss extensions of the standard cosmological model that relieve the tension and some novel ways to constrain them.

7.
Sci Adv ; 10(11): eadj6406, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489355

ABSTRACT

There is a compelling need to find drugs active against Mycobacterium tuberculosis (Mtb). 4'-Phosphopantetheinyl transferase (PptT) is an essential enzyme in Mtb that has attracted interest as a potential drug target. We optimized a PptT assay, used it to screen 422,740 compounds, and identified raltitrexed, an antineoplastic antimetabolite, as the most potent PptT inhibitor yet reported. While trying unsuccessfully to improve raltitrexed's ability to kill Mtb and remove its ability to kill human cells, we learned three lessons that may help others developing antibiotics. First, binding of raltitrexed substantially changed the configuration of the PptT active site, complicating molecular modeling of analogs based on the unliganded crystal structure or the structure of cocrystals with inhibitors of another class. Second, minor changes in the raltitrexed molecule changed its target in Mtb from PptT to dihydrofolate reductase (DHFR). Third, the structure-activity relationship for over 800 raltitrexed analogs only became interpretable when we quantified and characterized the compounds' intrabacterial accumulation and transformation.


Subject(s)
Mycobacterium tuberculosis , Neoplasms , Quinazolines , Thiophenes , Transferases (Other Substituted Phosphate Groups) , Humans , Mycobacterium tuberculosis/metabolism , Thymidylate Synthase/metabolism , Bacterial Proteins/metabolism
8.
ACS Med Chem Lett ; 14(7): 970-976, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37465309

ABSTRACT

4'-Phosphopantetheinyl transferase (PptT) is an essential enzyme for Mycobacterium tuberculosis (Mtb) survival and virulence and therefore an attractive target for a tuberculosis therapeutic. In this work, two modeling-informed approaches toward the isosteric replacement of the amidinourea moiety present in the previously reported PptT inhibitor AU 8918 are reported. Although a designed 3,5-diamino imidazole unexpectedly adopted an undesired tautomeric form and was inactive, replacement of the amidinourea moiety afforded a series of active PptT inhibitors containing 2,6-diaminopyridine scaffolds.

9.
J Med Chem ; 65(3): 1996-2022, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35044775

ABSTRACT

A newly validated target for tuberculosis treatment is phosphopantetheinyl transferase, an essential enzyme that plays a critical role in the biosynthesis of cellular lipids and virulence factors in Mycobacterium tuberculosis. The structure-activity relationships of a recently disclosed inhibitor, amidinourea (AU) 8918 (1), were explored, focusing on the biochemical potency, determination of whole-cell on-target activity for active compounds, and profiling of selective active congeners. These studies show that the AU moiety in AU 8918 is largely optimized and that potency enhancements are obtained in analogues containing a para-substituted aromatic ring. Preliminary data reveal that while some analogues, including 1, have demonstrated cardiotoxicity (e.g., changes in cardiomyocyte beat rate, amplitude, and peak width) and inhibit Cav1.2 and Nav1.5 ion channels (although not hERG channels), inhibition of the ion channels is largely diminished for some of the para-substituted analogues, such as 5k (p-benzamide) and 5n (p-phenylsulfonamide).


Subject(s)
Bacterial Proteins/metabolism , Guanidine/analogs & derivatives , Mycobacterium tuberculosis/enzymology , Transferases (Other Substituted Phosphate Groups)/metabolism , Urea/analogs & derivatives , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Guanidine/chemistry , Guanidine/metabolism , Guanidine/pharmacology , Kinetics , Microbial Sensitivity Tests , Molecular Conformation , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Urea/chemistry , Urea/metabolism , Urea/pharmacology
10.
ACS Infect Dis ; 8(3): 557-573, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35192346

