Cargando…

Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells

The CDH1 gene, encoding the cell adhesion protein E-cadherin, is one of the most frequently mutated genes in gastric cancer and inactivating germline CDH1 mutations are responsible for hereditary diffuse gastric cancer syndrome (HDGC). Using cell viability assays, we identified that breast (MCF10A)...

Descripción completa

Detalles Bibliográficos
Autores principales: Bougen-Zhukov, Nicola, Nouri, Yasmin, Godwin, Tanis, Taylor, Megan, Hakkaart, Christopher, Single, Andrew, Brew, Tom, Permina, Elizabeth, Chen, Augustine, Black, Michael A., Guilford, Parry
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6769709/
https://www.ncbi.nlm.nih.gov/pubmed/31540244
http://dx.doi.org/10.3390/cancers11091359
_version_ 1783455301109809152
author Bougen-Zhukov, Nicola
Nouri, Yasmin
Godwin, Tanis
Taylor, Megan
Hakkaart, Christopher
Single, Andrew
Brew, Tom
Permina, Elizabeth
Chen, Augustine
Black, Michael A.
Guilford, Parry
author_facet Bougen-Zhukov, Nicola
Nouri, Yasmin
Godwin, Tanis
Taylor, Megan
Hakkaart, Christopher
Single, Andrew
Brew, Tom
Permina, Elizabeth
Chen, Augustine
Black, Michael A.
Guilford, Parry
author_sort Bougen-Zhukov, Nicola
collection PubMed
description The CDH1 gene, encoding the cell adhesion protein E-cadherin, is one of the most frequently mutated genes in gastric cancer and inactivating germline CDH1 mutations are responsible for hereditary diffuse gastric cancer syndrome (HDGC). Using cell viability assays, we identified that breast (MCF10A) and gastric (NCI-N87) cells lacking CDH1 expression are more sensitive to allosteric AKT inhibitors than their CDH1-expressing isogenic counterparts. Apoptosis priming and total apoptosis assays in the isogenic MCF10A cells confirmed the enhanced sensitivity of E-cadherin-null cells to the AKT inhibitors. In addition, two of these inhibitors, ARQ-092 and MK2206, preferentially targeted mouse-derived gastric Cdh1(−/−) organoids for growth arrest. AKT protein expression and activation (as measured by phosphorylation of serine 473) were differentially regulated in E-cadherin-null MCF10A and NCI-N87 cells, with downregulation in the normal breast cells, but upregulation in the gastric cancer cells. Bioinformatic analysis of the TCGA STAD dataset revealed that AKT3, but not AKT1 or AKT2, is upregulated in the majority of E-cadherin-deficient gastric cancers. In conclusion, allosteric AKT inhibitors represent a promising class of drugs for chemoprevention and chemotherapy of cancers with E-cadherin loss.
format Online
Article
Text
id pubmed-6769709
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-67697092019-10-30 Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells Bougen-Zhukov, Nicola Nouri, Yasmin Godwin, Tanis Taylor, Megan Hakkaart, Christopher Single, Andrew Brew, Tom Permina, Elizabeth Chen, Augustine Black, Michael A. Guilford, Parry Cancers (Basel) Article The CDH1 gene, encoding the cell adhesion protein E-cadherin, is one of the most frequently mutated genes in gastric cancer and inactivating germline CDH1 mutations are responsible for hereditary diffuse gastric cancer syndrome (HDGC). Using cell viability assays, we identified that breast (MCF10A) and gastric (NCI-N87) cells lacking CDH1 expression are more sensitive to allosteric AKT inhibitors than their CDH1-expressing isogenic counterparts. Apoptosis priming and total apoptosis assays in the isogenic MCF10A cells confirmed the enhanced sensitivity of E-cadherin-null cells to the AKT inhibitors. In addition, two of these inhibitors, ARQ-092 and MK2206, preferentially targeted mouse-derived gastric Cdh1(−/−) organoids for growth arrest. AKT protein expression and activation (as measured by phosphorylation of serine 473) were differentially regulated in E-cadherin-null MCF10A and NCI-N87 cells, with downregulation in the normal breast cells, but upregulation in the gastric cancer cells. Bioinformatic analysis of the TCGA STAD dataset revealed that AKT3, but not AKT1 or AKT2, is upregulated in the majority of E-cadherin-deficient gastric cancers. In conclusion, allosteric AKT inhibitors represent a promising class of drugs for chemoprevention and chemotherapy of cancers with E-cadherin loss. MDPI 2019-09-13 /pmc/articles/PMC6769709/ /pubmed/31540244 http://dx.doi.org/10.3390/cancers11091359 Text en © 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Bougen-Zhukov, Nicola
Nouri, Yasmin
Godwin, Tanis
Taylor, Megan
Hakkaart, Christopher
Single, Andrew
Brew, Tom
Permina, Elizabeth
Chen, Augustine
Black, Michael A.
Guilford, Parry
Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title_full Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title_fullStr Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title_full_unstemmed Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title_short Allosteric AKT Inhibitors Target Synthetic Lethal Vulnerabilities in E-Cadherin-Deficient Cells
title_sort allosteric akt inhibitors target synthetic lethal vulnerabilities in e-cadherin-deficient cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6769709/
https://www.ncbi.nlm.nih.gov/pubmed/31540244
http://dx.doi.org/10.3390/cancers11091359
work_keys_str_mv AT bougenzhukovnicola allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT nouriyasmin allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT godwintanis allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT taylormegan allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT hakkaartchristopher allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT singleandrew allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT brewtom allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT perminaelizabeth allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT chenaugustine allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT blackmichaela allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells
AT guilfordparry allostericaktinhibitorstargetsyntheticlethalvulnerabilitiesinecadherindeficientcells