1
|
Zhang Y, Jin T, Dou Z, Wei B, Zhang B, Sun C. The dual role of the CD95 and CD95L signaling pathway in glioblastoma. Front Immunol 2022; 13:1029737. [PMID: 36505426 PMCID: PMC9730406 DOI: 10.3389/fimmu.2022.1029737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Binding of CD95, a cell surface death receptor, to its homologous ligand CD95L, transduces a cascade of downstream signals leading to apoptosis crucial for immune homeostasis and immune surveillance. Although CD95 and CD95L binding classically induces programmed cell death, most tumor cells show resistance to CD95L-induced apoptosis. In some cancers, such as glioblastoma, CD95-CD95L binding can exhibit paradoxical functions that promote tumor growth by inducing inflammation, regulating immune cell homeostasis, and/or promoting cell survival, proliferation, migration, and maintenance of the stemness of cancer cells. In this review, potential mechanisms such as the expression of apoptotic inhibitor proteins, decreased activity of downstream elements, production of nonapoptotic soluble CD95L, and non-apoptotic signals that replace apoptotic signals in cancer cells are summarized. CD95L is also expressed by other types of cells, such as endothelial cells, polymorphonuclear myeloid-derived suppressor cells, cancer-associated fibroblasts, and tumor-associated microglia, and macrophages, which are educated by the tumor microenvironment and can induce apoptosis of tumor-infiltrating lymphocytes, which recognize and kill cancer cells. The dual role of the CD95-CD95L system makes targeted therapy strategies against CD95 or CD95L in glioblastoma difficult and controversial. In this review, we also discuss the current status and perspective of clinical trials on glioblastoma based on the CD95-CD95L signaling pathway.
Collapse
Affiliation(s)
- Yanrui Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Taian Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhangqi Dou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Boxing Wei
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Buyi Zhang
- Department of Pathology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China,*Correspondence: Buyi Zhang, ; Chongran Sun,
| | - Chongran Sun
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China,Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, Zhejiang, China,*Correspondence: Buyi Zhang, ; Chongran Sun,
| |
Collapse
|
2
|
Hsia TC, Peng SF, Chueh FS, Lu KW, Yang JL, Huang AC, Hsu FT, Wu RSC. Bisdemethoxycurcumin Induces Cell Apoptosis and Inhibits Human Brain Glioblastoma GBM 8401/ Luc2 Cell Xenograft Tumor in Subcutaneous Nude Mice In Vivo. Int J Mol Sci 2022; 23:ijms23010538. [PMID: 35008959 PMCID: PMC8745075 DOI: 10.3390/ijms23010538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Bisdemethoxycurcumin (BDMC) has biological activities, including anticancer effects in vitro; however, its anticancer effects in human glioblastoma (GBM) cells have not been examined yet. This study aimed to evaluate the tumor inhibitory effect and molecular mechanism of BDMC on human GBM 8401/luc2 cells in vitro and in vivo. In vitro studies have shown that BDMC significantly reduced cell viability and induced cell apoptosis in GBM 8401/luc2 cells. Furthermore, BDMC induced apoptosis via inhibited Bcl-2 (anti-apoptotic protein) and increased Bax (pro-apoptotic proteins) and cytochrome c release in GBM 8401/luc2 cells in vitro. Then, twelve BALB/c-nude mice were xenografted with human glioblastoma GBM 8401/luc2 cancer cells subcutaneously, and the xenograft nude mice were treated without and with BDMC (30 and 60 mg/kg of BDMC treatment) every 3 days. GBM 8401/luc2 cell xenografts experiment showed that the growth of the tumors was significantly suppressed by BDMC administration at both doses based on the reduction of tumor size and weights. BDMC did not change the body weight and the H&E histopathology analysis of liver samples, indicating that BDMC did not induce systemic toxicity. Meanwhile, treatment with BDMC up-regulated the expressions of BAX and cleaved caspase-3, while it down-regulated the protein expressions of Bcl-2 and XIAP in the tumor tissues compared with the control group. This study has demonstrated that BDMC presents potent anticancer activity on the human glioblastoma GBM 8401/luc2 cell xenograft model by inducing apoptosis and inhibiting tumor cell proliferation and shows the potential for further development to the anti-GBM cancer drug.
Collapse
Affiliation(s)
- Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung 406, Taiwan;
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan;
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan;
| | - Kung-Wen Lu
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 406, Taiwan;
| | - Jiun-Long Yang
- Department of Nursing, St. Mary’s Junior College of Medicine, Nursing and Management, Yilan 266, Taiwan; (J.-L.Y.); (A.-C.H.)
| | - An-Cheng Huang
- Department of Nursing, St. Mary’s Junior College of Medicine, Nursing and Management, Yilan 266, Taiwan; (J.-L.Y.); (A.-C.H.)
