Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells

Recent studies suggest that inhibition of the ATR kinase can potentiate radiation-induced antitumor immune responses, but the extent and mechanisms of such responses in human cancers remain scarcely understood. We aimed to assess whether the ATR inhibitors VE822 and AZD6738, by abrogating the G2 che...

Full description

Bibliographic Details
Main Authors: Adrian Eek Mariampillai, Sissel Hauge, Inger Øynebråten, Gro Elise Rødland, Alexandre Corthay, Randi G. Syljuåsen
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-10-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fonc.2022.981332/full
_version_ 1811257634661072896
author Adrian Eek Mariampillai
Sissel Hauge
Inger Øynebråten
Gro Elise Rødland
Alexandre Corthay
Alexandre Corthay
Randi G. Syljuåsen
author_facet Adrian Eek Mariampillai
Sissel Hauge
Inger Øynebråten
Gro Elise Rødland
Alexandre Corthay
Alexandre Corthay
Randi G. Syljuåsen
author_sort Adrian Eek Mariampillai
collection DOAJ
description Recent studies suggest that inhibition of the ATR kinase can potentiate radiation-induced antitumor immune responses, but the extent and mechanisms of such responses in human cancers remain scarcely understood. We aimed to assess whether the ATR inhibitors VE822 and AZD6738, by abrogating the G2 checkpoint, increase cGAS-mediated type I IFN response after irradiation in human lung cancer and osteosarcoma cell lines. Supporting that the checkpoint may prevent IFN induction, radiation-induced IFN signaling declined when the G2 checkpoint arrest was prolonged at high radiation doses. G2 checkpoint abrogation after co-treatment with radiation and ATR inhibitors was accompanied by increased radiation-induced IFN signaling in four out of five cell lines tested. Consistent with the hypothesis that the cytosolic DNA sensor cGAS may detect DNA from ruptured micronuclei after G2 checkpoint abrogation, cGAS co-localized with micronuclei, and depletion of cGAS or STING abolished the IFN responses. Contrastingly, one lung cancer cell line showed no increase in IFN signaling despite irradiation and G2 checkpoint abrogation. This cell line showed a higher level of the exonuclease TREX1 than the other cell lines, but TREX1 depletion did not enhance IFN signaling. Rather, addition of a pan-caspase inhibitor restored the IFN response in this cell line and also increased the responses in the other cell lines. These results show that treatment-induced caspase activation can suppress the IFN response after co-treatment with radiation and ATR inhibitors. Caspase activation thus warrants further consideration as a possible predictive marker for lack of IFN signaling.
first_indexed 2024-04-12T17:59:21Z
format Article
id doaj.art-7fea646455fc4b5b8ee5d1fb69887e1b
institution Directory Open Access Journal
issn 2234-943X
language English
last_indexed 2024-04-12T17:59:21Z
publishDate 2022-10-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Oncology
spelling doaj.art-7fea646455fc4b5b8ee5d1fb69887e1b2022-12-22T03:22:12ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2022-10-011210.3389/fonc.2022.981332981332Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cellsAdrian Eek Mariampillai0Sissel Hauge1Inger Øynebråten2Gro Elise Rødland3Alexandre Corthay4Alexandre Corthay5Randi G. Syljuåsen6Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, NorwayDepartment of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, NorwayTumor Immunology Lab, Department of Pathology, Oslo University Hospital, Oslo, NorwayDepartment of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, NorwayTumor Immunology Lab, Department of Pathology, Oslo University Hospital, Oslo, NorwayHybrid Technology Hub – Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, NorwayDepartment of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, NorwayRecent studies suggest that inhibition of the ATR kinase can potentiate radiation-induced antitumor immune responses, but the extent and mechanisms of such responses in human cancers remain scarcely understood. We aimed to assess whether the ATR inhibitors VE822 and AZD6738, by abrogating the G2 checkpoint, increase cGAS-mediated type I IFN response after irradiation in human lung cancer and osteosarcoma cell lines. Supporting that the checkpoint may prevent IFN induction, radiation-induced IFN signaling declined when the G2 checkpoint arrest was prolonged at high radiation doses. G2 checkpoint abrogation after co-treatment with radiation and ATR inhibitors was accompanied by increased radiation-induced IFN signaling in four out of five cell lines tested. Consistent with the hypothesis that the cytosolic DNA sensor cGAS may detect DNA from ruptured micronuclei after G2 checkpoint abrogation, cGAS co-localized with micronuclei, and depletion of cGAS or STING abolished the IFN responses. Contrastingly, one lung cancer cell line showed no increase in IFN signaling despite irradiation and G2 checkpoint abrogation. This cell line showed a higher level of the exonuclease TREX1 than the other cell lines, but TREX1 depletion did not enhance IFN signaling. Rather, addition of a pan-caspase inhibitor restored the IFN response in this cell line and also increased the responses in the other cell lines. These results show that treatment-induced caspase activation can suppress the IFN response after co-treatment with radiation and ATR inhibitors. Caspase activation thus warrants further consideration as a possible predictive marker for lack of IFN signaling.https://www.frontiersin.org/articles/10.3389/fonc.2022.981332/fullcell cycle checkpointstype I interferon (IFN) signalingradiation therapy (radiotherapy)micronuclei (MN)ATRcaspase
spellingShingle Adrian Eek Mariampillai
Sissel Hauge
Inger Øynebråten
Gro Elise Rødland
Alexandre Corthay
Alexandre Corthay
Randi G. Syljuåsen
Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
Frontiers in Oncology
cell cycle checkpoints
type I interferon (IFN) signaling
radiation therapy (radiotherapy)
micronuclei (MN)
ATR
caspase
title Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
title_full Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
title_fullStr Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
title_full_unstemmed Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
title_short Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells
title_sort caspase activation counteracts interferon signaling after g2 checkpoint abrogation by atr inhibition in irradiated human cancer cells
topic cell cycle checkpoints
type I interferon (IFN) signaling
radiation therapy (radiotherapy)
micronuclei (MN)
ATR
caspase
url https://www.frontiersin.org/articles/10.3389/fonc.2022.981332/full
work_keys_str_mv AT adrianeekmariampillai caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT sisselhauge caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT ingerøynebraten caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT groeliserødland caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT alexandrecorthay caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT alexandrecorthay caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells
AT randigsyljuasen caspaseactivationcounteractsinterferonsignalingafterg2checkpointabrogationbyatrinhibitioninirradiatedhumancancercells