Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 186(5): 1013-1025.e24, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36827973

ABSTRACT

The emergence of drug-resistant tuberculosis has created an urgent need for new anti-tubercular agents. Here, we report the discovery of a series of macrolides called sequanamycins with outstanding in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb). Sequanamycins are bacterial ribosome inhibitors that interact with the ribosome in a similar manner to classic macrolides like erythromycin and clarithromycin, but with binding characteristics that allow them to overcome the inherent macrolide resistance of Mtb. Structures of the ribosome with bound inhibitors were used to optimize sequanamycin to produce the advanced lead compound SEQ-9. SEQ-9 was efficacious in mouse models of acute and chronic TB as a single agent, and it demonstrated bactericidal activity in a murine TB infection model in combination with other TB drugs. These results support further investigation of this series as TB clinical candidates, with the potential for use in new regimens against drug-susceptible and drug-resistant TB.


Subject(s)
Antitubercular Agents , Mycobacterium tuberculosis , Animals , Mice , Antitubercular Agents/pharmacology , Macrolides , Drug Resistance, Bacterial , Clarithromycin
2.
Antimicrob Agents Chemother ; 66(1): e0140021, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34723632

ABSTRACT

Cyclohexyl-griselimycin is a preclinical candidate for use against tuberculosis (TB). Here, we show that this oral cyclodepsipeptide is also active against the intrinsically drug-resistant nontuberculous mycobacterium Mycobacterium abscessus in vitro and in a mouse model of infection. This adds a novel advanced lead compound to the M. abscessus drug pipeline and supports a strategy of screening chemical matter generated in TB drug discovery efforts to fast-track the discovery of novel antibiotics against M. abscessus.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Mice , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria , Peptides, Cyclic
3.
Article in English | MEDLINE | ID: mdl-28874370

ABSTRACT

The mycobacterial phosphoglycosyltransferase WecA, which initiates arabinogalactan biosynthesis in Mycobacterium tuberculosis, has been proposed as a target of the caprazamycin derivative CPZEN-45, a preclinical drug candidate for the treatment of tuberculosis. In this report, we describe the functional characterization of mycobacterial WecA and confirm the essentiality of its encoding gene in M. tuberculosis by demonstrating that the transcriptional silencing of wecA is bactericidal in vitro and in macrophages. Silencing wecA also conferred hypersensitivity of M. tuberculosis to the drug tunicamycin, confirming its target selectivity for WecA in whole cells. Simple radiometric assays performed with mycobacterial membranes and commercially available substrates allowed chemical validation of other putative WecA inhibitors and resolved their selectivity toward WecA versus another attractive cell wall target, translocase I, which catalyzes the first membrane step in the biosynthesis of peptidoglycan. These assays and the mutant strain described herein will be useful for identifying potential antitubercular leads by screening chemical libraries for novel WecA inhibitors.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Bacterial Proteins/analysis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Evaluation, Preclinical/methods , Gene Expression Regulation, Bacterial/drug effects , Gene Silencing , Macrophages/microbiology , Microbial Sensitivity Tests , Molecular Targeted Therapy/methods , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Radiometry/methods , Transferases/analysis , Transferases/metabolism , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/genetics , Tuberculosis/microbiology , Tunicamycin/pharmacology , Uridine/analogs & derivatives , Uridine/pharmacology
4.
Bioorg Med Chem Lett ; 26(21): 5290-5299, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671498

ABSTRACT

A high throughput phenotypic screening against Mycobacterium smegmatis led us to the discovery of a new class of bacteriostatic, highly hydrophobic antitubercular quinazolinones that potently inhibited the in vitro growth of either extracellular or intramacrophagic M. tuberculosis (Mtb), via modulation of an unidentified but yet novel target. Optimization of the initial hit compound culminated in the identification of potent but poorly soluble Mtb growth inhibitors, three of which were progressed to in vivo efficacy studies. Despite nanomolar in vitro potency and attractive PK properties, none of these compounds was convincingly potent in our in vivo mouse tuberculosis models. This lack of efficacy may be linked to the poor drug-likeness of the test molecules and/or to the properties of the target.


