1
|
Roy PK, Deepak K, Das CK, Das A, Biswas A, Jena BC, Mandal M. PSMC2 promotes resistance against temozolomide in glioblastoma via suppressing JNK-mediated autophagic cell death. Biochem Pharmacol 2025; 233:116755. [PMID: 39824465 DOI: 10.1016/j.bcp.2025.116755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Temozolomide is universally used to treat glioblastoma due to its unique ability to cross the blood-brain barrier and inhibit tumor growth through DNA alkylation. However, over time, the inevitable emergence of resistance to temozolomide impedes successful treatment of this cancer. As a result, there is an urgent need to identify new therapeutic targets to improve treatment outcomes for this malignancy. In this work, acquired temozolomide-resistant glioblastoma cell lines LN18 (LN18-TR) and T98G (T98G-TR) exhibited stronger aggressiveness and lower endoplasmic reticulum (ER) stress than their parental cells.. Besides, temozolomide resistance was associated with elevated proteasome activity that suppressed ER stress, which was restored upon inhibition of the proteasome with MG132. Specifically, our study revealed that the 19S proteasomal regulatory subunit PSMC2, which was overexpressed in adapted temozolomide-resistant glioblastoma cells, reduced pro-death autophagy and decreased temozolomide sensitivity in parental cells when overexpressed. While autophagy increased in parental cells following temozolomide treatment, it was not elevated in temozolomide-resistant glioblastoma cells. Genetic suppression of PSMC2 triggered the JNK signalling pathway causing phosphorylation of BCL2, allowing Beclin1 to be released from the BCL2-Beclin1 complex. This boosted autophagosome nucleation, increased pro-death autophagy, and restored apoptosis in temozolomide-resistant glioblastoma cells. Finally, targeting PSMC2 provided a unique method for interrupting autophagy-mediated ER stress maintenance and temozolomide resistance in glioblastoma.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chandan Kanta Das
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, BRBII/III, Philadelphia, PA, 19104, USA
| | - Abhijit Das
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Bikash Chandra Jena
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
2
|
Skouras P, Markouli M, Papadatou I, Piperi C. Targeting epigenetic mechanisms of resistance to chemotherapy in gliomas. Crit Rev Oncol Hematol 2024; 204:104532. [PMID: 39406277 DOI: 10.1016/j.critrevonc.2024.104532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Glioma, an aggressive type of brain tumors of glial origin is highly heterogeneous, posing significant treatment challenges due to its intrinsic resistance to conventional therapeutic schemes. It is characterized by an interplay between epigenetic and genetic alterations in key signaling pathways which further endorse their resistance potential. Aberrant DNA methylation patterns, histone modifications and non-coding RNAs may alter the expression of genes associated with drug response and cell survival, induce gene silencing or deregulate key pathways contributing to glioma resistance. There is evidence that epigenetic plasticity enables glioma cells to adapt dynamically to therapeutic schemes and allow the formation of drug-resistant subpopulations. Furthermore, the tumor microenvironment adds an extra input on epigenetic regulation, increasing the complexity of resistance mechanisms. Herein, we discuss epigenetic changes conferring to drug resistance mechanisms in gliomas in order to delineate novel therapeutic targets and potential approaches that will enable personalized treatment.
Collapse
Affiliation(s)
- Panagiotis Skouras
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece; 1st Department of Neurosurgery, Evangelismos Hospital, National and Kapodistrian University of Athens, Greece.
| | - Mariam Markouli
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Ioanna Papadatou
- University Research Institute for the Study of Genetic & Malignant Disorders in Childhood, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece.
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece.
| |
Collapse
|
3
|
Zhou D, Yin M, Kang B, Yu X, Zeng H, Chen B, Wang G, Song Y, Liu X, He Q, Wu Q, Zhang L, Wu L, Wu Y, Qu N, Li X, Zhou W. CCT020312 exerts anti-prostate cancer effect by inducing G1 cell cycle arrest, apoptosis and autophagy through activation of PERK/eIF2α/ATF4/CHOP signaling. Biochem Pharmacol 2024; 221:116038. [PMID: 38286211 DOI: 10.1016/j.bcp.2024.116038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 01/31/2024]
Abstract
PERK/eIF2α/ATF4/CHOP signaling pathway is one of three major branches of unfolded protein response (UPR) and has been implicated in tumor progression. CCT020312 is a selective PERK activator and may have a potential anti-tumor effect. Here we investigated the anti-prostate cancer effect and its underlying mechanism of CCT020312. Our results showed that CCT020312 inhibited prostate cancer cell viability by inducing cell cycle arrest, apoptosis and autophagy through activation of PERK/eIF2α/ATF4/CHOP signaling. CCT020312 treatment caused cell cycle arrest at G1 phase and increased the levels of cleaved-Caspase3, cleaved-PARP and Bax in prostate cancer C4-2 and LNCaP cells. Moreover, CCT020312 increased LC3II/I, Atg12-Atg5 and Beclin1 levels and induced autophagosome formation. Furthermore, knockdown of CHOP reversed CCT020312-induced cell viability decrease, apoptosis and autophagy. Bafilomycin A1 reversed CCT020312-induced cell viability decrease but had no effect on CCT020312-induced CHOP activation in C4-2 and LNCaP cells. In vivo, CCT020312 suppressed tumor growth in C4-2 cells-derived xenograft mouse model, activated PERK pathway, and induced autophagy and apoptosis. Our study illustrates that CCT020312 exerts an anti-tumor effect in prostate cancer via activating the PERK pathway, thus indicating that CCT020312 may be a potential drug for prostate cancer.
Collapse
Affiliation(s)
- Duanfang Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China; Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing 401147, China
| | - Manjialan Yin
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Baoguo Kang
- Deputy Chief Physician, Department of Oncology, Liangjiang New District People's Hospital
| | - Xiaoping Yu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Hongfang Zeng
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Yi Song
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Xu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Qichen He
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Qiuya Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Limei Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Lihong Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China.
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing 400016, China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing 400016, China.
| |
Collapse
|
4
|
Li R, Chen Y, Yang B, Li Z, Wang S, He J, Zhou Z, Li X, Li J, Sun Y, Guo X, Wang X, Wu Y, Zhang W, Guo G. Integrated bioinformatics analysis and experimental validation identified CDCA families as prognostic biomarkers and sensitive indicators for rapamycin treatment of glioma. PLoS One 2024; 19:e0295346. [PMID: 38181024 PMCID: PMC10769025 DOI: 10.1371/journal.pone.0295346] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/21/2023] [Indexed: 01/07/2024] Open
Abstract
The cell division cycle associated (CDCA) genes regulate the cell cycle; however, their relationship with prognosis in glioma has been poorly reported in the literature. The Cancer Genome Atlas (TCGA) was utilized to probe the CDCA family in relation to the adverse clinical features of glioma. Glioma single-cell atlas reveals specific expression of CDCA3, 4, 5, 8 in malignant cells and CDCA7 in neural progenitor cells (NPC)-like malignant cells. Glioma data from TCGA, the China Glioma Genome Atlas Project (CGGA) and the gene expression omnibus (GEO) database all demonstrated that CDCA2, 3, 4, 5, 7 and 8 are prognostic markers for glioma. Further analysis identified CDCA2, 5 and 8 as independent prognostic factors for glioma. Lasso regression-based risk models for CDCA families demonstrated that high-risk patients were characterized by high tumor mutational burden (TMB), low levels of microsatellite instability (MSI), and low tumor immune dysfunction and rejection (TIDE) scores. These pointed to immunotherapy for glioma as a potentially viable treatment option Further CDCA clustering suggested that the high CDCA subtype exhibited a high macrophage phenotype and was associated with a higher antigen presentation capacity and high levels of immune escape. In addition, hsa-mir-15b-5p was predicted to be common regulator of CDCA3 and CDCA4, which was validated in U87 and U251 cells. Importantly, we found that CDCAs may indicate response to drug treatment, especially rapamycin, in glioma. In summary, our results suggest that CDCAs have potential applications in clinical diagnosis and as drug sensitivity markers in glioma.
Collapse
Affiliation(s)
- Ren Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yang Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Biao Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ziao Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shule Wang
- Department of General and Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianhang He
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zihan Zhou
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xuepeng Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiayu Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanqi Sun
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaolong Guo
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaogang Wang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongqiang Wu
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenju Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Geng Guo
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
5
|
Kusaczuk M, Ambel ET, Naumowicz M, Velasco G. Cellular stress responses as modulators of drug cytotoxicity in pharmacotherapy of glioblastoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189054. [PMID: 38103622 DOI: 10.1016/j.bbcan.2023.189054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Despite the extensive efforts to find effective therapeutic strategies, glioblastoma (GBM) remains a therapeutic challenge with dismal prognosis of survival. Over the last decade the role of stress responses in GBM therapy has gained a great deal of attention, since depending on the duration and intensity of these cellular programs they can be cytoprotective or promote cancer cell death. As such, initiation of the UPR, autophagy or oxidative stress may either impede or facilitate drug-mediated cell killing. In this review, we summarize the mechanisms that regulate ER stress, autophagy, and oxidative stress during GBM development and progression to later discuss the involvement of these stress pathways in the response to different treatments. We also discuss how a precise understanding of the molecular mechanisms regulating stress responses evoked by different pharmacological agents could decisively contribute to the design of novel and more effective combinational treatments against brain malignancies.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland.
| | - Elena Tovar Ambel
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain
| | - Monika Naumowicz
- Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, K. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
6
|
Marjanović M, Mikecin Dražić AM, Mioč M, Paradžik M, Kliček F, Novokmet M, Lauc G, Kralj M. Salinomycin disturbs Golgi function and specifically affects cells in epithelial-to-mesenchymal transition. J Cell Sci 2023; 136:jcs260934. [PMID: 37545292 DOI: 10.1242/jcs.260934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) gives rise to cells with properties similar to cancer stem cells (CSCs). Targeting the EMT program to selectively eliminate CSCs is a promising way to improve cancer therapy. Salinomycin (Sal), a K+/H+ ionophore, was identified as highly selective towards CSC-like cells, but its mechanism of action and selectivity remains elusive. Here, we show that Sal, similar to monensin and nigericin, disturbs the function of the Golgi. Sal alters the expression of Golgi-related genes and leads to marked changes in Golgi morphology, particularly in cells that have undergone EMT. Moreover, Golgi-disturbing agents severely affect post-translational modifications of proteins, including protein processing, glycosylation and secretion. We discover that the alterations induced by Golgi-disturbing agents specifically affect the viability of EMT cells. Collectively, our work reveals a novel vulnerability related to the EMT, suggesting an important role for the Golgi in the EMT and that targeting the Golgi could represent a novel therapeutic approach against CSCs.