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan
- Correspondence: (F.-T.H.); (R.S.-C.W.); Tel.: +886-4-2205-3366 (ext. 2532) (F.-T.H.); +886-4-2205-2121 (ext. 5242) (R.S.-C.W.); Fax: +886-4-2205-3764 (F.-T.H.); +886-4-2205-2121 (ext. 5237) (R.S.-C.W.)
| | - Rick Sai-Chuen Wu
- Department of Anesthesiology, China Medical University Hospital, Taichung 404, Taiwan
- Department of Anesthesiology, China Medical University, Taichung 404, Taiwan
- Correspondence: (F.-T.H.); (R.S.-C.W.); Tel.: +886-4-2205-3366 (ext. 2532) (F.-T.H.); +886-4-2205-2121 (ext. 5242) (R.S.-C.W.); Fax: +886-4-2205-3764 (F.-T.H.); +886-4-2205-2121 (ext. 5237) (R.S.-C.W.)
| |
Collapse
|
3
|
Nasrollahzadeh A, Momeny M, Bashash D, Yousefi H, Mousavi SA, Ghaffari SH. Blockade of Nuclear Factor-Κb (NF-Κb) Pathway Using Bay 11-7082 Enhances Arsenic Trioxide-Induced Antiproliferative Activity in U87 Glioblastoma Cells. Rep Biochem Mol Biol 2022; 10:602-613. [PMID: 35291620 PMCID: PMC8903363 DOI: 10.52547/rbmb.10.4.602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Glioblastoma (GBM), the most aggressive and common form of glioma, accounts for over 13,000 death per year in the United States which indicates the importance of developing novel strategies for the treatment of this fatal malignancy. Although Arsenic trioxide (ATO) hinders the growth and survival of GBM cells, the requirement of concentrations higher than 4 µM for triggering apoptotic cell death has questioned its safety profile. Since the NF-κB signaling pathway plays a crucial role in tumorigenesis and chemo-resistance, targeting this oncogenic pathway may sensitize GBM cells to lower concentrations of ATO. METHODS Anti-tumor effects of ATO as monotherapy and in combination with Bay 11-7082 were determined using MTT, crystal violet staining, Annexin V/PI staining and scratch assays. Quantitative reverse transcription-PCR (qRT-PCR) analysis was applied to elucidate the molecular mechanisms underlying the anti-tumor activity of this combination therapy. RESULTS Our results revealed that ATO and Bay 11-7082 synergistically inhibited the proliferation and survival of GBM cells. Also, it was revealed that NF-κB inhibition using Bay 11-7082 enhanced the inhibitory effects of ATO on migration of GBM cells via suppressing the expression of NF-κB target genes such as TWIST, MMP2, ICAM-1, and cathepsin B. Furthermore, combination treatment of GBM cells with ATO and Bay 11-7082 significantly induce apoptotic cell death coupled with downregulation of NF-κB anti-apoptotic target genes including Bcl-2 and IAP family members. CONCLUSION Altogether, these findings suggest that combination therapy with ATO and Bay 11-7082 may be a promising strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Ali Nasrollahzadeh
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | - Seyed Asadollah Mousavi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hamidollah Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Shanmugam MK, Sethi G. Molecular mechanisms of cell death. MECHANISMS OF CELL DEATH AND OPPORTUNITIES FOR THERAPEUTIC DEVELOPMENT 2022:65-92. [DOI: 10.1016/b978-0-12-814208-0.00002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
5
|
Devi Daimary U, Girisa S, Parama D, Verma E, Kumar A, Kunnumakkara AB. Embelin: A novel XIAP inhibitor for the prevention and treatment of chronic diseases. J Biochem Mol Toxicol 2021; 36:e22950. [PMID: 34842329 DOI: 10.1002/jbt.22950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
Chronic diseases are a serious health concern worldwide, especially in the elderly population. Most chronic diseases like cancer, cardiovascular ailments, neurodegenerative disorders, and autoimmune diseases are caused due to the abnormal functioning of multiple signaling pathways that give rise to critical anomalies in the body. Although a lot of advanced therapies are available, these have failed to entirely cure the disease due to their less efficacy. Apart from this, they have been shown to manifest disturbing side effects which hamper the patient's quality of life to the extreme. Since the last few decades, extensive studies have been done on natural herbs due to their excellent medicinal benefits. Components present in natural herbs target multiple signaling pathways involved in diseases and therefore hold high potential in the prevention and treatment of various chronic diseases. Embelin, a benzoquinone, is one such agent isolated from Embelia ribes, which has shown excellent biological activities toward several chronic ailments by upregulating a number of antioxidant enzymes (e.g., SOD, CAT, GSH, etc.), inhibiting anti-apoptotic genes (e.g., TRAIL, XIAP, survivin, etc.), modulating transcription factors (e.g., NF-κB, STAT3, etc.) blocking inflammatory biomarkers (e.g., NO, IL-1β, IL-6, TNF-α, etc.), monitoring cell cycle synchronizing genes (e.g., p53, cyclins, CDKs, etc.), and so forth. Several preclinical studies have confirmed its excellent therapeutic activities against malicious diseases like cancer, obesity, heart diseases, Alzheimer's, and so forth. This review presents an overview of embelin, its therapeutic prospective, and the molecular targets in different chronic diseases.