Subject(s)
Antitubercular Agents/pharmacology , Quinazolinones/chemistry , Quinazolinones/pharmacology , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Cell Line , High-Throughput Screening Assays , Humans , Mice , Microbial Sensitivity Tests , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/drug effects , Quinazolinones/pharmacokinetics , Structure-Activity Relationship
5.
J Antimicrob Chemother ; 70(11): 3070-3, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26245639

ABSTRACT

OBJECTIVES: The increasing number of clinical strains resistant to one or more of the front-line TB drugs complicates the management of this disease. To develop next-generation benzimidazole-based FtsZ inhibitors with improved efficacy, we employed iterative optimization strategies based on whole bacteria potency, bactericidal activity, plasma and metabolic stability and in vivo efficacy studies. METHODS: Candidate benzimidazoles were evaluated for potency against Mycobacterium tuberculosis H37Rv and select clinical strains, toxicity against Vero cells and compound stability in plasma and liver microsomes. The efficacy of lead compounds was assessed in the acute murine M. tuberculosis infection model via intraperitoneal and oral routes. RESULTS: MICs of SB-P17G-A33, SB-P17G-A38 and SB-P17G-A42 for M. tuberculosis H37Rv and select clinical strains were 0.18-0.39 mg/L. SB-P17G-A38 and SB-P17G-A42 delivered at 50 mg/kg twice daily intraperitoneally or orally demonstrated efficacy in reducing the bacterial load by 5.7-6.3 log10 cfu in the lungs and 3.9-5.0 log10 cfu in the spleen. SB-P17G-A33 delivered at 50 mg/kg twice daily intraperitoneally or orally also reduced the bacterial load by 1.7-2.1 log10 cfu in the lungs and 2.5-3.4 log10 cfu in the spleen. CONCLUSIONS: Next-generation benzimidazoles with excellent potency and efficacy against M. tuberculosis have been developed. This is the first report on benzimidazole-based FtsZ inhibitors showing an equivalent level of efficacy to isoniazid in an acute murine M. tuberculosis infection model.


Subject(s)
Antitubercular Agents/administration & dosage , Benzimidazoles/administration & dosage , Isoniazid/administration & dosage , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Administration, Oral , Animals , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Benzimidazoles/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Cytoskeletal Proteins/antagonists & inhibitors , Disease Models, Animal , Drug Stability , Inactivation, Metabolic , Injections, Intraperitoneal , Isoniazid/pharmacology , Mice , Microbial Sensitivity Tests , Treatment Outcome , Vero Cells
6.
Bioorg Med Chem Lett ; 25(7): 1455-9, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25770781

ABSTRACT

A series of imidazo[1,2-a]indeno[1,2-e]pyrazin-4-ones that potently inhibit M. tuberculosis glutamine synthetase (GlnA1) has been identified by high throughput screening. Exploration of this series was performed owing to a short chemistry program. Despite possibly nanomolar inhibitions, none of these compounds was active on whole cell Mtb, suggesting that GlnA1 may not be a suitable target to find new anti-tubercular drugs.


Subject(s)
Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glutamate-Ammonia Ligase/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Imidazoles/pharmacology , Mycobacterium tuberculosis/drug effects , Pyrazines/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glutamate-Ammonia Ligase/metabolism , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , High-Throughput Screening Assays , Imidazoles/chemical synthesis , Imidazoles/chemistry , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/enzymology , Pyrazines/chemical synthesis , Pyrazines/chemistry
7.
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
8.
Front Immunol ; 12: 668060, 2021.
Article in English | MEDLINE | ID: mdl-34276658

