Cargando…

Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy

The development of resistance to chemotherapeutic agents, such as Doxorubicin (DOX) and cytarabine (AraC), is one of the greatest challenges to the successful treatment of Acute Myeloid Leukemia (AML). Such acquisition is often underlined by a metabolic reprogramming that can provide a therapeutic o...

Descripción completa

Detalles Bibliográficos
Autores principales: Karakitsou, Effrosyni, Foguet, Carles, Contreras Mostazo, Miriam G., Kurrle, Nina, Schnütgen, Frank, Michaelis, Martin, Cinatl, Jindrich, Marin, Silvia, Cascante, Marta
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Research Network of Computational and Structural Biotechnology 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8326745/
https://www.ncbi.nlm.nih.gov/pubmed/34377370
http://dx.doi.org/10.1016/j.csbj.2021.06.049
_version_ 1783731904002916352
author Karakitsou, Effrosyni
Foguet, Carles
Contreras Mostazo, Miriam G.
Kurrle, Nina
Schnütgen, Frank
Michaelis, Martin
Cinatl, Jindrich
Marin, Silvia
Cascante, Marta
author_facet Karakitsou, Effrosyni
Foguet, Carles
Contreras Mostazo, Miriam G.
Kurrle, Nina
Schnütgen, Frank
Michaelis, Martin
Cinatl, Jindrich
Marin, Silvia
Cascante, Marta
author_sort Karakitsou, Effrosyni
collection PubMed
description The development of resistance to chemotherapeutic agents, such as Doxorubicin (DOX) and cytarabine (AraC), is one of the greatest challenges to the successful treatment of Acute Myeloid Leukemia (AML). Such acquisition is often underlined by a metabolic reprogramming that can provide a therapeutic opportunity, as it can lead to the emergence of vulnerabilities and dependencies to be exploited as targets against the resistant cells. In this regard, genome-scale metabolic models (GSMMs) have emerged as powerful tools to integrate multiple layers of data to build cancer-specific models and identify putative metabolic vulnerabilities. Here, we use genome-scale metabolic modelling to reconstruct a GSMM of the THP1 AML cell line and two derivative cell lines, one with acquired resistance to AraC and the second with acquired resistance to DOX. We also explore how, adding to the transcriptomic layer, the metabolomic layer enhances the selectivity of the resulting condition specific reconstructions. The resulting models enabled us to identify and experimentally validate that drug-resistant THP1 cells are sensitive to the FDA-approved antifolate methotrexate. Moreover, we discovered and validated that the resistant cell lines could be selectively targeted by inhibiting squalene synthase, providing a new and promising strategy to directly inhibit cholesterol synthesis in AML drug resistant cells.
format Online
Article
Text
id pubmed-8326745
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Research Network of Computational and Structural Biotechnology
record_format MEDLINE/PubMed
spelling pubmed-83267452021-08-09 Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy Karakitsou, Effrosyni Foguet, Carles Contreras Mostazo, Miriam G. Kurrle, Nina Schnütgen, Frank Michaelis, Martin Cinatl, Jindrich Marin, Silvia Cascante, Marta Comput Struct Biotechnol J Research Article The development of resistance to chemotherapeutic agents, such as Doxorubicin (DOX) and cytarabine (AraC), is one of the greatest challenges to the successful treatment of Acute Myeloid Leukemia (AML). Such acquisition is often underlined by a metabolic reprogramming that can provide a therapeutic opportunity, as it can lead to the emergence of vulnerabilities and dependencies to be exploited as targets against the resistant cells. In this regard, genome-scale metabolic models (GSMMs) have emerged as powerful tools to integrate multiple layers of data to build cancer-specific models and identify putative metabolic vulnerabilities. Here, we use genome-scale metabolic modelling to reconstruct a GSMM of the THP1 AML cell line and two derivative cell lines, one with acquired resistance to AraC and the second with acquired resistance to DOX. We also explore how, adding to the transcriptomic layer, the metabolomic layer enhances the selectivity of the resulting condition specific reconstructions. The resulting models enabled us to identify and experimentally validate that drug-resistant THP1 cells are sensitive to the FDA-approved antifolate methotrexate. Moreover, we discovered and validated that the resistant cell lines could be selectively targeted by inhibiting squalene synthase, providing a new and promising strategy to directly inhibit cholesterol synthesis in AML drug resistant cells. Research Network of Computational and Structural Biotechnology 2021-07-08 /pmc/articles/PMC8326745/ /pubmed/34377370 http://dx.doi.org/10.1016/j.csbj.2021.06.049 Text en © 2021 The Authors https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Research Article
Karakitsou, Effrosyni
Foguet, Carles
Contreras Mostazo, Miriam G.
Kurrle, Nina
Schnütgen, Frank
Michaelis, Martin
Cinatl, Jindrich
Marin, Silvia
Cascante, Marta
Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title_full Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title_fullStr Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title_full_unstemmed Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title_short Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy
title_sort genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against aml cells resistant to chemotherapy
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8326745/
https://www.ncbi.nlm.nih.gov/pubmed/34377370
http://dx.doi.org/10.1016/j.csbj.2021.06.049
work_keys_str_mv AT karakitsoueffrosyni genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT foguetcarles genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT contrerasmostazomiriamg genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT kurrlenina genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT schnutgenfrank genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT michaelismartin genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT cinatljindrich genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT marinsilvia genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy
AT cascantemarta genomescaleintegrationoftranscriptomeandmetabolomeunveilssqualenesynthaseanddihydrofolatereductaseastargetsagainstamlcellsresistanttochemotherapy