Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Language
Publication year range
1.
J Enzyme Inhib Med Chem ; 36(1): 847-855, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33752554

ABSTRACT

The dihydroneopterin aldolase (DHNA, EC 4.1.2.25) activity of FolB protein is required for the conversion of 7,8-dihydroneopterin (DHNP) to 6-hydroxymethyl-7,8-dihydropterin (HP) and glycolaldehyde (GA) in the folate pathway. FolB protein from Mycobacterium tuberculosis (MtFolB) is essential for bacilli survival and represents an important molecular target for drug development. S8-functionalized 8-mercaptoguanine derivatives were synthesised and evaluated for inhibitory activity against MtFolB. The compounds showed IC50 values in the submicromolar range. The inhibition mode and inhibition constants were determined for compounds that exhibited the strongest inhibition. Additionally, molecular docking analyses were performed to suggest enzyme-inhibitor interactions and ligand conformations. To the best of our knowledge, this study describes the first class of MtFolB inhibitors.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Guanosine/analogs & derivatives , Molecular Docking Simulation , Mycobacterium tuberculosis/drug effects , Thionucleosides/pharmacology , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanosine/chemical synthesis , Guanosine/chemistry , Guanosine/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/enzymology , Thionucleosides/chemical synthesis , Thionucleosides/chemistry
2.
J Biol Inorg Chem ; 25(6): 887-901, 2020 09.
Article in English | MEDLINE | ID: mdl-32728907

ABSTRACT

A pharmacophore design approach, based on the coordination chemistry of an intimate molecular hybrid of active metabolites of pro-drugs, known to release active species upon enzymatic oxidative activation, is devised. This is exemplified by combining two anti-mycobacterial drugs: pyrazinamide (first line) and delamanid (third line) whose active metabolites are pyrazinoic acid (PyzCOOH) and likely nitroxyl (HNO (or NO.)), respectively. Aiming to generate those active species, a hybrid compound was envisaged by coordination of pyrazine-2-hydroxamic acid (PyzCONHOH) with a Na3[FeII(CN)5] moiety. The corresponding pentacyanoferrate(II) complex Na4[FeII(CN)5(PyzCONHO-)] was synthesized and characterized by several spectroscopic techniques, cyclic voltammetry, and DFT calculations. Chemical oxidation of this complex with H2O2 was shown to induce the release of the metabolite PyzCOOH, without the need of the Mycobacterium tuberculosis (Mtb) pyrazinamidase enzyme (PncA). Control experiments show that both H2O2- and N-coordinated pyrazine FeII species are required, ruling out a direct hydrolysis of the hydroxamic acid or an alternative oxidative route through chelation of a metal center by a hydroxamic group. The release of HNO was observed using EPR spectroscopy in the presence of a spin trapping agent. The devised iron metal complex of pyrazine-2-hydroxamic acid was found inactive against an actively growing/non-resistant Mtb strain; however, it showed a strong dose-dependent and reversible vasodilatory activity with mostly lesser toxic effects than the reference drug sodium nitroprussiate, unveiling thus a potential indication for acute or chronic cardiovascular pathology. This is a priori a further indirect evidence of HNO release from this metal complex, standing as a possible pharmacophore model for an alternative vasodilator drug.


Subject(s)
Antitubercular Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Ferrous Compounds/chemical synthesis , Hydroxamic Acids/chemistry , Iron/chemistry , Mycobacterium tuberculosis/drug effects , Nitrogen Oxides/chemistry , Amidohydrolases/metabolism , Antitubercular Agents/pharmacology , Coordination Complexes/pharmacology , Drug Discovery , Electron Spin Resonance Spectroscopy , Hydrogen Peroxide/chemistry , Ligands , Nitrogen Oxides/metabolism , Oxidation-Reduction , Pyrazinamide/analogs & derivatives , Pyrazinamide/chemistry , Vasodilation
3.
Eur J Med Chem ; 192: 112179, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32113048

ABSTRACT

Using a classical molecular simplification approach, a series of 36 quinolines were synthesized and evaluated as in vitro inhibitors of Mycobacterium tuberculosis (M. tuberculosis) growth. Structure-activity relationship (SAR) studies leaded to potent antitubercular agents, with minimum inhibitory concentration (MIC) values as low as 0.3 µM against M. tuberculosis H37Rv reference strain. Furthermore, the lead compounds were active against multidrug-resistant strains, without cross-resistance with some first- and second-line drugs. Testing the molecules against a spontaneous mutant strain containing a single mutation in the qcrB gene (T313A) indicated that the synthesized quinolines targeted the cytochrome bc1 complex. In addition, leading compounds were devoid of apparent toxicity to HepG2 and Vero cells and showed moderate elimination rates in human liver S9 fractions. Finally, the selected structures inhibited M. tuberculosis growth in a macrophage model of tuberculosis infection. Taken together, these data indicate that this class of compounds may furnish candidates for the future development of antituberculosis drugs.