Collapse
Affiliation(s)
- Uzini Devi Daimary
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Dey Parama
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Elika Verma
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| |
Collapse
|
6
|
Liao CL, Ma YS, Hsia TC, Chou YC, Lien JC, Peng SF, Kuo CL, Hsu FT. Tetrandrine Suppresses Human Brain Glioblastoma GBM 8401/ luc2 Cell-Xenografted Subcutaneous Tumors in Nude Mice In Vivo. Molecules 2021; 26:molecules26237105. [PMID: 34885686 PMCID: PMC8659155 DOI: 10.3390/molecules26237105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/04/2023] Open
Abstract
Tetrandrine (TET), a bisbenzylisoquinoline (BBI) alkaloid, is isolated from the plant Stephania tetrandra S. Moore and has a wide range of biological activity, including anticancer properties in vitro and in vivo. At first, we established a luciferase-expressing stable clone that was named GBM 8401/luc2 cells. Herein, the primary results indicated that TET reduced the total cell viability and induced cell apoptosis in GBM 8401/luc2 human glioblastoma cells. However, there is no available information showing that TET suppresses glioblastoma cells in vivo. Thus, we investigated the effects and mechanisms of TET on a GBM 8401/luc2 cell-generated tumor in vivo. After the tumor volume reached 100-120 mm3 in subcutaneously xenografted nude mice, all of the mice were randomly divided into three groups: Group I was treated with phosphate-buffered solution (PBS) containing 0.1% dimethyl sulfoxide, Group II with 25 mg/kg of TET, and Group III with 50 mg/kg of TET. All mice were given the oral treatment of PBS or TET by gavage for 21 days, and the body weight and tumor volumes were recorded every 5 days. After treatment, individual tumors, kidneys, livers, and spleens were isolated from each group. The results showed that TET did not affect the body weights, but it significantly decreased the tumor volumes. The TET treatment at 50 mg/kg had a two-fold decrease in tumor volumes than that at 25 mg/kg when compared to the control. TET decreased the total photon flux, and treatment with TET at 50 mg/kg had a lower total photon flux than that at 25 mg/kg, as measured by a Xenogen IVIS imaging system. Moreover, the higher TET treatment had lower tumor volumes and weights than those of the lower dose. The apoptosis-associated protein expression in the tumor section was examined by immunohistochemical analysis, and the results showed that TET treatment reduced the levels of c-FLIP, MCL-1, and XIAP but increased the signals of cleaved-caspase-3, -8, and -9. Furthermore, the hematoxylin and eosin (H & E) staining of kidney, liver, and spleen tissues showed no significant difference between the TET-treated and control groups. Overall, these observations demonstrated that TET suppressed subcutaneous tumor growth in a nude-mice model via the induction of cell apoptosis.
Collapse
Affiliation(s)
- Ching-Lung Liao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40604, Taiwan;
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 840, Taiwan;
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung 824, Taiwan
| | - Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung 404, Taiwan;
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Cheng Chou
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung 404, Taiwan;
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan;
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan
- Correspondence: (C.-L.K.); (F.-T.H.); Tel.: +886-4-2205-3366 (ext. 5202) (C.-L.K.); +886-4-2205-3366 (ext. 2531) (F.-T.H.); Fax: +886-4-2205-3764 (C.-L.K. & F.-T.H.)
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
- Correspondence: (C.-L.K.); (F.-T.H.); Tel.: +886-4-2205-3366 (ext. 5202) (C.-L.K.); +886-4-2205-3366 (ext. 2531) (F.-T.H.); Fax: +886-4-2205-3764 (C.-L.K. & F.-T.H.)
| |
Collapse
|
7
|
Townsend PA, Kozhevnikova MV, Cexus ONF, Zamyatnin AA, Soond SM. BH3-mimetics: recent developments in cancer therapy. J Exp Clin Cancer Res 2021; 40:355. [PMID: 34753495 PMCID: PMC8576916 DOI: 10.1186/s13046-021-02157-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
The hopeful outcomes from 30 years of research in BH3-mimetics have indeed served a number of solid paradigms for targeting intermediates from the apoptosis pathway in a variety of diseased states. Not only have such rational approaches in drug design yielded several key therapeutics, such outputs have also offered insights into the integrated mechanistic aspects of basic and clinical research at the genetics level for the future. In no other area of medical research have the effects of such work been felt, than in cancer research, through targeting the BAX-Bcl-2 protein-protein interactions. With these promising outputs in mind, several mimetics, and their potential therapeutic applications, have also been developed for several other pathological conditions, such as cardiovascular disease and tissue fibrosis, thus highlighting the universal importance of the intrinsic arm of the apoptosis pathway and its input to general tissue homeostasis. Considering such recent developments, and in a field that has generated so much scientific interest, we take stock of how the broadening area of BH3-mimetics has developed and diversified, with a focus on their uses in single and combined cancer treatment regimens and recently explored therapeutic delivery methods that may aid the development of future therapeutics of this nature.
Collapse
Affiliation(s)
- Paul A Townsend
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
- University of Manchester, Manchester, UK.