ABSTRACT

Rising antimicrobial resistance challenges our ability to combat bacterial infections. The problem is acute for tuberculosis (TB), the leading cause of death from infection before COVID-19. Here, we developed a framework for multiple pharmaceutical companies to share proprietary information and compounds with multiple laboratories in the academic and government sectors for a broad examination of the ability of ß-lactams to kill Mycobacterium tuberculosis (Mtb). In the TB Drug Accelerator (TBDA), a consortium organized by the Bill & Melinda Gates Foundation, individual pharmaceutical companies collaborate with academic screening laboratories. We developed a higher order consortium within the TBDA in which four pharmaceutical companies (GlaxoSmithKline, Sanofi, MSD, and Lilly) collectively collaborated with screeners at Weill Cornell Medicine, the Infectious Disease Research Institute (IDRI), and the National Institute of Allergy and Infectious Diseases (NIAID), pharmacologists at Rutgers University, and medicinal chemists at the University of North Carolina to screen ∼8900 ß-lactams, predominantly cephalosporins, and characterize active compounds. In a striking contrast to historical expectation, 18% of ß-lactams screened were active against Mtb, many without a ß-lactamase inhibitor. One potent cephaloporin was active in Mtb-infected mice. The steps outlined here can serve as a blueprint for multiparty, intra- and intersector collaboration in the development of anti-infective agents.


Subject(s)
COVID-19 , Mycobacterium tuberculosis , Animals , Drug Industry , Mice , SARS-CoV-2 , Universities , beta-Lactams/pharmacology
11.
J Pediatr Gastroenterol Nutr ; 52(5): 514-22, 2011 May.
Article in English | MEDLINE | ID: mdl-21464757

ABSTRACT

OBJECTIVES: The Pediatric Gastroesophageal Reflux Disease Symptom and Quality of Life Questionnaire (PGSQ) represents 2 related age-stratified tools developed to assess pediatric gastroesophageal reflux disease (GERD). These include the PGSQ-Cp (for children ages 2 to 8 years, parent/caregiver report) and the PGSQ-A (for adolescents ages 9-17 years). The objective of the present study was to develop and evaluate PGSQ measurement properties. MATERIALS AND METHODS: The PGSQ items were generated based on information from focus groups, expert clinician review, and cognitive debriefing interviews. The symptoms of pediatric GERD and the effect of these symptoms were addressed. The tools were evaluated in a 3-week psychometric evaluation with participants from 11 clinical sites in the United States. The study included other measures such as the Pediatric Quality of Life questionnaire (PedsQL) and clinician-rated GERD severity. After item reduction, internal consistency, reproducibility, construct validity, known-group validity, and responsiveness were assessed. RESULTS: The 231 participants included 75 parents of children ages 2 to 8 years and 75 children ages 9 to 17 years with GERD and 41 parents of children and 40 children ages 9 to 17 years without GERD. Exploratory factor analysis demonstrated 4 symptom subscales for the PGSQ-Cp and 3 symptom subscales for the PGSQ-A. Both had subscales for total impact and school impact. High to moderate internal consistency was observed, ranging from 0.76 to 0.96 for the PGSQ-Cp and from 0.67 to 0.94 for the PGSQ-A. The PGSQ significantly differentiated between patients with GERD and controls (P < 0.0001, PGSQ-Cp; P < 0.0022-0.0001, PGSQ-A) and demonstrated responsiveness. CONCLUSIONS: These results support the reliability, validity, and responsiveness of both versions of the PGSQ. The instruments should be useful for clinical studies.


Subject(s)
Activities of Daily Living , Gastroesophageal Reflux , Psychometrics/methods , Quality of Life , Surveys and Questionnaires , Adolescent , Adult , Child , Child, Preschool , Gastroesophageal Reflux/complications , Humans , Parents , Pediatrics/methods , Reproducibility of Results , Schools , Severity of Illness Index , Surveys and Questionnaires/standards , United States
12.
Methods Mol Biol ; 2314: 611-635, 2021.
Article in English | MEDLINE | ID: mdl-34235673

ABSTRACT

Models of nonreplication help us understand the biology of persistent Mycobacterium tuberculosis. High throughput screening (HTS) against nonreplicating M. tuberculosis may lead to identification of tool compounds that affect pathways on which bacterial survival depends in such states and to development of drugs that can overcome phenotypic resistance to conventional antimycobacterial agents, which are mostly active against replicating M. tuberculosis. We describe a multistress model of nonreplication that mimics some of the microenvironmental conditions that M. tuberculosis faces in the host as adapted for HTS. The model includes acidic pH, mild hypoxia, a flux of nitric oxide, and other reactive nitrogen intermediates arising from nitrite at low pH and low concentrations of a fatty acid (butyrate) as a carbon source.