Subject(s)
Antitubercular Agents/pharmacology , Drug Design , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Cell Survival/drug effects , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Macrophages/drug effects , Macrophages/microbiology , Mice , Microbial Sensitivity Tests , Molecular Structure , RAW 264.7 Cells , Structure-Activity Relationship , Vero Cells
4.
Eur J Med Chem ; 155: 153-164, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29885576

ABSTRACT

Using a classical hybridization approach, a series of 1H-benzo[d]imidazoles and 3,4-dihydroquinazolin-4-ones were synthesized (39 examples) and evaluated as inhibitors of Mycobacterium tuberculosis growth. Chemical modification studies yielded potent antitubercular agents with minimum inhibitory concentration (MIC) values as low as 0.24 µM against M. tuberculosis H37Rv strain. Further, the synthesized compounds were active against four drug-resistant strains containing different levels of resistance for the first line drugs. These molecules were devoid of apparent toxicity to HepG2, HaCat, and Vero cells with IC50s > 30 µM. Viability in mammalian cell cultures was evaluated using MTT and neutral red assays. In addition, some 3,4-dihydroquinazolin-4-ones showed low risk of cardiac toxicity, no signals of neurotoxicity or morphological alteration in zebrafish (Danio rerio) toxicity models. 3,4-Dihydroquinazolin-4-ones 9q and 9w were considered the lead compounds of these series of molecules with MIC values of 0.24 µM and 0.94 µM against M. tuberculosis H37Rv, respectively. Taken together, these data indicate that this class of compounds may furnish candidates for future development of novel anti-TB drugs.


Subject(s)
Antitubercular Agents/pharmacology , Benzimidazoles/pharmacology , Mycobacterium tuberculosis/drug effects , Quinazolinones/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Structure-Activity Relationship , Zebrafish
5.
J Inorg Biochem ; 179: 71-81, 2018 02.
Article in English | MEDLINE | ID: mdl-29175704

ABSTRACT

The emergence of multidrug-resistant strains of Mycobacterium tuberculosis (MTB) represents a major threat to global health. Isoniazid (INH) is a prodrug used in the first-line treatment of tuberculosis. It undergoes oxidation by a catalase-peroxidase KatG, leading to generation of an isonicotinoyl radical that reacts with NAD(H) forming the INH-NADH adduct as the active metabolite. A redox-mediated activation of isoniazid using an iron metal complex was previously proposed as a strategy to overcome isoniazid resistance due to KatG mutations. Here, we have prepared a series of iron metal complexes with isoniazid and analogues, containing alkyl substituents at the hydrazide moiety, and also with pyrazinamide derivatives. These complexes were activated by H2O2 and studied by ESR and LC-MS. For the first time, the formation of the oxidized INH-NAD adduct from the pentacyano(isoniazid)ferrate(II) complex was detected by LC-MS, supporting a redox-mediated activation, for which a mechanistic proposition is reported. ESR data showed all alkylated hydrazides, in contrast to non-substituted hydrazides, only generated alkyl-based radicals. The structural modifications did not improve minimal inhibitory concentration (MIC) against MTB in comparison to isoniazid iron complex, providing support to isonicotinoyl radical formation as a requirement for activity. Nonetheless, the pyrazinoic acid hydrazide iron complex showed redox-mediated activation using H2O2 with generation of a pyrazinoyl radical intermediate and production of pyrazinoic acid, which is in fact the active metabolite of pyrazinamide prodrug. Thereby, this strategy can also unveil new opportunities for activation of this type of drug.


Subject(s)
Antitubercular Agents/pharmacology , Coordination Complexes/pharmacology , Ferrous Compounds/pharmacology , Isoniazid/analogs & derivatives , Isoniazid/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Ferrous Compounds/chemical synthesis , Ferrous Compounds/chemistry , Isoniazid/chemical synthesis , Isoniazid/chemistry , Microbial Sensitivity Tests , Models, Chemical , Mycobacterium tuberculosis/drug effects , Oxidation-Reduction
6.
Braz. arch. biol. technol ; 59: e16150485, 2016. graf
Article in English | LILACS | ID: biblio-951405

ABSTRACT

The esophagus and mouth tumors are very frequent malignancies worldwide. Lipopolysaccharides (LPS) are capable of regulating gene expression of pro-inflammatory cytokines by binding to toll-like receptor 4 (TLR4). Recent studies show that LPS can increase the migration ability of human esophageal cancer cell line HKESC-2 by increasing its adhesion properties. However, the effect of LPS has not been tested on viability of human esophageal and oral cancer cells. This study aimed to determine the action of LPS on the cell proliferation and viability in OE19 (adenocarcinoma) and OE21 (squamous carcinoma) cell lines, representative of human esophageal cancer, and HN30 cell line, representative of human oral carcinoma. LPS was used as treatment to OE19 and OE21 cells, and PgLPS (Porphyromonasgingivalis lipopolysaccharide) to HN30 cells. Viability was assessed by MTT assay and proliferation by cell counting. TLR4 expression was evaluated by real-time PCR. LPS at higher concentrations decreased significantly cell viability in both cell lines, adenocarcinoma (OE19) and squamous esophageal carcinoma (OE21) at different times of treatment. In addition, both cell lines, OE19 and OE21, expressed TLR4 receptor. Taken together, our data demonstrated that LPS at high concentrations might contribute to tumor death, in agreement with previously data.

SELECTION OF CITATIONS
SEARCH DETAIL
...