Collapse
Affiliation(s)
- Marko Marjanović
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička c. 54, 10000 Zagreb, Croatia
| | - Ana-Matea Mikecin Dražić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička c. 54, 10000 Zagreb, Croatia
| | - Marija Mioč
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička c. 54, 10000 Zagreb, Croatia
| | - Mladen Paradžik
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička c. 54, 10000 Zagreb, Croatia
| | - Filip Kliček
- GENOS, Glycoscience Research Laboratory, Borongajska c. 83, 10000 Zagreb, Croatia
| | - Mislav Novokmet
- GENOS, Glycoscience Research Laboratory, Borongajska c. 83, 10000 Zagreb, Croatia
| | - Gordan Lauc
- GENOS, Glycoscience Research Laboratory, Borongajska c. 83, 10000 Zagreb, Croatia
| | - Marijeta Kralj
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička c. 54, 10000 Zagreb, Croatia
| |
Collapse
|
7
|
Dong L, Xu M, Li Y, Xu W, Wu C, Zheng H, Xiao Z, Sun G, Ding L, Li X, Li W, Zhou L, Xia Q. SMURF1 attenuates endoplasmic reticulum stress by promoting the degradation of KEAP1 to activate NRF2 antioxidant pathway. Cell Death Dis 2023; 14:361. [PMID: 37316499 PMCID: PMC10267134 DOI: 10.1038/s41419-023-05873-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Cancer cells consistently utilize the unfolded protein response (UPR) to encounter the abnormal endoplasmic reticulum (ER) stress induced by the accumulation of misfolded proteins. Extreme activation of the UPR could also provoke maladaptive cell death. Previous reports have shown that NRF2 antioxidant signaling is activated by UPR and serves as noncanonical pathway to defense and reduce excessive ROS levels during ER stress. However, the mechanisms of regulating NRF2 signaling upon ER stress in glioblastoma have not been fully elucidated. Here we identify that SMURF1 protects against ER stress and facilitates glioblastoma cell survival by rewiring KEAP1-NRF2 pathway. We show that ER stress induces SMURF1 degradation. Knockdown of SMURF1 upregulates IRE1 and PERK signaling in the UPR pathway and prevents ER-associated protein degradation (ERAD) activity, leading to cell apoptosis. Importantly, SMURF1 overexpression activates NRF2 signaling to reduce ROS levels and alleviate UPR-mediated cell death. Mechanistically, SMURF1 interacts with and ubiquitinates KEAP1 for its degradation (NRF2 negative regulator), resulting in NRF2 nuclear import. Moreover, SMURF1 loss reduces glioblastoma cell proliferation and growth in subcutaneously implanted nude mice xenografts. Taken together, SMURF1 rewires KEAP1-NRF2 pathway to confer resistance to ER stress inducers and protect glioblastoma cell survival. ER stress and SMURF1 modulation may provide promising therapeutic targets for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Lei Dong
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Mengchuan Xu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yang Li
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Wanting Xu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Chengwei Wu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hanfei Zheng
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhenyu Xiao
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Guochen Sun
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lei Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaobo Li
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Wenming Li
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Liying Zhou
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Qin Xia
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
8
|
An C, Pipia I, Ruiz AS, Argüelles I, An M, Wase S, Peng G. The molecular link between obesity and genomic instability in cancer development. Cancer Lett 2023; 555:216035. [PMID: 36502927 DOI: 10.1016/j.canlet.2022.216035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Obesity has been known to be a major risk factor for various types of cancers for several decades. More recently, the relationship between dysregulated adipokines and cancer development has been the focus of much research. Adipose tissue is an important endocrine organ that secretes adipokines that affect both autocrine and paracrine signaling. These adipokines modulate inflammation, induce insulin resistance, and regulate their own behavior and production. Adipokine-production dysregulation is due to physiological changes in adipose tissue that prompt molecular modifications, including low-grade inflammation and the stimulatory production of reactive oxygen species. Additionally, studies have linked DNA damage response, genomic instability, and the innate immune response to tumorigenesis. Further investigation of adipokines and their role in the promotion of genomic instability may clarify the link between obesity and cancer, as well as elucidate potential pharmaceutical targets. In this review, we discuss the progress of recent literature, focusing on the impact of adipokines, genomic instability, and the innate immune response on increasing the risk of cancer.
Collapse
Affiliation(s)
- Clemens An
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Robert Larner, M.D. College of Medicine at The University of Vermont, Burlington, VT, USA.
| | - Ilissa Pipia
- Department of Biological Sciences, Cornell University, Ithaca, NY, USA
| | - Ana-Sofia Ruiz
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ivonne Argüelles
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martino An
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saima Wase
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Otolaryngology - Head & Neck Surgery, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Širvinskas D, Steponaitis G, Stakaitis R, Tamašauskas A, Vaitkienė P, Skiriutė D. Antisense lncRNA CHROMR is linked to glioma patient survival. Front Mol Biosci 2023; 10:1101953. [PMID: 36950523 PMCID: PMC10025505 DOI: 10.3389/fmolb.2023.1101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Background: Natural non-coding antisense transcripts (ncNATs) are long non-coding RNAs (lncRNA) transcribed from the opposite strand of a separate protein coding or non-coding gene. As such, ncNATs can increase overlapping mRNA (and the coded protein) levels by stabilizing mRNA, absorbing inhibitory miRNAs and protecting the mRNA from degradation, or conversely decrease mRNA (or protein) levels by directing the mRNA towards degradation or inhibiting protein translation. Recently, growing numbers of ncNATs were shown to be dysregulated in cancerous cells, however, actual impact of ncNATs on cancer progression remains largely unknown. We therefore investigated gene expression levels of natural antisense lncRNA CHROMR (Cholesterol Induced Regulator of Metabolism RNA) and its sense protein coding gene PRKRA (Protein Activator of Interferon Induced Protein Kinase EIF2AK2) in gliomas. Next, we checked CHROMR effect on the survival of glioma patients. Methods: We performed RNA-seq on post-surgical tumor samples from 26 glioma patients, and normal brain tissue. Gene expression in TPM values were extracted for CHROMR and PRKRA genes. These data were validated using the TCGA and GTEx gene expression databases. Results: The gene expression level of ncNAT lncRNA CHROMR in glioma tissue was significantly higher compared to healthy brain tissue, while the expression of its sense counterpart protein coding PRKRA mRNA did not differ between glioma and healthy samples. Survival analysis showed lower survival rates in patients with low mRNA PRKRA/lncRNA CHROMR gene expression ratio compared to high ratio showing a link between lncRNA CHROMR and glioma patient survival prognosis. Conclusion: Here we show that elevated levels of lncRNA CHROMR (i.e., low ratio of mRNA PRKRA/lncRNA CHROMR) is associated with poor prognosis for glioma patients.
Collapse
Affiliation(s)
- Dovydas Širvinskas
- Laboratory of Molecular Neurobiology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Giedrius Steponaitis
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rytis Stakaitis
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Arimantas Tamašauskas
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Paulina Vaitkienė
- Laboratory of Molecular Neurobiology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
- *Correspondence: Paulina Vaitkienė,
| | - Daina Skiriutė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
10
|
Mouawad R, Neamati N. Inhibition of Protein Disulfide Isomerase (PDIA1) Leads to Proteasome-Mediated Degradation of Ubiquitin-like PHD and RING Finger Domain-Containing Protein 1 (UHRF1) and Increased Sensitivity of Glioblastoma Cells to Topoisomerase II Inhibitors. ACS Pharmacol Transl Sci 2022; 6:100-114. [PMID: 36654750 PMCID: PMC9841782 DOI: 10.1021/acsptsci.2c00186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, and the prognosis remains poor with current available treatments. PDIA1 is considered a promising therapeutic target in GBM. In this study, we demonstrate that targeting PDIA1 results in increased GBM cell death by topoisomerase II (Top-II) inhibitors resulting in proteasome-mediated degradation of the oncogenic protein UHRF1. Combination of the PDIA1 inhibitor, bepristat-2a, produces strong synergy with doxorubicin, etoposide, and mitoxantrone in GBM and other cancer cell lines. Our bioinformatics analysis of multiple datasets revealed downregulation of UHRF1, upon PDIA1 inhibition. In addition, PDIA1 inhibition results in proteasome-mediated degradation of UHRF1 protein. Interestingly, treatment of GBM cells with bepristat-2a results in increased apoptosis and resistance to ferroptosis. Our findings emphasize the importance of PDIA1 as a therapeutic target in GBM and present a promising new therapeutic approach using Top-II inhibitors for GBM treatment.