ABSTRACT

Mycobacterium tuberculosis (Mtb), the etiological agent of tuberculosis, kills 1.5 to 1.7 million people every year. Macrophages are Mtb's main host cells and their inflammatory response is an essential component of the host defense against Mtb. However, Mtb is able to circumvent the macrophages' defenses by triggering an inappropriate inflammatory response. The ability of Mtb to hinder phagolysosome maturation and acidification, and to escape the phagosome into the cytosol, is closely linked to its virulence. The modulation of the host inflammatory response relies on Mtb virulence factors, but remains poorly studied. Understanding macrophage interactions with Mtb is crucial to develop strategies to control tuberculosis. The present study aims to determine the inflammatory response transcriptome and miRNome of human macrophages infected with the virulent H37Rv Mtb strain, to identify macrophage genetic networks specifically modulated by Mtb virulence. Using human macrophages infected with two different live strains of mycobacteria (live or heat-inactivated Mtb H37Rv and M. marinum), we quantified and analyzed 184 inflammatory mRNAs and 765 micro(mi)RNAs. Transcripts and miRNAs differently modulated by H37Rv in comparison with the two other conditions were analyzed using in silico approaches. We identified 30 host inflammatory response genes and 37 miRNAs specific for H37Rv virulence, and highlight evidence suggesting that Mtb intracellular-linked virulence depends on the inhibition of IL-1ß-dependent pro-inflammatory response, the repression of apoptosis and the delay of the recruitment and activation of adaptive immune cells. Our findings provide new potential targets for the development of macrophage-based therapeutic strategies against TB.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Lung/microbiology , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Adaptive Immunity , Apoptosis , Cytokines/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Lung/immunology , Lung/metabolism , Macrophages/immunology , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/immunology , Signal Transduction , THP-1 Cells , Transcriptome , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/metabolism , Virulence
9.
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
10.
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
11.
ACS Infect Dis ; 5(8): 1433-1445, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31184461

ABSTRACT

The historical view of ß-lactams as ineffective antimycobacterials has given way to growing interest in the activity of this class against Mycobacterium tuberculosis (Mtb) in the presence of a ß-lactamase inhibitor. However, most antimycobacterial ß-lactams kill Mtb only or best when the bacilli are replicating. Here, a screen of 1904 ß-lactams led to the identification of cephalosporins substituted with a pyrithione moiety at C3' that are active against Mtb under both replicating and nonreplicating conditions, neither activity requiring a ß-lactamase inhibitor. Studies showed that activity against nonreplicating Mtb required the in situ release of the pyrithione, independent of the known class A ß-lactamase, BlaC. In contrast, replicating Mtb could be killed both by released pyrithione and by the parent ß-lactam. Thus, the antimycobacterial activity of pyrithione-containing cephalosporins arises from two mechanisms that kill mycobacteria in different metabolic states.


Subject(s)
Antitubercular Agents/pharmacology , Cephalosporins/pharmacology , DNA Replication , Mycobacterium tuberculosis/drug effects , Pyridines/pharmacology , Thiones/pharmacology , Administration, Oral , Animals , Antitubercular Agents/administration & dosage , Callithrix , Cephalosporins/administration & dosage , Drug Discovery , Female , Hep G2 Cells , High-Throughput Screening Assays , Humans , Mice , Mycobacterium tuberculosis/physiology , Pyridines/administration & dosage , Thiones/administration & dosage
13.
ACS Infect Dis ; 3(1): 5-17, 2017 01 13.
Article in English | MEDLINE | ID: mdl-27726334

ABSTRACT

VCC234718, a molecule with growth inhibitory activity against Mycobacterium tuberculosis (Mtb), was identified by phenotypic screening of a 15344-compound library. Sequencing of a VCC234718-resistant mutant identified a Y487C substitution in the inosine monophosphate dehydrogenase, GuaB2, which was subsequently validated to be the primary molecular target of VCC234718 in Mtb. VCC234718 inhibits Mtb GuaB2 with a Ki of 100 nM and is uncompetitive with respect to IMP and NAD+. This compound binds at the NAD+ site, after IMP has bound, and makes direct interactions with IMP; therefore, the inhibitor is by definition uncompetitive. VCC234718 forms strong pi interactions with the Y487 residue side chain from the adjacent protomer in the tetramer, explaining the resistance-conferring mutation. In addition to sensitizing Mtb to VCC234718, depletion of GuaB2 was bactericidal in Mtb in vitro and in macrophages. When supplied at a high concentration (≥125 µM), guanine alleviated the toxicity of VCC234718 treatment or GuaB2 depletion via purine salvage. However, transcriptional silencing of guaB2 prevented Mtb from establishing an infection in mice, confirming that Mtb has limited access to guanine in this animal model. Together, these data provide compelling validation of GuaB2 as a new tuberculosis drug target.


