Cargando…

Low-cost anti-mycobacterial drug discovery using engineered E. coli

Whole-cell screening for Mycobacterium tuberculosis (Mtb) inhibitors is complicated by the pathogen’s slow growth and biocontainment requirements. Here we present a synthetic biology framework for assaying Mtb drug targets in engineered E. coli. We construct Target Essential Surrogate E. coli (TESEC...

Descripción completa

Detalles Bibliográficos
Autores principales: Bongaerts, Nadine, Edoo, Zainab, Abukar, Ayan A., Song, Xiaohu, Sosa-Carrillo, Sebastián, Haggenmueller, Sarah, Savigny, Juline, Gontier, Sophie, Lindner, Ariel B., Wintermute, Edwin H.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9262897/
https://www.ncbi.nlm.nih.gov/pubmed/35798732
http://dx.doi.org/10.1038/s41467-022-31570-3
_version_ 1784742599034142720
author Bongaerts, Nadine
Edoo, Zainab
Abukar, Ayan A.
Song, Xiaohu
Sosa-Carrillo, Sebastián
Haggenmueller, Sarah
Savigny, Juline
Gontier, Sophie
Lindner, Ariel B.
Wintermute, Edwin H.
author_facet Bongaerts, Nadine
Edoo, Zainab
Abukar, Ayan A.
Song, Xiaohu
Sosa-Carrillo, Sebastián
Haggenmueller, Sarah
Savigny, Juline
Gontier, Sophie
Lindner, Ariel B.
Wintermute, Edwin H.
author_sort Bongaerts, Nadine
collection PubMed
description Whole-cell screening for Mycobacterium tuberculosis (Mtb) inhibitors is complicated by the pathogen’s slow growth and biocontainment requirements. Here we present a synthetic biology framework for assaying Mtb drug targets in engineered E. coli. We construct Target Essential Surrogate E. coli (TESEC) in which an essential metabolic enzyme is deleted and replaced with an Mtb-derived functional analog, linking bacterial growth to the activity of the target enzyme. High throughput screening of a TESEC model for Mtb alanine racemase (Alr) revealed benazepril as a targeted inhibitor, a result validated in whole-cell Mtb. In vitro biochemical assays indicated a noncompetitive mechanism unlike that of clinical Alr inhibitors. We establish the scalability of TESEC for drug discovery by characterizing TESEC strains for four additional targets.
format Online
Article
Text
id pubmed-9262897
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-92628972022-07-09 Low-cost anti-mycobacterial drug discovery using engineered E. coli Bongaerts, Nadine Edoo, Zainab Abukar, Ayan A. Song, Xiaohu Sosa-Carrillo, Sebastián Haggenmueller, Sarah Savigny, Juline Gontier, Sophie Lindner, Ariel B. Wintermute, Edwin H. Nat Commun Article Whole-cell screening for Mycobacterium tuberculosis (Mtb) inhibitors is complicated by the pathogen’s slow growth and biocontainment requirements. Here we present a synthetic biology framework for assaying Mtb drug targets in engineered E. coli. We construct Target Essential Surrogate E. coli (TESEC) in which an essential metabolic enzyme is deleted and replaced with an Mtb-derived functional analog, linking bacterial growth to the activity of the target enzyme. High throughput screening of a TESEC model for Mtb alanine racemase (Alr) revealed benazepril as a targeted inhibitor, a result validated in whole-cell Mtb. In vitro biochemical assays indicated a noncompetitive mechanism unlike that of clinical Alr inhibitors. We establish the scalability of TESEC for drug discovery by characterizing TESEC strains for four additional targets. Nature Publishing Group UK 2022-07-07 /pmc/articles/PMC9262897/ /pubmed/35798732 http://dx.doi.org/10.1038/s41467-022-31570-3 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Bongaerts, Nadine
Edoo, Zainab
Abukar, Ayan A.
Song, Xiaohu
Sosa-Carrillo, Sebastián
Haggenmueller, Sarah
Savigny, Juline
Gontier, Sophie
Lindner, Ariel B.
Wintermute, Edwin H.
Low-cost anti-mycobacterial drug discovery using engineered E. coli
title Low-cost anti-mycobacterial drug discovery using engineered E. coli
title_full Low-cost anti-mycobacterial drug discovery using engineered E. coli
title_fullStr Low-cost anti-mycobacterial drug discovery using engineered E. coli
title_full_unstemmed Low-cost anti-mycobacterial drug discovery using engineered E. coli
title_short Low-cost anti-mycobacterial drug discovery using engineered E. coli
title_sort low-cost anti-mycobacterial drug discovery using engineered e. coli
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9262897/
https://www.ncbi.nlm.nih.gov/pubmed/35798732
http://dx.doi.org/10.1038/s41467-022-31570-3
work_keys_str_mv AT bongaertsnadine lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT edoozainab lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT abukarayana lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT songxiaohu lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT sosacarrillosebastian lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT haggenmuellersarah lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT savignyjuline lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT gontiersophie lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT lindnerarielb lowcostantimycobacterialdrugdiscoveryusingengineeredecoli
AT wintermuteedwinh lowcostantimycobacterialdrugdiscoveryusingengineeredecoli