Collapse
|
11
|
Sun S, Kiang KMY, Leung GKK. Chaperone protein P4HB predicts temozolomide response and prognosis in malignant glioma. Oncol Lett 2022; 24:264. [PMID: 35765277 DOI: 10.3892/ol.2022.13385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022] Open
Abstract
Prolyl 4-hydroxylase beta polypeptide (P4HB) is a chaperone protein associated with temozolomide (TMZ) resistance through the unfolded protein response. Cancer cells with constitutive activation of endoplasmic reticulum stress and upregulation of P4HB have been observed to show resistance against chemotherapies. The present study focused on the evaluation of the prognostic value of P4HB in subtypes of glioma with or without O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. P4HB expression was assessed by immunohistochemical staining in 73 grade I-IV gliomas and its association with the clinicopathological data was determined. It was indicated that P4HB expression was significantly associated with several parameters, including age, tumour grade and the number of TMZ treatment cycles received. In the Kaplan-Meier analysis, P4HB expression was positively associated with risk of mortality and disease progression. In patients treated with TMZ, high P4HB expression was significantly associated with poor overall survival (OS) and progression-free survival (PFS). The association between MGMT promoter methylation and P4HB expression was also assessed. Patients with MGMTMethP4HBLow tumours had the most favourable PFS (48 months) among cases with various combinations of MGMT methylation status and P4HB expression. Multivariate analysis revealed that P4HB may be used as an independent prognostic indicator of OS, particularly in high-grade gliomas. The present study uncovered the potential role of P4HB in a nuanced pathological stratification during clinical decision-making with respect to MGMT promoter methylation status and TMZ treatment.
Collapse
Affiliation(s)
- Stella Sun
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR 999077, P.R. China
| |
Collapse
|
12
|
Milano L, Charlier CF, Andreguetti R, Cox T, Healing E, Thomé MP, Elliott RM, Samson LD, Masson JY, Lenz G, Henriques JAP, Nohturfft A, Meira LB. A DNA repair-independent role for alkyladenine DNA glycosylase in alkylation-induced unfolded protein response. Proc Natl Acad Sci U S A 2022; 119:e2111404119. [PMID: 35197283 PMCID: PMC8892324 DOI: 10.1073/pnas.2111404119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/08/2022] [Indexed: 01/25/2023] Open
Abstract
Alkylating agents damage DNA and proteins and are widely used in cancer chemotherapy. While cellular responses to alkylation-induced DNA damage have been explored, knowledge of how alkylation affects global cellular stress responses is sparse. Here, we examined the effects of the alkylating agent methylmethane sulfonate (MMS) on gene expression in mouse liver, using mice deficient in alkyladenine DNA glycosylase (Aag), the enzyme that initiates the repair of alkylated DNA bases. MMS induced a robust transcriptional response in wild-type liver that included markers of the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) known to be controlled by XBP1, a key UPR effector. Importantly, this response is significantly reduced in the Aag knockout. To investigate how AAG affects alkylation-induced UPR, the expression of UPR markers after MMS treatment was interrogated in human glioblastoma cells expressing different AAG levels. Alkylation induced the UPR in cells expressing AAG; conversely, AAG knockdown compromised UPR induction and led to a defect in XBP1 activation. To verify the requirements for the DNA repair activity of AAG in this response, AAG knockdown cells were complemented with wild-type Aag or with an Aag variant producing a glycosylase-deficient AAG protein. As expected, the glycosylase-defective Aag does not fully protect AAG knockdown cells against MMS-induced cytotoxicity. Remarkably, however, alkylation-induced XBP1 activation is fully complemented by the catalytically inactive AAG enzyme. This work establishes that, besides its enzymatic activity, AAG has noncanonical functions in alkylation-induced UPR that contribute to cellular responses to alkylation.
Collapse
Affiliation(s)
- Larissa Milano
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, GU2 7WG Guildford, United Kingdom
- Center of Biotechnology, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
- Genome Stability Laboratory, CHU de Quebec Research Center, HDQ Pavilion, Oncology Axis, Québec City, QC G1R 3S3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Center, Quebec City, QC G1V 0A6, Canada
| | - Clara F Charlier
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, GU2 7WG Guildford, United Kingdom
| | - Rafaela Andreguetti
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, GU2 7WG Guildford, United Kingdom
| | - Thomas Cox
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, GU2 7WG Guildford, United Kingdom
| | - Eleanor Healing
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, GU2 7XH Guildford, United Kingdom
| | - Marcos P Thomé
- Department of Biophysics, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
| | - Ruan M Elliott
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, GU2 7XH Guildford, United Kingdom
| | - Leona D Samson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Quebec Research Center, HDQ Pavilion, Oncology Axis, Québec City, QC G1R 3S3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Center, Quebec City, QC G1V 0A6, Canada
| | - Guido Lenz
- Center of Biotechnology, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
- Department of Biophysics, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
| | - João Antonio P Henriques
- Center of Biotechnology, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
- Department of Biophysics, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, Brazil
| | - Axel Nohturfft
- Molecular and Clinical Sciences Research Institute, St. George's University of London, SW17 0RE London, United Kingdom
| | - Lisiane B Meira
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, GU2 7WG Guildford, United Kingdom;
| |
Collapse
|
13
|
Kim TW, Ko SG. The Herbal Formula JI017 Induces ER Stress via Nox4 in Breast Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10121881. [PMID: 34942984 PMCID: PMC8698338 DOI: 10.3390/antiox10121881] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 01/16/2023] Open
Abstract
Chemotherapy is a powerful anti-tumor therapeutic strategy; however, resistance to treatment remains a serious concern. To overcome chemoresistance, combination therapy with anticancer drugs is a potential strategy. We developed a novel herbal extract, JI017, with lower toxicity and lesser side effects. JI017 induced programmed cell death and excessive unfolded protein response through the release of intracellular reactive oxygen species (ROS) and calcium in breast cancer cells. JI017 treatment increased the expression of endoplasmic reticulum (ER) stress markers, including p-PERK, p-eIF2α, ATF4, and CHOP, via the activation of both exosomal GRP78 and cell lysate GRP78. The ROS inhibitors diphenyleneiodonium and N-acetyl cysteine suppressed apoptosis and excessive ER stress by inhibiting Nox4 in JI017-treated breast cancer cells. Furthermore, in paclitaxel-resistant breast cancer cell lines, MCF-7R and MDA-MB-231R, a combination of JI017 and paclitaxel overcame paclitaxel resistance by blocking epithelial-mesenchymal transition (EMT) processes, such as the downregulation of E-cadherin expression and the upregulation of HIF-1α, vimentin, Snail, and Slug expression. These findings suggested that JI017 exerts a powerful anti-cancer effect in breast cancer and a combination therapy of JI017 and paclitaxel may be a potential cancer therapy for paclitaxel resistant breast cancer.
Collapse
Affiliation(s)
| | - Seong-Gyu Ko
- Correspondence: ; Tel.: +82-2-961-0329; Fax: +82-2-961-1165
| |
Collapse
|
14
|
Qi D, Liu Y, Li J, Huang JH, Hu X, Wu E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med Res Rev 2021; 42:1037-1063. [PMID: 34786735 PMCID: PMC9298915 DOI: 10.1002/med.21870] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within a tumor that can both self‐renew and differentiate into other cell types forming the heterogeneous tumor bulk. Since CSCs are involved in all aspects of cancer development, including tumor initiation, cell proliferation, metastatic dissemination, therapy resistance, and recurrence, they have emerged as attractive targets for cancer treatment and management. Salinomycin, a widely used antibiotic in poultry farming, was identified by the Weinberg group as a potent anti‐CSC agent in 2009. As a polyether ionophore, salinomycin exerts broad‐spectrum activities, including the important anti‐CSC function. Studies on the mechanism of action of salinomycin against cancer have been continuously and rapidly published since then. Thus, it is imperative for us to update its literature of recent research findings in this area. We here summarize the notable work reported on salinomycin's anticancer activities, intracellular binding target(s), effects on tumor microenvironment, safety, derivatives, and tumor‐specific drug delivery; after that we also discuss the translational potential of salinomycin toward clinical application based on current multifaceted understandings.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA
| | - Yunyi Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Juan Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Xiaoxiao Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China.,Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA.,LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Pharmaceutical Sciences, Texas A&M University College of Pharmacy, College Station, Texas, USA
| |
Collapse
|
15
|
Wu Q, Berglund AE, Etame AB. The Impact of Epigenetic Modifications on Adaptive Resistance Evolution in Glioblastoma. Int J Mol Sci 2021; 22:8324. [PMID: 34361090 PMCID: PMC8347012 DOI: 10.3390/ijms22158324] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a highly lethal cancer that is universally refractory to the standard multimodal therapies of surgical resection, radiation, and chemotherapy treatment. Temozolomide (TMZ) is currently the best chemotherapy agent for GBM, but the durability of response is epigenetically dependent and often short-lived secondary to tumor resistance. Therapies that can provide synergy to chemoradiation are desperately needed in GBM. There is accumulating evidence that adaptive resistance evolution in GBM is facilitated through treatment-induced epigenetic modifications. Epigenetic alterations of DNA methylation, histone modifications, and chromatin remodeling have all been implicated as mechanisms that enhance accessibility for transcriptional activation of genes that play critical roles in GBM resistance and lethality. Hence, understanding and targeting epigenetic modifications associated with GBM resistance is of utmost priority. In this review, we summarize the latest updates on the impact of epigenetic modifications on adaptive resistance evolution in GBM to therapy.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| |
Collapse
|
16
|
Qiu X, Tan G, Wen H, Lian L, Xiao S. Forkhead box O1 targeting replication factor C subunit 2 expression promotes glioma temozolomide resistance and survival. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:692. [PMID: 33987390 PMCID: PMC8105996 DOI: 10.21037/atm-21-1523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Additional mechanisms of temozolomide (TMZ) resistance in gliomas remain uncertain. The aim of this study was to identify another DNA repair mechanism involving forkhead box O1 (FoxO1) and replicator C2 (RFC2) in gliomas. Methods We established glioma cells against TMZ, U87R, by exposure to TMZ. Proliferation rate Cell counting kit-8 (CCK8) was used, and epithelial-mesenchymal transition (EMT)-related markers were detected by western blot. The association between FoxO1 and RFC2 was analyzed by heat maps and scatter plot, and Real-time reverse transcription polymerase chain reaction (qRT-PCR) and Western blot were used to detect the effect of FoxO1 on the expression of RFC2. The regulation effect of FoxO1 on RFC2 expression was analyzed by luciferase reporter gene assay. Knockdown of FoxO1/RFC2 was achieved via short hairpin RNA (shRNA), the effect of knockdown on the proliferation was determined by CCK8 assay and colony formation assay, and apoptosis was examined by flow cytometry and immunoblotting. Results The TMZ-resistant glioma cell line, U87R, was established. The FoxO1 and RFC2 proteins increased significantly in U87R. The expression of FoxO1 and RFC2 were positively related in glioma tissues. We found that FoxO1 contributes to TMZ resistance and cell survival via regulating the expression of RFC2. Moreover, FoxO1 functions as a transcriptional activator to RFC2 by binding to the promoter of RFC2. Furthermore, knockdown of FoxO1/RFC2 suppressed cell proliferation, TMZ resistance, and induced apoptosis in U87R. Conclusions The FoxO1/RFC2 signaling pathway promotes glioma cell proliferation and TMZ resistance, suggesting that the FoxO1/RFC2 pathway may be a potential target for TMZ-resistant glioma therapy.