Subject(s)
Antitubercular Agents/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Mycobacterium/drug effects , Sulfones/pharmacology , Tuberculosis/drug therapy , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Discovery , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Genome, Bacterial , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mutation , Tuberculosis/microbiology
14.
Tuberculosis (Edinb) ; 100: 95-101, 2016 09.
Article in English | MEDLINE | ID: mdl-27553416

ABSTRACT

The lack of proper treatment for serious infectious diseases due to the emergence of multidrug resistance reinforces the need for the discovery of novel antibiotics. This is particularly true for tuberculosis (TB) for which 3.7% of new cases and 20% of previously treated cases are estimated to be caused by multi-drug resistant strains. In addition, in the case of TB, which claimed 1.5 million lives in 2014, the treatment of the least complicated, drug sensitive cases is lengthy and disagreeable. Therefore, new drugs with novel targets are urgently needed to control resistant Mycobacterium tuberculosis strains. In this manuscript we report the characterization of the thiopeptide micrococcin P1 as an anti-tubercular agent. Our biochemical experiments show that this antibiotic inhibits the elongation step of protein synthesis in mycobacteria. We have further identified micrococcin resistant mutations in the ribosomal protein L11 (RplK); the mutations were located in the proline loop at the N-terminus. Reintroduction of the mutations into a clean genetic background, confirmed that they conferred resistance, while introduction of the wild type RplK allele into resistant strains re-established sensitivity. We also identified a mutation in the 23S rRNA gene. These data, in good agreement with previous structural studies suggest that also in M. tuberculosis micrococcin P1 functions by binding to the cleft between the 23S rRNA and the L11 protein loop, thus interfering with the binding of elongation factors Tu and G (EF-Tu and EF-G) and inhibiting protein translocation.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Bacteriocins/pharmacology , Mycobacterium tuberculosis/drug effects , Peptides/pharmacology , Animals , Antibiotics, Antitubercular/administration & dosage , Bacterial Proteins/biosynthesis , Bacteriocins/administration & dosage , Cells, Cultured , Colony Count, Microbial , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical/methods , Drug Resistance, Bacterial/genetics , Humans , Macrophages/microbiology , Microbial Sensitivity Tests/methods , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/isolation & purification , Peptide Chain Elongation, Translational/drug effects , Peptides/administration & dosage , Ribosomal Proteins/genetics
15.
Science ; 348(6239): 1106-12, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26045430

ABSTRACT

The discovery of Streptomyces-produced streptomycin founded the age of tuberculosis therapy. Despite the subsequent development of a curative regimen for this disease, tuberculosis remains a worldwide problem, and the emergence of multidrug-resistant Mycobacterium tuberculosis has prioritized the need for new drugs. Here we show that new optimized derivatives from Streptomyces-derived griselimycin are highly active against M. tuberculosis, both in vitro and in vivo, by inhibiting the DNA polymerase sliding clamp DnaN. We discovered that resistance to griselimycins, occurring at very low frequency, is associated with amplification of a chromosomal segment containing dnaN, as well as the ori site. Our results demonstrate that griselimycins have high translational potential for tuberculosis treatment, validate DnaN as an antimicrobial target, and capture the process of antibiotic pressure-induced gene amplification.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Mycobacterium tuberculosis/drug effects , Peptides, Cyclic/pharmacology , Tuberculosis, Multidrug-Resistant/drug therapy , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Cell Line, Tumor , Crystallography, X-Ray , DNA-Directed DNA Polymerase , Disease Models, Animal , Drug Design , Humans , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Protein Structure, Secondary , Streptomyces/chemistry , Streptomyces/drug effects , Streptomyces/metabolism , Tuberculosis, Multidrug-Resistant/microbiology
16.
PLoS One ; 9(4): e93953, 2014.
Article in English | MEDLINE | ID: mdl-24736743

