Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells

Abstract Background Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known...

Full description

Bibliographic Details
Main Authors: Bryan G. Harder, Sen Peng, Christopher P. Sereduk, Andrej M. Sodoma, Gaspar J. Kitange, Joseph C. Loftus, Jann N. Sarkaria, Nhan L. Tran
Format: Article
Language:English
Published: BMC 2019-11-01
Series:Molecular Medicine
Subjects:
Online Access:http://link.springer.com/article/10.1186/s10020-019-0116-z
id doaj-11a55e0d187447f9ad9dbba577300d4d
record_format Article
spelling doaj-11a55e0d187447f9ad9dbba577300d4d2020-11-25T04:08:30ZengBMCMolecular Medicine1076-15511528-36582019-11-0125111110.1186/s10020-019-0116-zInhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cellsBryan G. Harder0Sen Peng1Christopher P. Sereduk2Andrej M. Sodoma3Gaspar J. Kitange4Joseph C. Loftus5Jann N. Sarkaria6Nhan L. Tran7Departments of Cancer Biology and Neurosurgery, Mayo Clinic ArizonaCancer and Cell Biology Division, Translational Genomics Research InstituteDepartments of Cancer Biology and Neurosurgery, Mayo Clinic ArizonaDepartments of Cancer Biology and Neurosurgery, Mayo Clinic ArizonaDepartment of Radiation Oncology, Mayo ClinicDepartment of Biochemistry and Molecular Biology, Mayo Clinic ArizonaDepartment of Radiation Oncology, Mayo ClinicDepartments of Cancer Biology and Neurosurgery, Mayo Clinic ArizonaAbstract Background Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known to promote cell survival, including autophagy. As such, targeting these survival pathways may represent new vulnerabilities of GBM after treatment with TMZ. Methods Using the T98G human glioma cell line, we assessed the molecular signaling associated with TMZ treatment, the cellular consequences of using the pan-PI3K inhibitor PX-866, and performed clonogenic assays to determine the effect sequential treatment of TMZ and PX-866 had on colony formation. Additionally, we also use subcutaneous GBM patient derived xenograft (PDX) tumors to show relative LC3 protein expression and correlations between survival pathways and molecular markers which dictate clinical responsiveness to TMZ. Results Here, we report that TMZ can induce autophagic flux in T98G glioma cells. GBM patient-derived xenograft (PDX) tumors treated with TMZ also display an increase in the autophagosome marker LC3 II. Additionally, O6-methylguanine-DNA-methyltransferase (MGMT) expression correlates with PI3K/AKT activity, suggesting that patients with inherent resistance to TMZ (MGMT-high) would benefit from PI3K/AKT inhibitors in addition to TMZ. Accordingly, we have identified that the blood-brain barrier (BBB) penetrant pan-PI3K inhibitor, PX-866, is an early-stage inhibitor of autophagic flux, while maintaining its ability to inhibit PI3K/AKT signaling in glioma cells. Lastly, due to the induction of autophagic flux by TMZ, we provide evidence for sequential treatment of TMZ followed by PX-866, rather than combined co-treatment, as a means to shut down autophagy-induced survival in GBM cells and to enhance apoptosis. Conclusions The understanding of how TMZ induces survival pathways, such as autophagy, may offer new therapeutic vulnerabilities and opportunities to use sequential inhibition of alternate pro-survival pathways that regulate autophagy. As such, identification of additional ways to inhibit TMZ-induced autophagy could enhance the efficacy of TMZ.http://link.springer.com/article/10.1186/s10020-019-0116-zGlioblastoma (GBM)AutophagyTemozolomide (TMZ)PX-866PI3KBafilomycin
collection DOAJ
language English
format Article
sources DOAJ
author Bryan G. Harder
Sen Peng
Christopher P. Sereduk
Andrej M. Sodoma
Gaspar J. Kitange
Joseph C. Loftus
Jann N. Sarkaria
Nhan L. Tran
spellingShingle Bryan G. Harder
Sen Peng
Christopher P. Sereduk
Andrej M. Sodoma
Gaspar J. Kitange
Joseph C. Loftus
Jann N. Sarkaria
Nhan L. Tran
Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
Molecular Medicine
Glioblastoma (GBM)
Autophagy
Temozolomide (TMZ)
PX-866
PI3K
Bafilomycin
author_facet Bryan G. Harder
Sen Peng
Christopher P. Sereduk
Andrej M. Sodoma
Gaspar J. Kitange
Joseph C. Loftus
Jann N. Sarkaria
Nhan L. Tran
author_sort Bryan G. Harder
title Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
title_short Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
title_full Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
title_fullStr Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
title_full_unstemmed Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
title_sort inhibition of phosphatidylinositol 3-kinase by px-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells
publisher BMC
series Molecular Medicine
issn 1076-1551
1528-3658
publishDate 2019-11-01
description Abstract Background Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known to promote cell survival, including autophagy. As such, targeting these survival pathways may represent new vulnerabilities of GBM after treatment with TMZ. Methods Using the T98G human glioma cell line, we assessed the molecular signaling associated with TMZ treatment, the cellular consequences of using the pan-PI3K inhibitor PX-866, and performed clonogenic assays to determine the effect sequential treatment of TMZ and PX-866 had on colony formation. Additionally, we also use subcutaneous GBM patient derived xenograft (PDX) tumors to show relative LC3 protein expression and correlations between survival pathways and molecular markers which dictate clinical responsiveness to TMZ. Results Here, we report that TMZ can induce autophagic flux in T98G glioma cells. GBM patient-derived xenograft (PDX) tumors treated with TMZ also display an increase in the autophagosome marker LC3 II. Additionally, O6-methylguanine-DNA-methyltransferase (MGMT) expression correlates with PI3K/AKT activity, suggesting that patients with inherent resistance to TMZ (MGMT-high) would benefit from PI3K/AKT inhibitors in addition to TMZ. Accordingly, we have identified that the blood-brain barrier (BBB) penetrant pan-PI3K inhibitor, PX-866, is an early-stage inhibitor of autophagic flux, while maintaining its ability to inhibit PI3K/AKT signaling in glioma cells. Lastly, due to the induction of autophagic flux by TMZ, we provide evidence for sequential treatment of TMZ followed by PX-866, rather than combined co-treatment, as a means to shut down autophagy-induced survival in GBM cells and to enhance apoptosis. Conclusions The understanding of how TMZ induces survival pathways, such as autophagy, may offer new therapeutic vulnerabilities and opportunities to use sequential inhibition of alternate pro-survival pathways that regulate autophagy. As such, identification of additional ways to inhibit TMZ-induced autophagy could enhance the efficacy of TMZ.
topic Glioblastoma (GBM)
Autophagy
Temozolomide (TMZ)
PX-866
PI3K
Bafilomycin
url http://link.springer.com/article/10.1186/s10020-019-0116-z
work_keys_str_mv AT bryangharder inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT senpeng inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT christopherpsereduk inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT andrejmsodoma inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT gasparjkitange inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT josephcloftus inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT jannnsarkaria inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
AT nhanltran inhibitionofphosphatidylinositol3kinasebypx866suppressestemozolomideinducedautophagyandpromotesapoptosisinglioblastomacells
_version_ 1724425403620130816