Collapse
Affiliation(s)
- Xingsheng Qiu
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guifeng Tan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Wen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lian Lian
- Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Lim YC, Ensbey KS, Offenhäuser C, D'souza RCJ, Cullen JK, Stringer BW, Quek H, Bruce ZC, Kijas A, Cianfanelli V, Mahboubi B, Smith F, Jeffree RL, Wiesmüeller L, Wiegmans AP, Bain A, Lombard FJ, Roberts TL, Khanna KK, Lavin MF, Kim B, Hamerlik P, Johns TG, Coster MJ, Boyd AW, Day BW. Simultaneous targeting of DNA replication and homologous recombination in glioblastoma with a polyether ionophore. Neuro Oncol 2021; 22:216-228. [PMID: 31504812 PMCID: PMC7442340 DOI: 10.1093/neuonc/noz159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite significant endeavor having been applied to identify effective therapies to treat glioblastoma (GBM), survival outcomes remain intractable. The greatest nonsurgical benefit arises from radiotherapy, though tumors typically recur due to robust DNA repair. Patients could therefore benefit from therapies with the potential to prevent DNA repair and synergize with radiotherapy. In this work, we investigated the potential of salinomycin to enhance radiotherapy and further uncover novel dual functions of this ionophore to induce DNA damage and prevent repair. METHODS In vitro primary GBM models and ex vivo GBM patient explants were used to determine the mechanism of action of salinomycin by immunoblot, flow cytometry, immunofluorescence, immunohistochemistry, and mass spectrometry. In vivo efficacy studies were performed using orthotopic GBM animal xenograft models. Salinomycin derivatives were synthesized to increase drug efficacy and explore structure-activity relationships. RESULTS Here we report novel dual functions of salinomycin. Salinomycin induces toxic DNA lesions and prevents subsequent recovery by targeting homologous recombination (HR) repair. Salinomycin appears to target the more radioresistant GBM stem cell-like population and synergizes with radiotherapy to significantly delay tumor formation in vivo. We further developed salinomycin derivatives which display greater efficacy in vivo while retaining the same beneficial mechanisms of action. CONCLUSION Our findings highlight the potential of salinomycin to induce DNA lesions and inhibit HR to greatly enhance the effect of radiotherapy. Importantly, first-generation salinomycin derivatives display greater efficacy and may pave the way for clinical testing of these agents.
Collapse
Affiliation(s)
- Yi Chieh Lim
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia.,Brain Tumor Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kathleen S Ensbey
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Carolin Offenhäuser
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Rochelle C J D'souza
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Jason K Cullen
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Brett W Stringer
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Hazel Quek
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Zara C Bruce
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | | | - Valentina Cianfanelli
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bijan Mahboubi
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Fiona Smith
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Rosalind L Jeffree
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, Queensland, Australia
| | - Lisa Wiesmüeller
- Department of Obstetrics and Gynaecology, University of Ulm, Ulm, Germany
| | - Adrian P Wiegmans
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Amanda Bain
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Fanny J Lombard
- University of Queensland, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Tara L Roberts
- School of Medicine, Ingham Institute, New South Wales, Australia
| | - Kum Kum Khanna
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Martin F Lavin
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia
| | - Baek Kim
- Center for Drug Discovery, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Mark J Coster
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Andrew W Boyd
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia.,University of Queensland, Queensland, Australia
| | - Bryan W Day
- Cell and Molecular Biology Department, QIMR Berghofer MRI, Queensland, Australia.,University of Queensland, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
| |
Collapse
|
18
|
Uddin MS, Mamun AA, Alghamdi BS, Tewari D, Jeandet P, Sarwar MS, Ashraf GM. Epigenetics of glioblastoma multiforme: From molecular mechanisms to therapeutic approaches. Semin Cancer Biol 2020; 83:100-120. [PMID: 33370605 DOI: 10.1016/j.semcancer.2020.12.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common form of brain cancer and one of the most aggressive cancers found in humans. Most of the signs and symptoms of GBM can be mild and slowly aggravated, although other symptoms might demonstrate it as an acute ailment. However, the precise mechanisms of the development of GBM remain unknown. Due to the improvement of molecular pathology, current researches have reported that glioma progression is strongly connected with different types of epigenetic phenomena, such as histone modifications, DNA methylation, chromatin remodeling, and aberrant microRNA. Furthermore, the genes and the proteins that control these alterations have become novel targets for treating glioma because of the reversibility of epigenetic modifications. In some cases, gene mutations including P16, TP53, and EGFR, have been observed in GBM. In contrast, monosomies, including removals of chromosome 10, particularly q23 and q25-26, are considered the standard markers for determining the development and aggressiveness of GBM. Recently, amid the epigenetic therapies, histone deacetylase inhibitors (HDACIs) and DNA methyltransferase inhibitors have been used for treating tumors, either single or combined. Specifically, HDACIs are served as a good choice and deliver a novel pathway to treat GBM. In this review, we focus on the epigenetics of GBM and the consequence of its mutations. We also highlight various treatment approaches, namely gene editing, epigenetic drugs, and microRNAs to combat GBM.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687, Reims Cedex 2, France
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
19
|
Drug repositioning of antiretroviral ritonavir for combinatorial therapy in glioblastoma. Eur J Cancer 2020; 140:130-139. [PMID: 33091717 DOI: 10.1016/j.ejca.2020.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND The protease inhibitor ritonavir (RTV) is a clinical-stage inhibitor of the human immunodeficiency virus. In a drug repositioning approach, we here exhibit the additional potential of RTV to augment current treatment of glioblastoma, the most aggressive primary brain tumour of adulthood. METHODS We explored the antitumour activity of RTV and mechanisms of action in a broad spectrum of short-term expanded clinical cell samples from primary and recurrent glioblastoma and in a cohort of conventional cell lines and non-tumour human neural controls in vitro. To validate RTV efficacy in monotherapeutic and in combinatorial settings, we used patient-derived xenograft models in a series of in vivo studies. RESULTS RTV monotherapy induced a selective antineoplastic response and demonstrated cytostatic and anti-migratory activity at clinical plasma peak levels. Additional exposure to temozolomide or irradiation further enhanced the effects synergistically, fostered by mechanisms of autophagy and increased endoplasmic reticulum stress. In xenograft models, we consequently observed increasing overall survival under the combinatorial effect of RTV and temozolomide. CONCLUSIONS Our data establish RTV as a valuable repositioning candidate for further exploration as an adjunct therapeutic in the clinical care of glioblastoma.
Collapse
|
20
|
Xu H, Liu P, Yan Y, Fang K, Liang D, Hou X, Zhang X, Wu S, Ma J, Wang R, Li T, Piao H, Meng S. FKBP9 promotes the malignant behavior of glioblastoma cells and confers resistance to endoplasmic reticulum stress inducers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:44. [PMID: 32111229 PMCID: PMC7048151 DOI: 10.1186/s13046-020-1541-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/05/2020] [Indexed: 02/08/2023]
Abstract
Background FK506-binding protein 9 (FKBP9) is amplified in high-grade gliomas (HGGs). However, the roles and mechanism(s) of FKBP9 in glioma are unknown. Methods The expression of FKBP9 in clinical glioma tissues was detected by immunohistochemistry (IHC). The correlation between FKBP9 expression levels and the clinical prognosis of glioma patients was examined by bioinformatic analysis. Glioblastoma (GBM) cell lines stably depleted of FKBP9 were established using lentiviruses expressing shRNAs against FKBP9. The effects of FKBP9 on GBM cells were determined by cell-based analyses, including anchorage-independent growth, spheroid formation, transwell invasion assay, confocal microscopy, immunoblot (IB) and coimmunoprecipitation assays. In vivo tumor growth was determined in both chick chorioallantoic membrane (CAM) and mouse xenograft models. Results High FKBP9 expression correlated with poor prognosis in glioma patients. Knockdown of FKBP9 markedly suppressed the malignant phenotype of GBM cells in vitro and inhibited tumor growth in vivo. Mechanistically, FKBP9 expression induced the activation of p38MAPK signaling via ASK1. Furthermore, ASK1-p38 signaling contributed to the FKBP9-mediated effects on GBM cell clonogenic growth. In addition, depletion of FKBP9 activated the IRE1α-XBP1 pathway, which played a role in the FKBP9-mediated oncogenic effects. Importantly, FKBP9 expression conferred GBM cell resistance to endoplasmic reticulum (ER) stress inducers that caused FKBP9 ubiquitination and degradation. Conclusions Our findings suggest an oncogenic role for FKBP9 in GBM and reveal FKBP9 as a novel mediator in the IRE1α-XBP1 pathway.