Subject(s)
Antitubercular Agents/pharmacology , High-Throughput Screening Assays/methods , Hypoxia/physiopathology , Mycobacterium tuberculosis/growth & development , Nitric Oxide/metabolism , Stress, Physiological , Tuberculosis/drug therapy , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Tuberculosis/microbiology , Tuberculosis/pathology
13.
Sci Transl Med ; 13(621): eabg2612, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34818059

ABSTRACT

"Viable but nonculturable" states of bacteria pose challenges for environmental and clinical microbiology, but their biological mechanisms remain obscure. Mycobacterium tuberculosis (Mtb), the leading cause of death from infection until the coronavirus disease 2019 pandemic, affords a notable example of this phenotype. Mtb can enter into a "differentially detectable" (DD) state associated with phenotypic antimicrobial resistance. In this state, Mtb cells are viable but undetectable as colony-forming units. We found that Mtb cells enter the DD state when they undergo sublethal oxidative stress that damages their DNA, proteins, and lipids. In addition, their replication process is delayed, allowing time for repair. Mycobacterium bovis and its derivative, BCG, fail to enter the DD state under similar conditions. These findings have implications for tuberculosis latency, detection, relapse, treatment monitoring, and development of regimens that overcome phenotypic antimicrobial resistance.


Subject(s)
COVID-19 , Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/metabolism , Oxidative Stress , SARS-CoV-2 , Tuberculosis/metabolism
14.
Microbiol Spectr ; 9(2): e0092821, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34550010

ABSTRACT

Phosphopantetheinyl hydrolase, PptH (Rv2795c), is a recently discovered enzyme from Mycobacterium tuberculosis that removes 4'-phosphopantetheine (Ppt) from holo-carrier proteins (CPs) and thereby opposes the action of phosphopantetheinyl transferases (PPTases). PptH is the first structurally characterized enzyme of the phosphopantetheinyl hydrolase family. However, conditions for optimal activity of PptH have not been defined, and only one substrate has been identified. Here, we provide biochemical characterization of PptH and demonstrate that the enzyme hydrolyzes Ppt in vitro from more than one M. tuberculosis holo-CP as well as holo-CPs from other organisms. PptH provided the only detectable activity in mycobacterial lysates that dephosphopantetheinylated acyl carrier protein M (AcpM), suggesting that PptH is the main Ppt hydrolase in M. tuberculosis. We could not detect a role for PptH in coenzyme A (CoA) salvage, and PptH was not required for virulence of M. tuberculosis during infection of mice. It remains to be determined why mycobacteria conserve a broadly acting phosphohydrolase that removes the Ppt prosthetic group from essential CPs. We speculate that the enzyme is critical for aspects of the life cycle of M. tuberculosis that are not routinely modeled. IMPORTANCE Tuberculosis (TB), caused by Mycobacterium tuberculosis, was the leading cause of death from an infectious disease before COVID, yet the in vivo essentiality and function of many of the protein-encoding genes expressed by M. tuberculosis are not known. We biochemically characterize M. tuberculosis's phosphopantetheinyl hydrolase, PptH, a protein unique to mycobacteria that removes an essential posttranslational modification on proteins involved in synthesis of lipids important for the bacterium's cell wall and virulence. We demonstrate that the enzyme has broad substrate specificity, but it does not appear to have a role in coenzyme A (CoA) salvage or virulence in a mouse model of TB.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Pantetheine/analogs & derivatives , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Wall/metabolism , Female , Humans , Lipids/biosynthesis , Mice , Mice, Inbred C57BL , Pantetheine/metabolism , Protein Processing, Post-Translational , Tuberculosis/pathology , Virulence/physiology
15.
Nat Chem Biol ; 4(10): 609-16, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18724363

