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










Database
Language
Publication year range
1.
Bioorg Med Chem Lett ; 26(14): 3300-3306, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27261180

ABSTRACT

In this report, we describe the synthesis of 1-(Phthalazin-4-yl)-hydrazine using bronsted acidic ionic liquids and demonstrate their ability to inhibit asexual stage development of human malaria parasite, Plasmodium falciparum. Through computational studies, we short-listed chemical scaffolds with potential binding affinity to an essential parasite protein, dihydroorotate dehydrogenase (DHODH). Further, these compounds were synthesized in the lab and tested against P. falciparum. Several compounds from our library showed inhibitory activity at low micro-molar concentrations with minimal cytotoxic effects. These results indicate the potential of hydralazine derivatives as reference scaffolds to develop novel antimalarials.


Subject(s)
Antimalarials/pharmacology , Phthalazines/pharmacology , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Line , Dogs , Dose-Response Relationship, Drug , Molecular Structure , Parasitic Sensitivity Tests , Phthalazines/chemical synthesis , Phthalazines/chemistry , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 26(3): 1056-1060, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26725030

ABSTRACT

Bcl homologs prominently contribute to apoptotic resistance in cancer cells and serve as molecular targets in treatment of various cancers. Herein, we report the synthesis of biphenyl-adamantane derivatives by a ligand free palladium on carbon based Suzuki reaction using diisopropylamine as a base for the coupling of adamantane based aryl chloride with a variety of aryl boronic acids. Among the biphenyl derivatives synthesized, compound 3'-(adamantan-1-yl)-4'-methoxy-[1,1'-biphenyl]-3-ol (AMB) displayed cytotoxic activity against hepatocellular carcinoma cell lines without significantly affecting the normal cell lines. Further, AMB caused increased accumulation of the HCC cells in subG1 phase, decreased the expression of Bcl-2, Bcl-xL, cyclin D1, caspase-3, survivin and increased the cleavage of PARP in a time-dependent manner. In silico molecular interaction studies between Bcl homologs and AMB showed that the biphenyl scaffold is predicted to form π-π interactions with Phe-101 and Tyr-105 and the adamantyl fragment is predicted to occupy another hydrophobic region in the kink region of the binding groove. In summary, we report on the synthesis and biological characterization of adamantyl-tethered biphenylic compounds that induce apoptosis in tumor cells most likely by targeting Bcl homologs.


Subject(s)
Adamantane/chemistry , Apoptosis Regulatory Proteins/metabolism , Biphenyl Compounds/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins/chemistry , Binding Sites , Biphenyl Compounds/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Molecular Docking Simulation , Protein Structure, Tertiary
3.
PLoS One ; 10(10): e0139798, 2015.
Article in English | MEDLINE | ID: mdl-26470029

ABSTRACT

In this work, we describe the 'green' synthesis of novel 6-(adamantan-1-yl)-2-substituted-imidazo[2,1-b][1,3,4]thiadiazoles (AITs) by ring formation reactions using 1-(adamantan-1-yl)-2-bromoethanone and 5-alkyl/aryl-2-amino1,3,4-thiadiazoles on a nano material base in ionic liquid media. Given the established activity of imidazothiadiazoles against M. tuberculosis, we next examined the anti-TB activity of AITs against the H37Rv strain using Alamar blue assay. Among the tested compounds 6-(adamantan-1-yl)-2-(4-methoxyphenyl)imidazo[2,1-b][1,3,4]thiadiazole (3f) showed potent inhibitory activity towards M. tuberculosis with an MIC value of 8.5 µM. The inhibitory effect of this molecule against M. tuberculosis was comparable to the standard drugs such as Pyrazinamide, Streptomycin, and Ciprofloxacin drugs. Mechanistically, an in silico analysis predicted sterol 14α-demethylase (CYP51) as the likely target and experimental activity of 3f in this system corroborated the in silico target prediction. In summary, we herein report the synthesis and biological evaluation of novel AITs against M. tuberculosis that likely target CYP51 to induce their antimycobacterial activity.


Subject(s)
Adamantane/chemistry , Ionic Liquids/chemistry , Magnesium Oxide/chemistry , Mycobacterium tuberculosis/drug effects , Sterol 14-Demethylase/metabolism , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Aspergillus fumigatus/drug effects , Catalysis , Chemistry Techniques, Synthetic , Drug Discovery , Green Chemistry Technology , Models, Molecular , Mycobacterium tuberculosis/enzymology , Nanostructures/chemistry , Protein Conformation , Sterol 14-Demethylase/chemistry , Thiadiazoles/chemical synthesis
4.
Org Biomol Chem ; 13(43): 10681-90, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26347024

ABSTRACT

Malaria parasites are currently gaining drug-resistance rapidly, across countries and continents. Hence, the discovery and development of novel chemical scaffolds, with superior antimalarial activity remain an important priority, for the developing world. Our report describes the development, characterization and evaluation of novel bepotastine-based sulphonamide antimalarials inhibiting asexual stage development of Plasmodium falciparum parasites in vitro. The screening results showed potent inhibitory activity of a number of novel sulphonamides against P. falciparum at low micromolar concentrations, in particular in late-stage parasite development. Based on computational studies we hypothesize N-myristoyltransferase as the target of the compounds developed here. Our results demonstrate the value of novel bepotastine-based sulphonamide compounds for targeting the asexual developmental stages of P. falciparum.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Antimalarials/chemical synthesis , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/microbiology , Models, Molecular , Piperidines/chemical synthesis , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis
5.
Bioorg Med Chem ; 23(18): 6157-65, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26299825

ABSTRACT

1,2,3-Triazole-based heterocycles have previously been shown to possess significant anticancer activity in various tumor models. In the present study, we attached a 1,2,3-triazole moiety to the third position of a 1,2-benzisoxazole heterocycle via copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) with various alkynes and established for the title compounds significant antiproliferative effect against human acute myeloid leukemia (AML) cells. Among the tested compounds, 3-(4-(4-phenoxyphenyl)-1H-1,2,3-triazol-1-yl)benzo[d]isoxazole (PTB) was found to be the most potent antiproliferative agent with an IC50 of 2 µM against MV4-11 cells using MTT assay. Notably, PTB induced cytotoxicity in MOLM13, MOLM14 and MV4-11 cells with selectivity over normal bone marrow cells (C57BL/6). Furthermore, PTB was found to induce cytotoxicity by increasing apoptosis of AML cells (MOLM13, MOLM14 and MV4-11) as well as sub-G1 cell population and apoptotic cells at submicromolar concentrations, as shown by flow cytometry and Annexin-V staining, respectively. On the protein level we suggested histone deacetylases (HDACs) as the potential protein target of those compounds in silico, and the predicted target was next experimentally validated by measuring the variations in the levels of p21, cyclin D and acetylation of histone H3 and tubulin. Molecular docking analysis of the title compounds with the second deacetylase domain of HDAC6 displayed high degree of shape complementarity to the binding site of the enzyme, forming multiple molecular interactions in the hydrophobic region as well as a hydrogen bond to the phenol side-chain of Tyr-782. Thus, 1,2,3-triazole derivatives appear to represent a class of novel, biologically active ligands against histone deacetylases which deserve to be further evaluated in their applications in the cancer field.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylases/chemistry , Triazoles/chemistry , Tubulin/metabolism , Acetylation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/toxicity , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Isoxazoles/chemistry , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Triazoles/chemical synthesis , Triazoles/toxicity
6.
PLoS One ; 9(9): e106364, 2014.
Article in English | MEDLINE | ID: mdl-25184206

ABSTRACT

The classical antivenom therapy has appreciably reduced snakebite mortality rate and thus is the only savior drug available. Unfortunately, it considerably fails to shield the viper bite complications like hemorrhage, local tissue degradation and necrosis responsible for severe morbidity. Moreover, the therapy is also tagged with limitations including anaphylaxis, serum sickness and poor availability. Over the last decade, snake venom metalloproteases (SVMPs) are reported to be the primary component responsible for hemorrhage and tissue degradation at bitten site. Thus, antivenom inability to offset viper venom-induced local toxicity has been a basis for an insistent search for SVMP inhibitors. Here we report the inhibitory effect of compound 5d, an apigenin based molecule against SVMPs both in silico and in vivo. Several apigenin analogues are synthesized using multicomponent Ugi reactions. Among them, compound 5d effectively abrogated Echis carinatus (EC) venom-induced local hemorrhage, tissue necrosis and myotoxicity in a dose dependant fashion. The histopathological study further conferred effective inhibition of basement membrane degradation, and accumulation of inflammatory leucocytes at the site of EC venom inoculation. The compound also protected EC venom-induced fibrin and fibrinogen degradation. The molecular docking of compound 5d and bothropasin demonstrated the direct interaction of hydroxyl group of compound with Glu146 present in hydrophobic pocket of active site and does not chelate Zn2+. Hence, it is concluded that compound 5d could be a potent agent in viper bite management.


Subject(s)
Apigenin/administration & dosage , Hemorrhage/drug therapy , Metalloproteases/antagonists & inhibitors , Snake Bites/drug therapy , Animals , Apigenin/chemical synthesis , Apigenin/chemistry , Crotalid Venoms/chemistry , Fibrin/chemistry , Fibrin/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Hemorrhage/pathology , Metalloendopeptidases/chemistry , Metalloproteases/metabolism , Mice , Molecular Docking Simulation , Snake Bites/pathology , Snake Venoms/antagonists & inhibitors , Snake Venoms/chemistry , Snake Venoms/enzymology , Viperidae
SELECTION OF CITATIONS
SEARCH DETAIL
...