Collapse
Affiliation(s)
- Huizhe Xu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Peng Liu
- Department of General Surgery, Shenzhen University General Hospital, No. 1098, Xueyuan avenue, Shenzhen, 518055, China
| | - Yumei Yan
- The First Department of Ultrasound, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116021, Liaoning Province, China
| | - Kun Fang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Dapeng Liang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Xiukun Hou
- The First Department of Ultrasound, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116021, Liaoning Province, China
| | - Xiaohong Zhang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Songyan Wu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Jianmei Ma
- Department of Anatomy, Dalian Medical University, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China
| | - Ruoyu Wang
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, No.6 Jiefang Street, Dalian, 116001, Liaoning Province, China.
| | - Tao Li
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, Liaoning Province, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, 9 Lvshun Road South, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
21
|
da Silva DC, Valentão P, Andrade PB, Pereira DM. Endoplasmic reticulum stress signaling in cancer and neurodegenerative disorders: Tools and strategies to understand its complexity. Pharmacol Res 2020; 155:104702. [PMID: 32068119 DOI: 10.1016/j.phrs.2020.104702] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022]
Abstract
The endoplasmic reticulum (ER) comprises a network of tubules and vesicles that constitutes the largest organelle of the eukaryotic cell. Being the location where most proteins are synthesized and folded, it is crucial for the upkeep of cellular homeostasis. Disturbed ER homeostasis triggers the activation of a conserved molecular machinery, termed the unfolded protein response (UPR), that comprises three major signaling branches, initiated by the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1) and the activating transcription factor 6 (ATF6). Given the impact of this intricate signaling network upon an extensive list of cellular processes, including protein turnover and autophagy, ER stress is involved in the onset and progression of multiple diseases, including cancer and neurodegenerative disorders. There is, for this reason, an increasing number of publications focused on characterizing and/or modulating ER stress, which have resulted in a wide array of techniques employed to study ER-related molecular events. This review aims to sum up the essentials on the current knowledge of the molecular biology of endoplasmic reticulum stress, while highlighting the available tools used in studies of this nature.
Collapse
Affiliation(s)
- Daniela Correia da Silva
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-213, Porto, Portugal
| | - Patrícia Valentão
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-213, Porto, Portugal
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-213, Porto, Portugal
| | - David M Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-213, Porto, Portugal.
| |
Collapse
|
22
|
Chang CY, Li JR, Wu CC, Wang JD, Liao SL, Chen WY, Wang WY, Chen CJ. Endoplasmic Reticulum Stress Contributes to Indomethacin-Induced Glioma Apoptosis. Int J Mol Sci 2020; 21:557. [PMID: 31952288 PMCID: PMC7013513 DOI: 10.3390/ijms21020557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
The dormancy of cellular apoptotic machinery has been highlighted as a crucial factor in therapeutic resistance, recurrence, and poor prognosis in patients with malignancy, such as malignant glioma. Increasing evidence indicates that nonsteroidal anti-inflammatory drugs (NSAIDs) confer chemopreventive effects, and indomethacin has been shown to have a novel chemotherapeutic application targeting glioma cells. To extend these findings, herein, we studied the underlying mechanisms of apoptosis activation caused by indomethacin in human H4 and U87 glioma cells. We found that the glioma cell-killing effects of indomethacin involved both death receptor- and mitochondria-mediated apoptotic cascades. Indomethacin-induced glioma cell apoptosis was accompanied by a series of biochemical changes, including reactive oxygen species generation, endoplasmic reticulum (ER) stress, apoptosis signal-regulating kinase-1 (Ask1) activation, p38 hyperphosphorylation, protein phosphatase 2A (PP2A) activation, Akt dephosphorylation, Mcl-1 and FLICE-inhibiting protein (FLIP) downregulation, Bax mitochondrial distribution, and caspases 3/caspase 8/caspase 9 activation. Data on pharmacological inhibition related to oxidative stress, ER stress, free Ca2+, and p38 revealed that the axis of oxidative stress/ER stress/Ask1/p38/PP2A/Akt comprised an apoptotic cascade leading to Mcl-1/FLIP downregulation and glioma apoptosis. Since indomethacin is an emerging choice in chemotherapy and its antineoplastic effects have been demonstrated in glioma tumor-bearing models, the findings further strengthen the argument for turning on the aforementioned axis in order to activate the apoptotic machinery of glioma cells.
Collapse
Affiliation(s)
- Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan;
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Financial Engineering, Providence University, Taichung City 433, Taiwan
- Department of Data Science and Big Data Analytics, Providence University, Taichung City 433, Taiwan
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City 407, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan;
| | - Wen-Yi Wang
- Department of Nursing, HungKuang University, Taichung City 433, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
| |
Collapse
|
23
|
Martinez-Velez N, Marigil M, García-Moure M, Gonzalez-Huarriz M, Aristu JJ, Ramos-García LI, Tejada S, Díez-Valle R, Patiño-García A, Becher OJ, Gomez-Manzano C, Fueyo J, Alonso MM. Delta-24-RGD combined with radiotherapy exerts a potent antitumor effect in diffuse intrinsic pontine glioma and pediatric high grade glioma models. Acta Neuropathol Commun 2019; 7:64. [PMID: 31036068 PMCID: PMC6487528 DOI: 10.1186/s40478-019-0714-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 04/02/2019] [Indexed: 01/17/2023] Open
Abstract
Pediatric high grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPGs), are aggressive tumors with a dismal outcome. Radiotherapy (RT) is part of the standard of care of these tumors; however, radiotherapy only leads to a transient clinical improvement. Delta-24-RGD is a genetically engineered tumor-selective adenovirus that has shown safety and clinical efficacy in adults with recurrent gliomas. In this work, we evaluated the feasibility, safety and therapeutic efficacy of Delta-24-RGD in combination with radiotherapy in pHGGs and DIPGs models. Our results showed that the combination of Delta-24-RGD with radiotherapy was feasible and resulted in a synergistic anti-glioma effect in vitro and in vivo in pHGG and DIPG models. Interestingly, Delta-24-RGD treatment led to the downregulation of relevant DNA damage repair proteins, further sensitizing tumors cells to the effect of radiotherapy. Additionally, Delta-24-RGD/radiotherapy treatment significantly increased the trafficking of immune cells (CD3, CD4+ and CD8+) to the tumor niche compared with single treatments. In summary, administration of the Delta-24-RGD/radiotherapy combination to pHGG and DIPG models is safe and significantly increases the overall survival of mice bearing these tumors. Our data offer a rationale for the combination Delta-24-RGD/radiotherapy as a therapeutic option for children with these tumors. SIGNIFICANCE: Delta-24-RGD/radiotherapy administration is safe and significantly increases the survival of treated mice. These positive data underscore the urge to translate this approach to the clinical treatment of children with pHGG and DIPGs.
Collapse
|
24
|
Pinkham K, Park DJ, Hashemiaghdam A, Kirov AB, Adam I, Rosiak K, da Hora CC, Teng J, Cheah PS, Carvalho L, Ganguli-Indra G, Kelly A, Indra AK, Badr CE. Stearoyl CoA Desaturase Is Essential for Regulation of Endoplasmic Reticulum Homeostasis and Tumor Growth in Glioblastoma Cancer Stem Cells. Stem Cell Reports 2019; 12:712-727. [PMID: 30930246 PMCID: PMC6450460 DOI: 10.1016/j.stemcr.2019.02.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/23/2019] [Accepted: 02/26/2019] [Indexed: 12/20/2022] Open
Abstract
Inherent plasticity and various survival cues allow glioblastoma stem-like cells (GSCs) to survive and proliferate under intrinsic and extrinsic stress conditions. Here, we report that GSCs depend on the adaptive activation of ER stress and subsequent activation of lipogenesis and particularly stearoyl CoA desaturase (SCD1), which promotes ER homeostasis, cytoprotection, and tumor initiation. Pharmacological targeting of SCD1 is particularly toxic due to the accumulation of saturated fatty acids, which exacerbates ER stress, triggers apoptosis, impairs RAD51-mediated DNA repair, and achieves a remarkable therapeutic outcome with 25%-100% cure rate in xenograft mouse models. Mechanistically, divergent cell fates under varying levels of ER stress are primarily controlled by the ER sensor IRE1, which either promotes SCD1 transcriptional activation or converts to apoptotic signaling when SCD1 activity is impaired. Taken together, the dependence of GSCs on fatty acid desaturation presents an exploitable vulnerability to target glioblastoma.
Collapse
Affiliation(s)
- Kelsey Pinkham
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - David Jaehyun Park
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Arsalan Hashemiaghdam
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Aleksandar B Kirov
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Isam Adam
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Kamila Rosiak
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Cintia C da Hora
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Jian Teng
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Pike See Cheah
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Selangor 43400, Malaysia
| | - Litia Carvalho
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Avalon Kelly
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Christian E Badr
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
25
|
He Y, Su J, Lan B, Gao Y, Zhao J. Targeting off-target effects: endoplasmic reticulum stress and autophagy as effective strategies to enhance temozolomide treatment. Onco Targets Ther 2019; 12:1857-1865. [PMID: 30881038 PMCID: PMC6413742 DOI: 10.2147/ott.s194770] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive adult primary central nervous system tumor. Unfortunately, GBM is resistant to the classic chemotherapy drug, temozolomide (TMZ). As well as its classic DNA-targeting effects, the off-target effects of TMZ can have pro-survival or pro-death roles and regulate GBM chemoradiation sensitivity. Endoplasmic reticulum (ER) stress is one of the most common off-target effects. ER stress and its downstream induction of autophagy, apoptosis, and other events have important roles in regulating TMZ sensitivity. Autophagy is an evolutionarily conserved cellular homeostasis mechanism that is closely associated with ER stress-induced apoptosis. Under ER stress, autophagy cannot only remove misfolded/unfolded proteins and damaged organelles and degrade and inhibit apoptosis-related caspase activation to reduce cell damage, but may also promote apoptosis dependent on ER stress intensity. Although some protein interactions between autophagy and apoptosis and common upstream signaling pathways have been found, the underlying regulatory mechanisms are still not fully understood. This review summarizes the possible mechanisms underlying the current known off-target roles of ER stress and downstream autophagy in the regulation of cell fate and evaluates their role in TMZ treatment and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yichun He
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China,
| | - Jing Su
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Beiwu Lan
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China,
| | - Yufei Gao
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China,
| | - Jingxia Zhao
- Experimental Teaching Center, School of Nursing, Jilin University, Changchun, Jilin, China,
| |
Collapse
|
26
|
Yang B, Ma YB, Chu SH. Silencing SATB1 overcomes temozolomide resistance by downregulating MGMT expression and upregulating SLC22A18 expression in human glioblastoma cells. Cancer Gene Ther 2018; 25:309-316. [PMID: 30140041 DOI: 10.1038/s41417-018-0040-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant tumor of the central nervous system and has a very poor prognosis. Currently, patients were treated by resection followed by radiotherapy plus concurrent temozolomide (TMZ) chemotherapy. However, many patients are resistant to TMZ-induced DNA damage because of upregulated expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT). In this study, upregulation of SATB1 and MGMT, and downregulation of SLC22A18 resulted in acquisition of TMZ resistance in GBM U87 cells. Inactivation of special AT-rich sequence-binding protein 1 (SATB1) using short hairpin RNA (shRNA) downregulated MGMT expression and upregulated solute carrier family 22 member 18 (SLC22A18) expression in GBM cells. This suggested SATB1-mediated posttranscriptional regulation of the MGMT and SLC22A18 protein levels. Immunohistochemical analysis of malignant glioma specimens demonstrated a significant positive correlation between the levels of MGMT and SATB1, and a negative correlation between the levels of SLC22A18 and SATB1. Importantly, in recurrent, compared with the primary, lesions in 15 paired identical tumors, the SATB1 and MGMT protein levels were increased and the SLC22A18 levels were decreased. Finally, in TMZ-resistant GBM, SATB1 knockdown enhanced TMZ efficacy. Consequently, SATB1 inhibition might be a promising strategy combined with TMZ chemotherapy to treat TMZ-resistant GBM.
Collapse
Affiliation(s)
- Biao Yang
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Yan-Bin Ma
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Sheng-Hua Chu
- Department of Neurosurgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
27
|
Zang L, Kondengaden SM, Che F, Wang L, Heng X. Potential Epigenetic-Based Therapeutic Targets for Glioma. Front Mol Neurosci 2018; 11:408. [PMID: 30498431 PMCID: PMC6249994 DOI: 10.3389/fnmol.2018.00408] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
Glioma is characterized by a high recurrence rate, short survival times, high rates of mortality and treatment difficulties. Surgery, chemotherapy and radiation (RT) are the standard treatments, but outcomes rarely improve even after treatment. With the advancement of molecular pathology, recent studies have found that the development of glioma is closely related to various epigenetic phenomena, including DNA methylation, abnormal microRNA (miRNA), chromatin remodeling and histone modifications. Owing to the reversibility of epigenetic modifications, the proteins and genes that regulate these changes have become new targets in the treatment of glioma. In this review, we present a summary of the potential therapeutic targets of glioma and related effective treating drugs from the four aspects mentioned above. We further illustrate how epigenetic mechanisms dynamically regulate the pathogenesis and discuss the challenges of glioma treatment. Currently, among the epigenetic treatments, DNA methyltransferase (DNMT) inhibitors and histone deacetylase inhibitors (HDACIs) can be used for the treatment of tumors, either individually or in combination. In the treatment of glioma, only HDACIs remain a good option and they provide new directions for the treatment. Due to the complicated pathogenesis of glioma, epigenetic applications to glioma clinical treatment are still limited.
Collapse
Affiliation(s)
- Lanlan Zang
- Central Laboratory and Key Laboratory of Neurophysiology, Linyi People's Hospital, Shandong University, Linyi, China.,Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Shukkoor Muhammed Kondengaden
- Chemistry Department and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, United States
| | - Fengyuan Che
- Central Laboratory and Key Laboratory of Neurophysiology, Linyi People's Hospital, Shandong University, Linyi, China.,Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, China
| | - Lijuan Wang
- Central Laboratory and Key Laboratory of Neurophysiology, Linyi People's Hospital, Shandong University, Linyi, China
| | - Xueyuan Heng
- Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, China
| |
Collapse
|
28
|
Thanasupawat T, Glogowska A, Burg M, Krcek J, Beiko J, Pitz M, Zhang G, Hombach‐Klonisch S, Klonisch T. C1q/TNF-related peptide 8 (CTRP8) promotes temozolomide resistance in human glioblastoma. Mol Oncol 2018; 12:1464-1479. [PMID: 29949238 PMCID: PMC6120254 DOI: 10.1002/1878-0261.12349] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 06/09/2018] [Accepted: 06/10/2018] [Indexed: 02/05/2023] Open
Abstract
The C1q/TNF-related peptide 8 (CTRP8) has recently emerged as a novel ligand of the G protein-coupled receptor RXFP1 in the fatal brain tumor glioblastoma (GBM). We previously demonstrated that the CTRP8-RXFP1 ligand-receptor system promotes motility and matrix invasion of patient GBM and U87 MG cells by specific phosphorylation of PI3 kinase and protein kinase C. Here, we demonstrate a novel role for CTRP8 in protecting human GBM cells against the DNA alkylating damage of temozolomide (TMZ), the standard chemotherapy drug used to treat GBM. This DNA protective role of CTRP8 required a functional RXFP1-STAT3 signaling cascade in GBM cells. We identified N-methylpurine DNA glycosylase (MPG), a monofunctional glycosylase that initiates base excision repair pathway by generating an apurinic/apyrimidinic (AP) site, as a new CTRP8-RXFP1-STAT3 target in GBM. Upon TMZ exposure, treatment with CTRP8 reduced the formation of AP sites and double-strand DNA breaks in GBM cells. This CTRP8 effect was independent of cellular MGMT levels and was associated with decreased caspase 3/7 activity and increased survival of human GBM. CTRP8-induced RXFP1 activation caused an increase in cellular protein levels of the anti-apoptotic Bcl members and STAT3 targets Bcl-2 and Bcl-XL in human GBM. Collectively, our results demonstrate a novel multipronged and clinically relevant mechanism by which the CTRP8-RXFP1 ligand-receptor system exerts a DNA protective function against TMZ chemotherapeutic stress in GBM. This CTRP8-RXFP1-STAT3 axis is a novel determinant of TMZ responsiveness/chemoresistance and an emerging new drug target for improved treatment of human GBM.
Collapse
Affiliation(s)
- Thatchawan Thanasupawat
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Aleksandra Glogowska
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Maxwell Burg
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Jerry Krcek
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
- Department of SurgeryFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Jason Beiko
- Department of SurgeryFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Marshall Pitz
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
- Department of Internal MedicineFaculty of MedicineUniversity of ManitobaWinnipegCanada
- Research Institute in Oncology and Hematology (RIOH)CancerCare ManitobaWinnipegCanada
| | - Guo‐Jun Zhang
- ChangJiang Scholar's LaboratoryShantou University Medical CollegeChina
| | - Sabine Hombach‐Klonisch
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell ScienceFaculty of MedicineUniversity of ManitobaWinnipegCanada
- Department of SurgeryFaculty of MedicineUniversity of ManitobaWinnipegCanada
- Research Institute in Oncology and Hematology (RIOH)CancerCare ManitobaWinnipegCanada
- Department of Medical Microbiology & Infectious DiseasesFaculty of MedicineUniversity of ManitobaWinnipegCanada
| |
Collapse
|
29
|
Wang C, Jiang M, Hou H, Lin Q, Yan Z, Zhang X. Apatinib suppresses cell growth and metastasis and promotes antitumor activity of temozolomide in glioma. Oncol Lett 2018; 16:5607-5614. [PMID: 30344715 PMCID: PMC6176256 DOI: 10.3892/ol.2018.9355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/30/2018] [Indexed: 01/14/2023] Open
Abstract
Apatinib, a small-molecule multi-targeted tyrosine kinase inhibitor, is widely used to treat various types of solid tumors. In the present study, it was investigated whether apatinib has therapeutic potential for glioma. Cell Counting Kit-8 and colony formation assays were utilized to determine the cell viability of p53- and EGFR-mutated U251MG glioma cells, and wild-type U-87MG ATCC glioma cells. Furthermore, apoptosis, and the invasion and migration abilities of glioma cells were investigated by flow cytometry, invasion assays and wound-healing assays. The potential of the combination of apatinib with temozolomide (TMZ) for glioma therapy was also investigated. The results demonstrate that apatinib significantly inhibited cell proliferation and colony formation through promoting cell apoptosis in p53- and EGFR-mutated and wild-type glioma cells. Cell invasion and migration abilities were notably decreased following treatment with apatinib. Overall, the present study indicates a synergistic antitumor effect of apatinib and TMZ in glioma, and presents a basis for the use of apatinib in glioma treatment.
Collapse
Affiliation(s)
- Chao Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Man Jiang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Helei Hou
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Qiang Lin
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhiyong Yan
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaochun Zhang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
30
|
Roberts NB, Alqazzaz A, Hwang JR, Qi X, Keegan AD, Kim AJ, Winkles JA, Woodworth GF. Oxaliplatin disrupts pathological features of glioma cells and associated macrophages independent of apoptosis induction. J Neurooncol 2018; 140:497-507. [PMID: 30132163 DOI: 10.1007/s11060-018-2979-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/06/2018] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Emerging evidence suggests that effective treatment of glioblastoma (GBM), the most common and deadly form of adult primary brain cancer, will likely require concurrent treatment of multiple aspects of tumor pathobiology to overcome tumor heterogeneity and the complex tumor-supporting microenvironment. Recent studies in non-central nervous system (CNS) tumor cells have demonstrated that oxaliplatin (OXA) can induce multi-faceted anti-tumor effects, in particular at drug concentrations below those required to induce apoptosis. These findings motivated re-investigation of OXA for the treatment of GBM. METHODS The effects of OXA on murine KR158 and GL261 glioma cells including cell growth, cell death, inhibition of signal transducer and activator of transcription (STAT) activity, O-6-methylguanine-DNA methyltransferase (MGMT) expression, and immunogenic cell death (ICD) initiation, were evaluated by cytotoxicity assays, Western blot analysis, STAT3-luciferase reporter assays, qRT-PCR assays, and flow cytometry. Chemical inhibitors of endoplasmic reticulum (ER) stress were used to investigate the contribution of this cell damage response to the observed OXA effects. The effect of OXA on bone marrow-derived macrophages (BMDM) exposed to glioma conditioned media (GCM) was also analyzed by Western blot analysis. RESULTS We identified the OXA concentration threshold for induction of apoptosis and from this determined the drug dose and treatment period for sub-cytotoxic treatments of glioma cells. Under these experimental conditions, OXA reduced STAT3 activity, reduced MGMT levels and increased temozolomide sensitivity. In addition, there was evidence of immunogenic cell death (elevated EIF2α phosphorylation and calreticulin exposure) following prolonged OXA treatment. Notably, inhibition of ER stress reversed the OXA-mediated inhibition of STAT3 activity and MGMT expression in the tumor cells. In BMDMs exposed to GCM, OXA also reduced levels of phosphorylated STAT3 and decreased expression of Arginase 1, an enzyme known to contribute to pro-tumor functions in the tumor-immune environment. CONCLUSIONS OXA can induce notable multi-faceted biological effects in glioma cells and BMDMs at relatively low drug concentrations. These findings may have significant therapeutic relevance against GBM and warrant further investigation.
Collapse
Affiliation(s)
- Nathan B Roberts
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Aymen Alqazzaz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Jacqueline R Hwang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Xiulan Qi
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Research and Development Service, U.S. Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Jeffrey A Winkles
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA. .,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Surgery, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD, 21201, USA.
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
31
|
Hoja S, Schulze M, Rehli M, Proescholdt M, Herold-Mende C, Hau P, Riemenschneider MJ. Molecular dissection of the valproic acid effects on glioma cells. Oncotarget 2018; 7:62989-63002. [PMID: 27556305 PMCID: PMC5325342 DOI: 10.18632/oncotarget.11379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/12/2016] [Indexed: 11/25/2022] Open
Abstract
Many glioblastoma patients suffer from seizures why they are treated with antiepileptic agents. Valproic acid (VPA) is a histone deacetylase inhibitor that apart from its anticonvulsive effects in some retrospective studies has been suggested to lead to a superior outcome of glioblastoma patients. However, the exact molecular effects of VPA treatment on glioblastoma cells have not yet been deciphered. We treated glioblastoma cells with VPA, recorded the functional effects of this treatment and performed a global and unbiased next generation sequencing study on the chromatin (ChIP) and RNA level. 1) VPA treatment clearly sensitized glioma cells to temozolomide: A protruding VPA-induced molecular feature in this context was the transcriptional upregulation/reexpression of numerous solute carrier (SLC) transporters that was also reflected by euchromatinization on the histone level and a reexpression of SLC transporters in human biopsy samples after VPA treatment. DNA repair genes were adversely reduced. 2) VPA treatment, however, also reduced cell proliferation in temozolomide-naive cells: On the molecular level in this context we observed a transcriptional upregulation/reexpression and euchromatinization of several glioblastoma relevant tumor suppressor genes and a reduction of stemness markers, while transcriptional subtype classification (mesenchymal/proneural) remained unaltered. Taken together, these findings argue for both temozolomide-dependent and -independent effects of VPA. VPA might increase the uptake of temozolomide and simultaneously lead to a less malignant glioblastoma phenotype. From a mere molecular perspective these findings might indicate a surplus value of VPA in glioblastoma therapy and could therefore contribute an additional ratio for clinical decision making.
Collapse
Affiliation(s)
- Sabine Hoja
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Markus Schulze
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Michael Rehli
- Department of Internal Medicine III, Regensburg University Hospital, Regensburg, Germany.,RCI Regensburg Centre for Interventional Immunology, Regensburg University Hospital, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany.,Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Christel Herold-Mende
- Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Hau
- Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany.,Department of Neurology, Regensburg University, Regensburg, Germany
| | - Markus J Riemenschneider
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany.,Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
32
|
Kinnersley B, Houlston RS, Bondy ML. Genome-Wide Association Studies in Glioma. Cancer Epidemiol Biomarkers Prev 2018; 27:418-428. [PMID: 29382702 DOI: 10.1158/1055-9965.epi-17-1080] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 01/23/2023] Open
Abstract
Since the first reports in 2009, genome-wide association studies (GWAS) have been successful in identifying germline variants associated with glioma susceptibility. In this review, we describe a chronological history of glioma GWAS, culminating in the most recent study comprising 12,496 cases and 18,190 controls. We additionally summarize associations at the 27 glioma-risk SNPs that have been reported so far. Future efforts are likely to be principally focused on assessing association of germline-risk SNPs with particular molecular subgroups of glioma, as well as investigating the functional basis of the risk loci in tumor formation. These ongoing studies will be important to maximize the impact of research into glioma susceptibility, both in terms of insight into tumor etiology as well as opportunities for clinical translation. Cancer Epidemiol Biomarkers Prev; 27(4); 418-28. ©2018 AACRSee all articles in this CEBP Focus section, "Genome-Wide Association Studies in Cancer."
Collapse
Affiliation(s)
- Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Melissa L Bondy
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
33
|
Weatherbee JL, Kraus JL, Ross AH. ER stress in temozolomide-treated glioblastomas interferes with DNA repair and induces apoptosis. Oncotarget 2018; 7:43820-43834. [PMID: 27286262 PMCID: PMC5190062 DOI: 10.18632/oncotarget.9907] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/19/2016] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a deadly grade IV brain tumor. Radiation in combination with temozolomide (TMZ), the current chemotherapeutic for GBMs, only provides 12–14 months survival post diagnosis. Because GBMs are dependent on both activation of the DNA damage pathway and the endoplasmic reticulum (ER) stress response, we asked if a novel ER stress inducing agent, JLK1486, increases the efficacy of TMZ. We found that the combination of TMZ+JLK1486 resulted in decreased proliferation in a panel of adherent GBM cells lines and reduced secondary sphere formation in non-adherent and primary lines. Decreased proliferation correlated with increased cell death due to apoptosis. We found prolonged ER stress in TMZ+JLK1486 treated cells that resulted in sustained activation of the unfolded protein response (UPR) through increased levels of BiP, ATF4, and CHOP. In addition, TMZ+JLK1486 treatment caused decreased RAD51 levels, impairing DNA damage repair. Furthermore, we found delayed time to tumor doubling in TMZ+JLK1486 treated mice. Our data shows that the addition of JLK1486 to TMZ increases the efficaciousness of the treatment by decreasing proliferation and inducing cell death. We propose increased cell death is due to two factors. One, prolonged ER stress driving the expression of the pro-apoptotic transcription factor CHOP, and, second, unresolved DNA double strand breaks, due to decreased RAD51 levels. The combination of TMZ+JLK1486 is a potential novel therapeutic combination and suggests an inverse relationship between unresolved ER stress and the DNA damage response pathway.
Collapse
Affiliation(s)
- Jessica L Weatherbee
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jean-Louis Kraus
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix-Marseille University (AMU) and CNRS, UMR 7288, IBDML, Case 907, Marseille, France
| | - Alonzo H Ross
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
34
|
Melin BS, Barnholtz-Sloan JS, Wrensch MR, Johansen C, Il'yasova D, Kinnersley B, Ostrom QT, Labreche K, Chen Y, Armstrong G, Liu Y, Eckel-Passow JE, Decker PA, Labussière M, Idbaih A, Hoang-Xuan K, Di Stefano AL, Mokhtari K, Delattre JY, Broderick P, Galan P, Gousias K, Schramm J, Schoemaker MJ, Fleming SJ, Herms S, Heilmann S, Nöthen MM, Wichmann HE, Schreiber S, Swerdlow A, Lathrop M, Simon M, Sanson M, Andersson U, Rajaraman P, Chanock S, Linet M, Wang Z, Yeager M, Wiencke JK, Hansen H, McCoy L, Rice T, Kosel ML, Sicotte H, Amos CI, Bernstein JL, Davis F, Lachance D, Lau C, Merrell RT, Shildkraut J, Ali-Osman F, Sadetzki S, Scheurer M, Shete S, Lai RK, Claus EB, Olson SH, Jenkins RB, Houlston RS, Bondy ML. Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors. Nat Genet 2017; 49:789-794. [PMID: 28346443 PMCID: PMC5558246 DOI: 10.1038/ng.3823] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 03/01/2017] [Indexed: 01/07/2023]
Abstract
Genome-wide association studies (GWAS) have transformed our understanding of glioma susceptibility, but individual studies have had limited power to identify risk loci. We performed a meta-analysis of existing GWAS and two new GWAS, which totaled 12,496 cases and 18,190 controls. We identified five new loci for glioblastoma (GBM) at 1p31.3 (rs12752552; P = 2.04 × 10-9, odds ratio (OR) = 1.22), 11q14.1 (rs11233250; P = 9.95 × 10-10, OR = 1.24), 16p13.3 (rs2562152; P = 1.93 × 10-8, OR = 1.21), 16q12.1 (rs10852606; P = 1.29 × 10-11, OR = 1.18) and 22q13.1 (rs2235573; P = 1.76 × 10-10, OR = 1.15), as well as eight loci for non-GBM tumors at 1q32.1 (rs4252707; P = 3.34 × 10-9, OR = 1.19), 1q44 (rs12076373; P = 2.63 × 10-10, OR = 1.23), 2q33.3 (rs7572263; P = 2.18 × 10-10, OR = 1.20), 3p14.1 (rs11706832; P = 7.66 × 10-9, OR = 1.15), 10q24.33 (rs11598018; P = 3.39 × 10-8, OR = 1.14), 11q21 (rs7107785; P = 3.87 × 10-10, OR = 1.16), 14q12 (rs10131032; P = 5.07 × 10-11, OR = 1.33) and 16p13.3 (rs3751667; P = 2.61 × 10-9, OR = 1.18). These data substantiate that genetic susceptibility to GBM and non-GBM tumors are highly distinct, which likely reflects different etiology.
Collapse
Affiliation(s)
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Margaret R Wrensch
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Christoffer Johansen
- Institute of Cancer Epidemiology, Danish Cancer Society, Copenhagen, Denmark and Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Dora Il'yasova
- Department of Epidemiology and Biostatistics, School of Public Health, Georgia State University, Atlanta, Georgia, USA
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Cancer Control and Prevention Program, Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Ben Kinnersley
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Quinn T Ostrom
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Karim Labreche
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
| | - Yanwen Chen
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Georgina Armstrong
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Yanhong Liu
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Jeanette E Eckel-Passow
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Paul A Decker
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Marianne Labussière
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
| | - Ahmed Idbaih
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | - Khe Hoang-Xuan
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | - Anna-Luisa Di Stefano
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | - Karima Mokhtari
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | - Jean-Yves Delattre
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | - Peter Broderick
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Pilar Galan
- Université Paris 13 Sorbonne Paris Cité, INSERM U557, INRA U1125, CNAM, Paris, France
| | | | - Johannes Schramm
- Department of Neurosurgery, University of Bonn Medical Center, Bonn, Germany
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Sarah J Fleming
- Centre for Epidemiology and Biostatistics, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Stefan Herms
- Centre for Epidemiology and Biostatistics, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Heinz-Erich Wichmann
- Helmholtz Center Munich, Institute of Epidemiology I, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig Maximilians University, Munich, Germany
- Institute of Medical Statistics and Epidemiology, Technical University Munich, Munich, Germany
| | - Stefan Schreiber
- 1st Medical Department, University Clinic Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Anthony Swerdlow
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, Institute of Cancer Research, London, UK
| | - Mark Lathrop
- Génome Québec, Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Matthias Simon
- Department of Neurosurgery, University of Bonn Medical Center, Bonn, Germany
| | - Marc Sanson
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | | | - Preetha Rajaraman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Martha Linet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - John K Wiencke
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | - Helen Hansen
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Lucie McCoy
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Terri Rice
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Matthew L Kosel
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Hugues Sicotte
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Jonine L Bernstein
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Faith Davis
- School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Dan Lachance
- Department of Neurology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Ching Lau
- Department of Pediatrics, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Ryan T Merrell
- Department of Neurology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Joellen Shildkraut
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Cancer Control and Prevention Program, Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Francis Ali-Osman
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Siegal Sadetzki
- Cancer and Radiation Epidemiology Unit, Gertner Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michael Scheurer
- Department of Pediatrics, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Sanjay Shete
- Department of Biostatistics, University of Texas Maryland Anderson Cancer Center, Houston, Texas, USA
| | - Rose K Lai
- Departments of Neurology and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Elizabeth B Claus
- School of Public Health, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard S Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Melissa L Bondy
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
35
|
Obacz J, Avril T, Le Reste PJ, Urra H, Quillien V, Hetz C, Chevet E. Endoplasmic reticulum proteostasis in glioblastoma—From molecular mechanisms to therapeutic perspectives. Sci Signal 2017; 10:10/470/eaal2323. [DOI: 10.1126/scisignal.aal2323] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Yuan XS, Wang ZT, Hu YJ, Bao FC, Yuan P, Zhang C, Cao JL, Lv W, Hu J. Downregulation of RUVBL1 inhibits proliferation of lung adenocarcinoma cells by G1/S phase cell cycle arrest via multiple mechanisms. Tumour Biol 2016; 37:16015–16027. [PMID: 27722820 DOI: 10.1007/s13277-016-5452-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/23/2016] [Indexed: 02/08/2023] Open
Abstract
Lung cancer remains a leading cause of cancer-related mortality and morbidity worldwide, of which non-small cell lung cancer (NSCLC) accounts for 80 %. RUVBL1 is a highly conserved eukaryotic AAA+ adenosine 5'-triphosphatase (ATPase) that has many functions highly relevant to cancer. We therefore attempted to determine the potential role of RUVBL1 in the biogenesis of lung adenocarcinoma and obtained some interesting results. Our study revealed that RUVBL1 expression was higher in lung adenocarcinoma specimens than in those of adjacent non-tumor tissues and in lung cancer cell lines than in normal lung cell lines. RUVBL1 knockdown via siRNA reduced proliferation and caused G1/S phase cell cycle arrest in lung adenocarcinoma cell lines. The G1/S phase cell cycle arrest triggered by RUVBL1 downregulation could be attributed, at least in part, to repression of the AKT/GSK-3β/cyclin D1 pathway and probably to the activation of IRE1α-mediated endoplasmic reticulum (ER) stress. We thus demonstrated for the first time that a knockdown of RUVBL1 could effectively inhibit the proliferation of lung adenocarcinoma A549 and H292 cells through the induction of G1/S phase cell cycle arrest via multiple mechanisms. These observations strongly suggested that RUVBL1 should be considered a promising target for the prevention or therapy of lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiao-Shuai Yuan
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Zhi-Tian Wang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Ye-Ji Hu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Fei-Chao Bao
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Ping Yuan
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Chong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Jin-Lin Cao
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Wang Lv
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China
| | - Jian Hu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No.79, Qingchun Road, Hangzhou, China.
| |
Collapse
|
37
|
Brun E, Sicard-Roselli C. Actual questions raised by nanoparticle radiosensitization. Radiat Phys Chem Oxf Engl 1993 2016. [DOI: 10.1016/j.radphyschem.2016.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Ma Z, Fan C, Yang Y, Di S, Hu W, Li T, Zhu Y, Han J, Xin Z, Wu G, Zhao J, Li X, Yan X. Thapsigargin sensitizes human esophageal cancer to TRAIL-induced apoptosis via AMPK activation. Sci Rep 2016; 6:35196. [PMID: 27731378 PMCID: PMC5059685 DOI: 10.1038/srep35196] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent for esophageal squamous cell carcinoma (ESCC). Forced expression of CHOP, one of the key downstream transcription factors during endoplasmic reticulum (ER) stress, upregulates the death receptor 5 (DR5) levels and promotes oxidative stress and cell death. In this study, we show that ER stress mediated by thapsigargin promoted CHOP and DR5 synthesis thus sensitizing TRAIL treatment, which induced ESCC cells apoptosis. These effects were reversed by DR5 siRNA in vitro and CHOP siRNA both in vitro and in vivo. Besides, chemically inhibition of AMPK by Compound C and AMPK siRNA weakened the anti-cancer effect of thapsigargin and TRAIL co-treatment. Therefore, our findings suggest ER stress effectively sensitizes human ESCC to TRAIL-mediated apoptosis via the TRAIL-DR5-AMPK signaling pathway, and that activation of ER stress may be beneficial for improving the efficacy of TRAIL-based anti-cancer therapy.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi’an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi’an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi’an 710032, China
| | - Yifang Zhu
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Zhenlong Xin
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi’an 710032, China
| | - Guiling Wu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi’an 710032, China
| | - Jing Zhao
- Department of Thoracic Surgery, Beijing Military General Hospital, 5 DongSi ShiTiao Road 100070, Beijing 100700, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
| |
Collapse
|