ABSTRACT

A screen of a genomic library from Mycobacterium tuberculosis (Mtb) identified a small, unannotated open reading frame (MT0196) that encodes a 4.9-kDa, cysteine-rich protein. Despite extensive nucleotide divergence, the amino acid sequence is highly conserved among mycobacteria that are pathogenic in vertebrate hosts. We synthesized the protein and found that it preferentially binds up to six Cu(I) ions in a solvent-shielded core. Copper, cadmium and compounds that generate nitric oxide or superoxide induced the gene's expression in Mtb up to 1,000-fold above normal expression. The native protein bound copper within Mtb and partially protected Mtb from copper toxicity. We propose that the product of the MT0196 gene be named mycobacterial metallothionein (MymT). To our knowledge, MymT is the first metallothionein of a Gram-positive bacterium with a demonstrated function.


Subject(s)
Copper/chemistry , Metallothionein/genetics , Mycobacterium tuberculosis/enzymology , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Gene Expression Regulation, Bacterial , Metallothionein/chemistry , Molecular Sequence Data , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Sequence Alignment
16.
Front Microbiol ; 11: 1248, 2020.
Article in English | MEDLINE | ID: mdl-32655524

ABSTRACT

As ß-lactams are reconsidered for the treatment of tuberculosis (TB), their targets are assumed to be peptidoglycan transpeptidases, as verified by adduct formation and kinetic inhibition of Mycobacterium tuberculosis (Mtb) transpeptidases by carbapenems active against replicating Mtb. Here, we investigated the targets of recently described cephalosporins that are selectively active against non-replicating (NR) Mtb. NR-active cephalosporins failed to inhibit recombinant Mtb transpeptidases. Accordingly, we used alkyne analogs of NR-active cephalosporins to pull down potential targets through unbiased activity-based protein profiling and identified over 30 protein binders. None was a transpeptidase. Several of the target candidates are plausibly related to Mtb's survival in an NR state. However, biochemical tests and studies of loss of function mutants did not identify a unique target that accounts for the bactericidal activity of these beta-lactams against NR Mtb. Instead, NR-active cephalosporins appear to kill Mtb by collective action on multiple targets. These results highlight the ability of these ß-lactams to target diverse classes of proteins.

17.
ACS Infect Dis ; 5(2): 272-280, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30501173

ABSTRACT

To find new inhibitors of Mycobacterium tuberculosis that have novel mechanisms of action, we miniaturized a high throughput screen to identify compounds that disrupt pH homeostasis. We adapted and validated a 384-well format assay to determine intrabacterial pH using a ratiometric green fluorescent protein. We screened 89000 small molecules under nonreplicating conditions and confirmed 556 hits that reduced intrabacterial pH (below pH 6.5). We selected five compounds that disrupt intrabacterial pH homeostasis and also showed some activity against nonreplicating bacteria in a 4-stress model, but with no (or greatly reduced) activity against replicating bacteria. The compounds selected were two benzamide sulfonamides, a benzothiadiazole, a bissulfone, and a thiadiazole, none of which are known antibacterial agents. All of these five compounds demonstrated bactericidal activity against nonreplicating bacteria in buffer. Four of the five compounds demonstrated increased activity under low pH conditions. None of the five compounds acted as ionophores or as general disrupters of membrane potential. These compounds are useful starting points for work to elucidate their mechanism of action and their utility for drug discovery.


Subject(s)
Antitubercular Agents/isolation & purification , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Drug Discovery , Green Fluorescent Proteins , High-Throughput Screening Assays , Homeostasis , Hydrogen-Ion Concentration , Microbial Sensitivity Tests
18.
J Antibiot (Tokyo) ; 72(6): 469-475, 2019 06.
Article in English | MEDLINE | ID: mdl-30903100

ABSTRACT

A systematic study of the stability of a set of cephalosporins in mouse plasma reveals that cephalosporins lacking an acidic moiety at C-2 may be vulnerable to ß-lactam cleavage in mouse plasma.