ABSTRACT

Trisubstituted benzimidazoles have demonstrated potency against Gram-positive and Gram-negative bacterial pathogens. Previously, a library of novel trisubstituted benzimidazoles was constructed for high throughput screening, and compounds were identified that exhibited potency against M. tuberculosis H37Rv and clinical isolates, and were not toxic to Vero cells. A new series of 2-cyclohexyl-5-acylamino-6-N, N-dimethylaminobenzimidazoles derivatives has been developed based on SAR studies. Screening identified compounds with potency against M. tuberculosis. A lead compound from this series, SB-P17G-A20, was discovered to have an MIC of 0.16 µg/mL and demonstrated efficacy in the TB murine acute model of infection based on the reduction of bacterial load in the lungs and spleen by 1.73 ± 0.24 Log10 CFU and 2.68 ± Log10 CFU, respectively, when delivered at 50 mg/kg by intraperitoneal injection (IP) twice daily (bid). The activity of SB-P17G-A20 was determined to be concentration dependent and to have excellent stability in mouse and human plasma, and liver microsomes. Together, these studies demonstrate that SB-P17G-A20 has potency against M. tuberculosis clinical strains with varying susceptibility and efficacy in animal models of infection, and that trisubstituted benzimidazoles continue to be a platform for the development of novel inhibitors with efficacy.


Subject(s)
Antitubercular Agents/pharmacology , Benzimidazoles/pharmacology , Cell Division/drug effects , Mycobacterium tuberculosis/drug effects , Animals , Antitubercular Agents/administration & dosage , Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzimidazoles/administration & dosage , Benzimidazoles/chemistry , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Stability , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Protein Multimerization/drug effects , Rifamycins/administration & dosage , Rifamycins/pharmacology , Treatment Outcome , Tuberculosis/drug therapy , Tuberculosis/microbiology
18.
J Biol Chem ; 277(25): 22386-94, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-11950831

ABSTRACT

Substance P (SP) interacts with the neurokinin-1 (NK-1) G-protein-coupled receptor, which has been cloned in several species. In the present study, the domains of the NK-1 receptor involved in the binding of SP and SP-(7-11) C-terminal fragment have been analyzed using two peptide analogs containing the photoreactive amino acid para-benzoylphenylalanine ((p-Bz)Phe) in position 8 of their sequence. This study was carried out with [BAPA-Lys(6),(p-Bz)Phe(8),Pro(9),Met(O(2))(11)]SP-(7-11) and [BAPA(0),(p-Bz)Phe(8)]SP on both rat and human NK-1 receptors expressed in CHO cells. Combined trypsin and endo-GluC enzymatic complete digestions and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis led to the identification of the same domain of covalent interaction, (173)TMPSR(177), for the two photoactivatable peptides. Further digestion of this fragment with carboxypeptidase Y led to the identification of (173)TMP(175) in the second extracellular loop (E2) of the NK-1 receptor as the site of covalent attachment. Models of the conformation of this E2 loop in the human NK-1 receptor were generated using two different strategies, one based on homology with bovine rhodopsin and the other based on the solution conformation preferences of a synthetic peptide corresponding to the E2 loop.


Subject(s)
Phenylalanine/analogs & derivatives , Receptors, Neurokinin-1/chemistry , Substance P/chemistry , Amino Acid Sequence , Animals , CHO Cells , Carboxypeptidases/chemistry , Carboxypeptidases/metabolism , Cathepsin A , Cattle , Cell Membrane/metabolism , Cricetinae , Humans , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Peptide Biosynthesis , Peptides/chemistry , Phenylalanine/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Rats , Receptors, Neurokinin-1/metabolism , Rhodopsin/chemistry , Salicylates , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptavidin/pharmacology , Substance P/metabolism , Trypsin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL