Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors

Abstract Translocation-related sarcomas (TRSs) harbor an oncogenic fusion gene generated by chromosome translocation and account for approximately one-third of all sarcomas; however, effective targeted therapies have yet to be established. We previously reported that a pan-phosphatidylinositol 3-kin...

Full description

Bibliographic Details
Main Authors: Sho Isoyama, Naomi Tamaki, Yutaka Noguchi, Mutsumi Okamura, Yuki Yoshimatsu, Tadashi Kondo, Takeshi Suzuki, Shin-ichi Yaguchi, Shingo Dan
Format: Article
Language:English
Published: Nature Publishing Group 2023-02-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-023-05690-7
_version_ 1797863389282697216
author Sho Isoyama
Naomi Tamaki
Yutaka Noguchi
Mutsumi Okamura
Yuki Yoshimatsu
Tadashi Kondo
Takeshi Suzuki
Shin-ichi Yaguchi
Shingo Dan
author_facet Sho Isoyama
Naomi Tamaki
Yutaka Noguchi
Mutsumi Okamura
Yuki Yoshimatsu
Tadashi Kondo
Takeshi Suzuki
Shin-ichi Yaguchi
Shingo Dan
author_sort Sho Isoyama
collection DOAJ
description Abstract Translocation-related sarcomas (TRSs) harbor an oncogenic fusion gene generated by chromosome translocation and account for approximately one-third of all sarcomas; however, effective targeted therapies have yet to be established. We previously reported that a pan-phosphatidylinositol 3-kinase (PI3K) inhibitor, ZSTK474, was effective for the treatment of sarcomas in a phase I clinical trial. We also demonstrated the efficacy of ZSTK474 in a preclinical model, particularly in cell lines from synovial sarcoma (SS), Ewing’s sarcoma (ES) and alveolar rhabdomyosarcoma (ARMS), all of which harbor chromosomal translocations. ZSTK474 selectively induced apoptosis in all these sarcoma cell lines, although the precise mechanism underlying the induction of apoptosis remained unclear. In the present study, we aimed to determine the antitumor effect of PI3K inhibitors, particularly with regards to the induction of apoptosis, against various TRS subtypes using cell lines and patient-derived cells (PDCs). All of the cell lines derived from SS (six), ES (two) and ARMS (one) underwent apoptosis accompanied by the cleavage of poly-(ADP-ribose) polymerase (PARP) and the loss of mitochondrial membrane potential. We also observed apoptotic progression in PDCs from SS, ES and clear cell sarcoma (CCS). Transcriptional analyses revealed that PI3K inhibitors triggered the induction of PUMA and BIM and the knockdown of these genes by RNA interference efficiently suppressed apoptosis, suggesting their functional involvement in the progression of apoptosis. In contrast, TRS-derived cell lines/PDCs from alveolar soft part sarcoma (ASPS), CIC-DUX4 sarcoma and dermatofibrosarcoma protuberans failed to undergo apoptosis nor induce PUMA and BIM expression, as well as cell lines derived from non-TRSs and carcinomas. Thus, we conclude that PI3K inhibitors induce apoptosis in selective TRSs such as ES and SS via the induction of PUMA and BIM and the subsequent loss of mitochondrial membrane potential. This represents proof of concept for PI3K-targeted therapy, particularly such TRS patients.
first_indexed 2024-04-09T22:34:47Z
format Article
id doaj.art-449aec3bd3674ec2a73c663ca55309a8
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-04-09T22:34:47Z
publishDate 2023-02-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-449aec3bd3674ec2a73c663ca55309a82023-03-22T12:32:15ZengNature Publishing GroupCell Death and Disease2041-48892023-02-0114211010.1038/s41419-023-05690-7Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitorsSho Isoyama0Naomi Tamaki1Yutaka Noguchi2Mutsumi Okamura3Yuki Yoshimatsu4Tadashi Kondo5Takeshi Suzuki6Shin-ichi Yaguchi7Shingo Dan8Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchDivision of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchDivision of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchDivision of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchDepartment of Patient-derived Cancer Model, Tochigi Cancer CenterDivision of Rare Cancer Research, National Cancer Center Research InstituteDivision of Functional Genomics, Cancer Research Institute, Kanazawa UniversityDivision of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchDivision of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer ResearchAbstract Translocation-related sarcomas (TRSs) harbor an oncogenic fusion gene generated by chromosome translocation and account for approximately one-third of all sarcomas; however, effective targeted therapies have yet to be established. We previously reported that a pan-phosphatidylinositol 3-kinase (PI3K) inhibitor, ZSTK474, was effective for the treatment of sarcomas in a phase I clinical trial. We also demonstrated the efficacy of ZSTK474 in a preclinical model, particularly in cell lines from synovial sarcoma (SS), Ewing’s sarcoma (ES) and alveolar rhabdomyosarcoma (ARMS), all of which harbor chromosomal translocations. ZSTK474 selectively induced apoptosis in all these sarcoma cell lines, although the precise mechanism underlying the induction of apoptosis remained unclear. In the present study, we aimed to determine the antitumor effect of PI3K inhibitors, particularly with regards to the induction of apoptosis, against various TRS subtypes using cell lines and patient-derived cells (PDCs). All of the cell lines derived from SS (six), ES (two) and ARMS (one) underwent apoptosis accompanied by the cleavage of poly-(ADP-ribose) polymerase (PARP) and the loss of mitochondrial membrane potential. We also observed apoptotic progression in PDCs from SS, ES and clear cell sarcoma (CCS). Transcriptional analyses revealed that PI3K inhibitors triggered the induction of PUMA and BIM and the knockdown of these genes by RNA interference efficiently suppressed apoptosis, suggesting their functional involvement in the progression of apoptosis. In contrast, TRS-derived cell lines/PDCs from alveolar soft part sarcoma (ASPS), CIC-DUX4 sarcoma and dermatofibrosarcoma protuberans failed to undergo apoptosis nor induce PUMA and BIM expression, as well as cell lines derived from non-TRSs and carcinomas. Thus, we conclude that PI3K inhibitors induce apoptosis in selective TRSs such as ES and SS via the induction of PUMA and BIM and the subsequent loss of mitochondrial membrane potential. This represents proof of concept for PI3K-targeted therapy, particularly such TRS patients.https://doi.org/10.1038/s41419-023-05690-7
spellingShingle Sho Isoyama
Naomi Tamaki
Yutaka Noguchi
Mutsumi Okamura
Yuki Yoshimatsu
Tadashi Kondo
Takeshi Suzuki
Shin-ichi Yaguchi
Shingo Dan
Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
Cell Death and Disease
title Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
title_full Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
title_fullStr Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
title_full_unstemmed Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
title_short Subtype-selective induction of apoptosis in translocation-related sarcoma cells induced by PUMA and BIM upon treatment with pan-PI3K inhibitors
title_sort subtype selective induction of apoptosis in translocation related sarcoma cells induced by puma and bim upon treatment with pan pi3k inhibitors
url https://doi.org/10.1038/s41419-023-05690-7
work_keys_str_mv AT shoisoyama subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT naomitamaki subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT yutakanoguchi subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT mutsumiokamura subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT yukiyoshimatsu subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT tadashikondo subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT takeshisuzuki subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT shinichiyaguchi subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors
AT shingodan subtypeselectiveinductionofapoptosisintranslocationrelatedsarcomacellsinducedbypumaandbimupontreatmentwithpanpi3kinhibitors