Subject(s)
Anti-Bacterial Agents/blood , Anti-Bacterial Agents/chemistry , Cephalosporins/blood , Cephalosporins/chemistry , Animals , Mice , Molecular Structure , Structure-Activity Relationship
19.
mBio ; 10(4)2019 07 09.
Article in English | MEDLINE | ID: mdl-31289182

ABSTRACT

A defining characteristic of treating tuberculosis is the need for prolonged administration of multiple drugs. This may be due in part to subpopulations of slowly replicating or nonreplicating Mycobacterium tuberculosis bacilli exhibiting phenotypic tolerance to most antibiotics in the standard treatment regimen. Confounding this problem is the increasing incidence of heritable multidrug-resistant M. tuberculosis A search for new antimycobacterial chemical scaffolds that can kill phenotypically drug-tolerant mycobacteria uncovered tricyclic 4-hydroxyquinolines and a barbituric acid derivative with mycobactericidal activity against both replicating and nonreplicating M. tuberculosis Both families of compounds depleted M. tuberculosis of intrabacterial magnesium. Complete or partial resistance to both chemotypes arose from mutations in the putative mycobacterial Mg2+/Co2+ ion channel, CorA. Excess extracellular Mg2+, but not other divalent cations, diminished the compounds' cidality against replicating M. tuberculosis These findings establish depletion of intrabacterial magnesium as an antimicrobial mechanism of action and show that M. tuberculosis magnesium homeostasis is vulnerable to disruption by structurally diverse, nonchelating, drug-like compounds.IMPORTANCE Antimycobacterial agents might shorten the course of treatment by reducing the number of phenotypically tolerant bacteria if they could kill M. tuberculosis in diverse metabolic states. Here we report two chemically disparate classes of agents that kill M. tuberculosis both when it is replicating and when it is not. Under replicating conditions, the tricyclic 4-hydroxyquinolines and a barbituric acid analogue deplete intrabacterial magnesium as a mechanism of action, and for both compounds, mutations in CorA, a putative Mg2+/Co2+ transporter, conferred resistance to the compounds when M. tuberculosis was under replicating conditions but not under nonreplicating conditions, illustrating that a given compound can kill M. tuberculosis in different metabolic states by disparate mechanisms. Targeting magnesium metallostasis represents a previously undescribed antimycobacterial mode of action that might cripple M. tuberculosis in a Mg2+-deficient intraphagosomal environment of macrophages.


Subject(s)
Antitubercular Agents/pharmacology , Cation Transport Proteins/genetics , Magnesium/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , DNA Replication , Homeostasis , Mutation
20.
Science ; 363(6426)2019 02 01.
Article in English | MEDLINE | ID: mdl-30705156

ABSTRACT

Mycobacterium tuberculosis (Mtb) is the leading infectious cause of death in humans. Synthesis of lipids critical for Mtb's cell wall and virulence depends on phosphopantetheinyl transferase (PptT), an enzyme that transfers 4'-phosphopantetheine (Ppt) from coenzyme A (CoA) to diverse acyl carrier proteins. We identified a compound that kills Mtb by binding and partially inhibiting PptT. Killing of Mtb by the compound is potentiated by another enzyme encoded in the same operon, Ppt hydrolase (PptH), that undoes the PptT reaction. Thus, loss-of-function mutants of PptH displayed antimicrobial resistance. Our PptT-inhibitor cocrystal structure may aid further development of antimycobacterial agents against this long-sought target. The opposing reactions of PptT and PptH uncover a regulatory pathway in CoA physiology.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Coenzyme A/metabolism , Guanidine/analogs & derivatives , Hydrolases/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Urea/analogs & derivatives , Acyl Carrier Protein/metabolism , Animals , Catalytic Domain , Drug Resistance, Bacterial/genetics , Female , Guanidine/pharmacology , Hydrolases/genetics , Lipid Metabolism , Loss of Function Mutation , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Operon , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL