Cargando…

Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase

L-Aspartate α-decarboxylase (ADC) belongs to a class of pyruvoyl dependent enzymes and catalyzes the conversion of aspartate to β-alanine in the pantothenate pathway, which is critical for the growth of several micro-organisms, including Mycobacterium tuberculosis (Mtb). Its presence only in micro-o...

Descripción completa

Detalles Bibliográficos
Autores principales: Sharma, Reetu, Kothapalli, Roopa, Van Dongen, Antonius M. J., Swaminathan, Kunchithapadam
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3314653/
https://www.ncbi.nlm.nih.gov/pubmed/22470451
http://dx.doi.org/10.1371/journal.pone.0033521
_version_ 1782228121753223168
author Sharma, Reetu
Kothapalli, Roopa
Van Dongen, Antonius M. J.
Swaminathan, Kunchithapadam
author_facet Sharma, Reetu
Kothapalli, Roopa
Van Dongen, Antonius M. J.
Swaminathan, Kunchithapadam
author_sort Sharma, Reetu
collection PubMed
description L-Aspartate α-decarboxylase (ADC) belongs to a class of pyruvoyl dependent enzymes and catalyzes the conversion of aspartate to β-alanine in the pantothenate pathway, which is critical for the growth of several micro-organisms, including Mycobacterium tuberculosis (Mtb). Its presence only in micro-organisms, fungi and plants and its absence in animals, particularly human, make it a promising drug target. We have followed a chemoinformatics-based approach to identify potential drug-like inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase (MtbADC). The structure-based high throughput virtual screening (HTVS) mode of the Glide program was used to screen 333,761 molecules of the Maybridge, National Cancer Institute (NCI) and Food and Drug Administration (FDA) approved drugs databases. Ligands were rejected if they cross-reacted with S-adenosylmethionine (SAM) decarboxylase, a human pyruvoyl dependent enzyme. The lead molecules were further analyzed for physicochemical and pharmacokinetic parameters, based on Lipinski's rule of five, and ADMET (absorption, distribution, metabolism, excretion and toxicity) properties. This analysis resulted in eight small potential drug-like inhibitors that are in agreement with the binding poses of the crystallographic ADC:fumarate and ADC:isoasparagine complex structures and whose backbone scaffolds seem to be suitable for further experimental studies in therapeutic development against tuberculosis.
format Online
Article
Text
id pubmed-3314653
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-33146532012-04-02 Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase Sharma, Reetu Kothapalli, Roopa Van Dongen, Antonius M. J. Swaminathan, Kunchithapadam PLoS One Research Article L-Aspartate α-decarboxylase (ADC) belongs to a class of pyruvoyl dependent enzymes and catalyzes the conversion of aspartate to β-alanine in the pantothenate pathway, which is critical for the growth of several micro-organisms, including Mycobacterium tuberculosis (Mtb). Its presence only in micro-organisms, fungi and plants and its absence in animals, particularly human, make it a promising drug target. We have followed a chemoinformatics-based approach to identify potential drug-like inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase (MtbADC). The structure-based high throughput virtual screening (HTVS) mode of the Glide program was used to screen 333,761 molecules of the Maybridge, National Cancer Institute (NCI) and Food and Drug Administration (FDA) approved drugs databases. Ligands were rejected if they cross-reacted with S-adenosylmethionine (SAM) decarboxylase, a human pyruvoyl dependent enzyme. The lead molecules were further analyzed for physicochemical and pharmacokinetic parameters, based on Lipinski's rule of five, and ADMET (absorption, distribution, metabolism, excretion and toxicity) properties. This analysis resulted in eight small potential drug-like inhibitors that are in agreement with the binding poses of the crystallographic ADC:fumarate and ADC:isoasparagine complex structures and whose backbone scaffolds seem to be suitable for further experimental studies in therapeutic development against tuberculosis. Public Library of Science 2012-03-28 /pmc/articles/PMC3314653/ /pubmed/22470451 http://dx.doi.org/10.1371/journal.pone.0033521 Text en Sharma et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Sharma, Reetu
Kothapalli, Roopa
Van Dongen, Antonius M. J.
Swaminathan, Kunchithapadam
Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title_full Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title_fullStr Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title_full_unstemmed Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title_short Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase
title_sort chemoinformatic identification of novel inhibitors against mycobacterium tuberculosis l-aspartate α-decarboxylase
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3314653/
https://www.ncbi.nlm.nih.gov/pubmed/22470451
http://dx.doi.org/10.1371/journal.pone.0033521
work_keys_str_mv AT sharmareetu chemoinformaticidentificationofnovelinhibitorsagainstmycobacteriumtuberculosislaspartateadecarboxylase
AT kothapalliroopa chemoinformaticidentificationofnovelinhibitorsagainstmycobacteriumtuberculosislaspartateadecarboxylase
AT vandongenantoniusmj chemoinformaticidentificationofnovelinhibitorsagainstmycobacteriumtuberculosislaspartateadecarboxylase
AT swaminathankunchithapadam chemoinformaticidentificationofnovelinhibitorsagainstmycobacteriumtuberculosislaspartateadecarboxylase