| | - Maria V Kozhevnikova
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
- Sirius University of Science and Technology, Sochi, Russian Federation
| | - Surinder M Soond
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
8
|
Momeny M, Shamsaiegahkani S, Kashani B, Hamzehlou S, Esmaeili F, Yousefi H, Irani S, Mousavi SA, Ghaffari SH. Cediranib, a pan-inhibitor of vascular endothelial growth factor receptors, inhibits proliferation and enhances therapeutic sensitivity in glioblastoma cells. Life Sci 2021; 287:120100. [PMID: 34715143 DOI: 10.1016/j.lfs.2021.120100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/26/2022]
Abstract
AIMS Glioblastoma (GB) is the most aggressive type of brain tumor. Rapid progression, active angiogenesis, and therapy resistance are major reasons for its high mortality. Elevated expression of members of the vascular endothelial growth factor (VEGF) family suggests that anti-VEGF therapies may be potent anti-glioma therapeutic approaches. Here, we evaluated the anti-tumor activity of cediranib, a pan inhibitor of the VEGF receptors, on GB cells. MATERIALS AND METHODS Anti-proliferative effects of cediranib were determined using MTT, crystal-violet staining, clonogenic and anoikis resistance assays. Apoptosis induction was assessed by Annexin V/PI staining and Western blot analysis and aggressive abilities of GB cells were investigated using cell migration/invasion assays and zymography. Small-interfering RNA (siRNA)-mediated Knockdown was used to study resistance mechanisms. The anti-proliferative and apoptotic effects of cediranib in combination with radiotherapy, temozolomide, bevacizumab were also evaluated using MTT, Annexin V/PI staining and Western blot analysis for cleaved PARP-1. KEY FINDINGS Cediranib reduced GB cell proliferation, induced apoptotic cell death and inhibited the aggressive abilities of GB cells. Cediranib synergistically increased the anti-proliferative and apoptotic effects of radiotherapy and bevacizumab and augmented the sensitivity of GB cells to temozolomide chemotherapy. In addition, knockdown of MET and AKT potentiated cediranib sensitivity in cediranib-resistant GB cells. SIGNIFICANCE These findings suggest that cediranib, alone or in combination with other therapeutics, is a promising strategy for the treatment of GB and provide a rationale for further investigation of the therapeutic potential of cediranib for the treatment of this fatal malignancy.
Collapse
Affiliation(s)
| | - Sahar Shamsaiegahkani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kashani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Esmaeili
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Centre, New Orleans, USA
| | - Shiva Irani
- Department of Biology Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed A Mousavi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Burster T, Traut R, Yermekkyzy Z, Mayer K, Westhoff MA, Bischof J, Knippschild U. Critical View of Novel Treatment Strategies for Glioblastoma: Failure and Success of Resistance Mechanisms by Glioblastoma Cells. Front Cell Dev Biol 2021; 9:695325. [PMID: 34485282 PMCID: PMC8415230 DOI: 10.3389/fcell.2021.695325] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
According to the invasive nature of glioblastoma, which is the most common form of malignant brain tumor, the standard care by surgery, chemo- and radiotherapy is particularly challenging. The presence of glioblastoma stem cells (GSCs) and the surrounding tumor microenvironment protects glioblastoma from recognition by the immune system. Conventional therapy concepts have failed to completely remove glioblastoma cells, which is one major drawback in clinical management of the disease. The use of small molecule inhibitors, immunomodulators, immunotherapy, including peptide and mRNA vaccines, and virotherapy came into focus for the treatment of glioblastoma. Although novel strategies underline the benefit for anti-tumor effectiveness, serious challenges need to be overcome to successfully manage tumorigenesis, indicating the significance of developing new strategies. Therefore, we provide insights into the application of different medications in combination to boost the host immune system to interfere with immune evasion of glioblastoma cells which are promising prerequisites for therapeutic approaches to treat glioblastoma patients.
Collapse
Affiliation(s)
- Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Rebecca Traut
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Zhanerke Yermekkyzy
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Katja Mayer
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
10
|
Gaggianesi M, Di Franco S, Pantina VD, Porcelli G, D'Accardo C, Verona F, Veschi V, Colarossi L, Faldetta N, Pistone G, Bongiorno MR, Todaro M, Stassi G. Messing Up the Cancer Stem Cell Chemoresistance Mechanisms Supported by Tumor Microenvironment. Front Oncol 2021; 11:702642. [PMID: 34354950 PMCID: PMC8330815 DOI: 10.3389/fonc.2021.702642] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in cancer patient management and in the development of targeted therapies, systemic chemotherapy is currently used as a first-line treatment for many cancer types. After an initial partial response, patients become refractory to standard therapy fostering rapid tumor progression. Compelling evidence highlights that the resistance to chemotherapeutic regimens is a peculiarity of a subpopulation of cancer cells within tumor mass, known as cancer stem cells (CSCs). This cellular compartment is endowed with tumor-initiating and metastasis formation capabilities. CSC chemoresistance is sustained by a plethora of grow factors and cytokines released by neighboring tumor microenvironment (TME), which is mainly composed by adipocytes, cancer-associated fibroblasts (CAFs), immune and endothelial cells. TME strengthens CSC refractoriness to standard and targeted therapies by enhancing survival signaling pathways, DNA repair machinery, expression of drug efflux transporters and anti-apoptotic proteins. In the last years many efforts have been made to understand CSC-TME crosstalk and develop therapeutic strategy halting this interplay. Here, we report the combinatorial approaches, which perturb the interaction network between CSCs and the different component of TME.
Collapse
Affiliation(s)
- Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Caterina D'Accardo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | | | - Naida Faldetta
- Department of Surgery, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
11
|
Chu X, Zheng W, Chen Q, Wang C, Fan S, Shao C. HBXIP contributes to radioresistance through NF-κB-mediated expression of XIAP in breast cancer. RADIATION MEDICINE AND PROTECTION 2021. [DOI: 10.1016/j.radmp.2021.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
12
|
Huang YP, Ma YS, Kuo CL, Liao CL, Chen PY, Peng SF, Hsu FT, Lai KC. Demethoxycurcumin Suppresses Human Brain Glioblastoma Multiforme GBM 8401 Cell Xenograft Tumor in Nude Mice In Vivo. Int J Mol Sci 2021; 22:ijms22115503. [PMID: 34071132 PMCID: PMC8197162 DOI: 10.3390/ijms22115503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022] Open
Abstract
Demethoxycurcumin (DMC), a derivate of curcumin, has been shown to induce apoptotic cell death in human glioblastoma multiforme GBM 8401 cells via cell cycle arrest and induction of cell apoptosis. However, there is no report showing DMC suppresses glioblastoma multiforme cells in vivo. In the present study, we investigated the effects of DMC on GBM8401 cells in vivo. At first, we established a luciferase-expressing stable clone named GBM 8401/luc2. Second, mice were inoculated subcutaneously with GBM 8401/luc2 cells to generate a xenograft tumor mice model. After inoculation, tumor volume reached 100-120 mm3, and all mice were randomly divided into three groups: Group I was treated with 110 µL phosphate-buffered solution (PBS) containing 0.1% dimethyl sulfoxide, Group II with 30 mg/kg of DMC, and Group III with 60 mg/kg of DMC. Mice from each group were given the oral treatment of DMC by gavage for 21 days. The body weight and tumor volume were recorded every 3 days. DMC significantly decreased the tumor volumes, and 60 mg/kg treatment showed a higher decrease in tumor volumes than that of 30 mg/kg, However, DMC did not affect the body weights. The photons emitted from mice tumors were detected with Xenogen IVIS imaging system, DMC at both doses decreased the total photon flux and 60 mg/kg treatment of DMC has low total photon flux than that of 30 mg/kg. The tumor volumes and weights in 60 mg/kg treatment of DMC were lower than that of 30 mg/kg. Immunohistochemical analysis was used to measure protein expression of tumors and results showed that DMC treatment led to lightly staining with anti-Bcl-2 and -XIAP and 60 mg/kg treatment of DMC has lighter staining with anti-Bcl-2 and -XIAP than that of 30 mg/kg. The higher dose (60 mg/kg) of DMC has higher signals of cleaved-caspase-3 than that of the lower dose (30 mg/kg). Furthermore, the hematoxylin and eosin (H&E) staining of liver tissues showed no significant difference between DMC-treated and control-groups. Overall, these observations showed that DMC suppressed tumor properties in vivo and DMC may be used against human glioblastoma multiforme in the future.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, School of Medicine, China Medical University, Taichung 406, Taiwan;
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 840, Taiwan;
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung 824, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 406, Taiwan;
| | - Ching-Lung Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung 406, Taiwan;
| | - Po-Yuan Chen
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan; (P.-Y.C.); (S.-F.P.)
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan; (P.-Y.C.); (S.-F.P.)
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan; (P.-Y.C.); (S.-F.P.)
- Correspondence: (F.-T.H.); (K.-C.L.); Tel.: +886-4-22053366 (ext. 2532) (F.-T.H.); +886-5-7837901 (ext. 1161) (K.-C.L.); Fax: +886-4-22053764 (F.-T.H.); +886-5-7831121 (K.-C.L.)
| | - Kuang-Chi Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medical Technology, Chung Hwa University of Medical Technology, Tainan 717, Taiwan
- Department of Surgery, China Medical University Beigang Hospital, Beigang, Yunlin 651, Taiwan
- Correspondence: (F.-T.H.); (K.-C.L.); Tel.: +886-4-22053366 (ext. 2532) (F.-T.H.); +886-5-7837901 (ext. 1161) (K.-C.L.); Fax: +886-4-22053764 (F.-T.H.); +886-5-7831121 (K.-C.L.)
| |
Collapse
|
13
|
Castelli V, Giordano A, Benedetti E, Giansanti F, Quintiliani M, Cimini A, d’Angelo M. The Great Escape: The Power of Cancer Stem Cells to Evade Programmed Cell Death. Cancers (Basel) 2021; 13:328. [PMID: 33477367 PMCID: PMC7830655 DOI: 10.3390/cancers13020328] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the primary causes of death worldwide. Tumour malignancy is related to tumor heterogeneity, which has been suggested to be due to a small subpopulation of tumor cells named cancer stem cells (CSCs). CSCs exert a key role in metastasis development, tumor recurrence, and also epithelial-mesenchymal transition, apoptotic resistance, self-renewal, tumorigenesis, differentiation, and drug resistance. Several current therapies fail to eradicate tumors due to the ability of CSCs to escape different programmed cell deaths. Thus, developing CSC-selective and programmed death-inducing therapeutic approaches appears to be of primary importance. In this review, we discuss the main programmed cell death occurring in cancer and the promising CSC-targeting agents developed in recent years. Even if the reported studies are encouraging, further investigations are necessary to establish a combination of agents able to eradicate CSCs or inhibit their growth and proliferation.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
| | - Antonio Giordano
- Department of Medical Biotechnology, University of Siena, 53100 Siena, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
| | - Francesco Giansanti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
| | - Massimiliano Quintiliani
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (E.B.); (F.G.); (M.Q.)
| |
Collapse
|
14
|
The role of E3 ubiquitin ligases in the development and progression of glioblastoma. Cell Death Differ 2021; 28:522-537. [PMID: 33432111 PMCID: PMC7862665 DOI: 10.1038/s41418-020-00696-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
Despite recent advances in our understanding of the disease, glioblastoma (GB) continues to have limited treatment options and carries a dismal prognosis for patients. Efforts to stratify this heterogeneous malignancy using molecular classifiers identified frequent alterations in targetable proteins belonging to several pathways including the receptor tyrosine kinase (RTK) and mitogen-activated protein kinase (MAPK) signalling pathways. However, these findings have failed to improve clinical outcomes for patients. In almost all cases, GB becomes refractory to standard-of-care therapy, and recent evidence suggests that disease recurrence may be associated with a subpopulation of cells known as glioma stem cells (GSCs). Therefore, there remains a significant unmet need for novel therapeutic strategies. E3 ubiquitin ligases are a family of >700 proteins that conjugate ubiquitin to target proteins, resulting in an array of cellular responses, including DNA repair, pro-survival signalling and protein degradation. Ubiquitin modifications on target proteins are diverse, ranging from mono-ubiquitination through to the formation of polyubiquitin chains and mixed chains. The specificity in substrate tagging and chain elongation is dictated by E3 ubiquitin ligases, which have essential regulatory roles in multiple aspects of brain cancer pathogenesis. In this review, we begin by briefly summarising the histological and molecular classification of GB. We comprehensively describe the roles of E3 ubiquitin ligases in RTK and MAPK, as well as other, commonly altered, oncogenic and tumour suppressive signalling pathways in GB. We also describe the role of E3 ligases in maintaining glioma stem cell populations and their function in promoting resistance to ionizing radiation (IR) and chemotherapy. Finally, we consider how our knowledge of E3 ligase biology may be used for future therapeutic interventions in GB, including the use of blood-brain barrier permeable proteolysis targeting chimeras (PROTACs).
Collapse
|
15
|
Uzunparmak B, Gao M, Lindemann A, Erikson K, Wang L, Lin E, Frank SJ, Gleber-Netto FO, Zhao M, Skinner HD, Newton J, Sikora AG, Myers JN, Pickering CR. Caspase-8 loss radiosensitizes head and neck squamous cell carcinoma to SMAC mimetic-induced necroptosis. JCI Insight 2020; 5:139837. [PMID: 33108350 PMCID: PMC7714407 DOI: 10.1172/jci.insight.139837] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022] Open
Abstract
Caspase-8 (CASP8) is one of the most frequently mutated genes in head and neck squamous carcinomas (HNSCCs), and CASP8 mutations are associated with poor survival. The distribution of these mutations in HNSCCs suggests that they are likely to be inactivating. Inhibition of CASP8 has been reported to sensitize cancer cells to necroptosis, a regulated cell death mechanism. Here, we show that knockdown of CASP8 renders HNSCCs susceptible to necroptosis by a second mitochondria-derived activator of caspase (SMAC) mimetic, birinapant, in combination with pan-caspase inhibitors Z-VAD-FMK or emricasan and radiation. In a syngeneic mouse model of oral cancer, birinapant, particularly when combined with radiation, delayed tumor growth and enhanced survival under CASP8 loss. Exploration of molecular underpinnings of necroptosis sensitivity confirmed that the level of functional receptor-interacting serine/threonine protein kinase 3 (RIP3) determines susceptibility to this mode of death. Although an in vitro screen revealed that low RIP3 levels rendered many HNSCC cell lines resistant to necroptosis, patient tumors maintained RIP3 expression and should therefore remain sensitive. Collectively, these results suggest that targeting the necroptosis pathway with SMAC mimetics, especially in combination with radiation, may be relevant therapeutically in HNSCC with compromised CASP8 status, provided that RIP3 function is maintained.
Collapse
Affiliation(s)
- Burak Uzunparmak
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas USA
| | - Meng Gao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Antje Lindemann
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly Erikson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Li Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eric Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven J. Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederico O. Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Heath D. Skinner
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jared Newton
- Bobby R. Alford Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew G. Sikora
- Bobby R. Alford Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Jeffrey N. Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Curtis R. Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Hsu FT, Chen WT, Wu CT, Chung JG. Hyperforin induces apoptosis through extrinsic/intrinsic pathways and inhibits EGFR/ERK/NF-κB-mediated anti-apoptotic potential in glioblastoma. ENVIRONMENTAL TOXICOLOGY 2020; 35:1058-1069. [PMID: 32485087 DOI: 10.1002/tox.22942] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Glioblastoma is the most common primary brain tumor with poor survival rate and without effective treatment strategy. Notably, amplification and active mutation of epidermal growth factor receptor (EGFR) occur frequently in glioblastoma patient that may be a potential treatment target. Several studies indicated that various type of herbal compounds not only regulate anti-depressant effect but also shown capacity to suppress glioblastoma growth via inducing apoptosis and inhibiting oncogene signaling transduction. Hyperforin, an herb compound derived from St. John's wort was used to treat depressive disorder by inhibiting neuronal reuptake of several neurotransmitters. Although hyperforin can reduce matrix metallopeptidases-2 (MMPs) and -9-mediated metastasis of glioblastoma, the detail mechanism of hyperforin on glioblastoma is remaining unclear. Here, we suggested that hyperforin may induce extrinsic/intrinsic apoptosis and suppress anti-apoptotic related proteins expression of glioblastoma. We also indicated that hyperforin-mediated anti-apoptotic potential of glioblastoma was correlated to inactivation of EGFR/extracellular signal-regulated kinases (ERK)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Wei-Ting Chen
- Department of Psychiatry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Ching-Te Wu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
17
|
Xia Q, Ali S, Liu L, Li Y, Liu X, Zhang L, Dong L. Role of Ubiquitination in PTEN Cellular Homeostasis and Its Implications in GB Drug Resistance. Front Oncol 2020; 10:1569. [PMID: 32984016 PMCID: PMC7492558 DOI: 10.3389/fonc.2020.01569] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) is the most common and aggressive brain malignancy, characterized by heterogeneity and drug resistance. PTEN, a crucial tumor suppressor, exhibits phosphatase-dependent (PI3K-AKT-mTOR pathway)/independent (nucleus stability) activities to maintain the homeostatic regulation of numerous physiological processes. Premature and absolute loss of PTEN activity usually tends to cellular senescence. However, monoallelic loss of PTEN is frequently observed at tumor inception, and absolute loss of PTEN activity also occurs at the late stage of gliomagenesis. Consequently, aberrant PTEN homeostasis, mainly regulated at the post-translational level, renders cells susceptible to tumorigenesis and drug resistance. Ubiquitination-mediated degradation or deregulated intracellular localization of PTEN hijacks cell growth rheostat control for neoplastic remodeling. Functional inactivation of PTEN mediated by the overexpression of ubiquitin ligases (E3s) renders GB cells adaptive to PTEN loss, which confers resistance to EGFR tyrosine kinase inhibitors and immunotherapies. In this review, we discuss how glioma cells develop oncogenic addiction to the E3s-PTEN axis, promoting their growth and proliferation. Antitumor strategies involving PTEN-targeting E3 ligase inhibitors can restore the tumor-suppressive environment. E3 inhibitors collectively reactivate PTEN and may represent next-generation treatment against deadly malignancies such as GB.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Sakhawat Ali
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Xuefeng Liu
- School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Lei Dong
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
18
|
Choudhary S, Burns SC, Mirsafian H, Li W, Vo DT, Qiao M, Lei X, Smith AD, Penalva LO. Genomic analyses of early responses to radiation inglioblastoma reveal new alterations at transcription,splicing, and translation levels. Sci Rep 2020; 10:8979. [PMID: 32488114 PMCID: PMC7265345 DOI: 10.1038/s41598-020-65638-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
High-dose radiation is the main component of glioblastoma therapy. Unfortunately, radio-resistance is a common problem and a major contributor to tumor relapse. Understanding the molecular mechanisms driving response to radiation is critical for identifying regulatory routes that could be targeted to improve treatment response. We conducted an integrated analysis in the U251 and U343 glioblastoma cell lines to map early alterations in the expression of genes at three levels: transcription, splicing, and translation in response to ionizing radiation. Changes at the transcriptional level were the most prevalent response. Downregulated genes are strongly associated with cell cycle and DNA replication and linked to a coordinated module of expression. Alterations in this group are likely driven by decreased expression of the transcription factor FOXM1 and members of the E2F family. Genes involved in RNA regulatory mechanisms were affected at the mRNA, splicing, and translation levels, highlighting their importance in radiation-response. We identified a number of oncogenic factors, with an increased expression upon radiation exposure, including BCL6, RRM2B, IDO1, FTH1, APIP, and LRIG2 and lncRNAs NEAT1 and FTX. Several of these targets have been previously implicated in radio-resistance. Therefore, antagonizing their effects post-radiation could increase therapeutic efficacy. Our integrated analysis provides a comprehensive view of early response to radiation in glioblastoma. We identify new biological processes involved in altered expression of various oncogenic factors and suggest new target options to increase radiation sensitivity and prevent relapse.
Collapse
Affiliation(s)
- Saket Choudhary
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Suzanne C Burns
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Hoda Mirsafian
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Wenzheng Li
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Dat T Vo
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Texas, USA
| | - Mei Qiao
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Xiufen Lei
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Andrew D Smith
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Luiz O Penalva
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA.
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, Texas, USA.
| |
Collapse
|
19
|
Matteoni S, Abbruzzese C, Matarrese P, De Luca G, Mileo AM, Miccadei S, Schenone S, Musumeci F, Haas TL, Sette G, Carapella CM, Amato R, Perrotti N, Signore M, Paggi MG. The kinase inhibitor SI113 induces autophagy and synergizes with quinacrine in hindering the growth of human glioblastoma multiforme cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:202. [PMID: 31101126 PMCID: PMC6525441 DOI: 10.1186/s13046-019-1212-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022]
Abstract
Background Glioblastoma multiforme (GBM), due to its location, aggressiveness, heterogeneity and infiltrative growth, is characterized by an exceptionally dismal clinical outcome. The small molecule SI113, recently identified as a SGK1 inhibitor, has proven to be effective in restraining GBM growth in vitro and in vivo, showing also encouraging results when employed in combination with other antineoplastic drugs or radiotherapy. Our aim was to explore the pharmacological features of SI113 in GBM cells in order to elucidate the pivotal molecular pathways affected by the drug. Such knowledge would be of invaluable help in conceiving a rational offensive toward GBM. Methods We employed GBM cell lines, either established or primary (neurospheres), and used a Reverse-Phase Protein Arrays (RPPA) platform to assess the effect of SI113 upon 114 protein factors whose post-translational modifications are associated with activation or repression of specific signal transduction cascades. Results SI113 strongly affected the PI3K/mTOR pathway, evoking a pro-survival autophagic response in neurospheres. These results suggested the use of SI113 coupled, for maximum efficiency, with autophagy inhibitors. Indeed, the association of SI113 with an autophagy inhibitor, the antimalarial drug quinacrine, induced a strong synergistic effect in inhibiting GBM growth properties in all the cells tested, including neurospheres. Conclusions RPPA clearly identified the molecular pathways influenced by SI113 in GBM cells, highlighting their vulnerability when the drug was administered in association with autophagy inhibitors, providing a strong molecular rationale for testing SI113 in clinical trials in associative GBM therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1212-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvia Matteoni
- Section of Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Claudia Abbruzzese
- Section of Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriele De Luca
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna M Mileo
- Tumor Immunology and Immunotherapy, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Stefania Miccadei
- Tumor Immunology and Immunotherapy, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | | | | | - Tobias L Haas
- Department of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Sette
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Carmine M Carapella
- Division of Neurosurgery, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Rosario Amato
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Nicola Perrotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Michele Signore
- RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00162, Rome, Italy.
| | - Marco G Paggi
- Section of Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
20
|
Smac mimetic suppresses tunicamycin-induced apoptosis via resolution of ER stress. Cell Death Dis 2019; 10:155. [PMID: 30770792 PMCID: PMC6377606 DOI: 10.1038/s41419-019-1381-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/02/2018] [Accepted: 12/19/2018] [Indexed: 01/02/2023]
Abstract
Since Inhibitor of Apoptosis (IAP) proteins have been implicated in cellular adaptation to endoplasmic reticulum (ER) stress, we investigated the regulation of ER stress-induced apoptosis by small-molecule second mitochondria-derived activator of caspase (Smac) mimetics that antagonize IAP proteins. Here, we discover that Smac mimetic suppresses tunicamycin (TM)-induced apoptosis via resolution of the unfolded protein response (UPR) and ER stress. Smac mimetics such as BV6 selectively inhibit apoptosis triggered by pharmacological or genetic inhibition of protein N-glycosylation using TM or knockdown of DPAGT1, the enzyme that catalyzes the first step of protein N-glycosylation. In contrast, BV6 does not rescue cell death induced by other typical ER stressors (i.e., thapsigargin (TG), dithiothreitol, brefeldin A, bortezomib, or 2-deoxyglucose). The protection from TM-triggered apoptosis is found for structurally different Smac mimetics and for genetic knockdown of cellular IAP (cIAP) proteins in several cancer types, underlining the broader relevance. Interestingly, lectin microarray profiling reveals that BV6 counteracts TM-imposed inhibition of protein glycosylation. BV6 consistently abolishes TM-stimulated accumulation of ER stress markers such as glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) and reduces protein kinase RNA-like ER kinase (PERK) phosphorylation and X box-binding protein 1 (XBP1) splicing upon TM treatment. BV6-stimulated activation of nuclear factor-κB (NF-κB) contributes to the resolution of ER stress, since NF-κB inhibition by overexpression of dominant-negative IκBα superrepressor counteracts the suppression of TM-stimulated transcriptional activation of CHOP and GRP78 by BV6. Thus, our study is the first to show that Smac mimetic protects from TM-triggered apoptosis by resolving the UPR and ER stress. This provides new insights into the regulation of cellular stress responses by Smac mimetics.
Collapse
|
21
|
Cong H, Xu L, Wu Y, Qu Z, Bian T, Zhang W, Xing C, Zhuang C. Inhibitor of Apoptosis Protein (IAP) Antagonists in Anticancer Agent Discovery: Current Status and Perspectives. J Med Chem 2019; 62:5750-5772. [DOI: 10.1021/acs.jmedchem.8b01668] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Hui Cong
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Lijuan Xu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yougen Wu
- College of Tropical Agriculture and Forestry, Hainan University, 58 Renmin Avenue, Haikou 570228, China
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Tengfei Bian
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
22
|
Signal transduction pathways and resistance to targeted therapies in glioma. Semin Cancer Biol 2019; 58:118-129. [PMID: 30685341 DOI: 10.1016/j.semcancer.2019.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
Abstract
Although surgical techniques and adjuvant therapies have undergone progressive development for decades, the therapeutic outcomes for treating glioblastoma (GBM) remain poor. The main reasons for the poor prognosis of gliomas are that limited tumor tissue that can be resected (to preserve brain functions) and that residual tumors are often resistant to irradiation and chemotherapy. Therefore, overcoming the resistance of residual tumors against adjuvant therapy is urgently needed for glioma treatment. Recent large cohort studies of genetic alterations in GBM demonstrated that both genetic information and intracellular molecular signaling are networked in gliomas and that such information may help clarify which molecules or signals serve essential roles in resistance against radiation or chemotherapy, highlighting them as potential novel therapeutic targets against refractory gliomas. In this review, we summarize the current understanding of molecular networks that govern glioma biology, mainly based on cohort studies or recent evidence, with a focus on how intracellular signaling molecules in gliomas associate with each other and regulate refractoriness against current therapy.
Collapse
|
23
|
Ma YS, Lin JJ, Lin CC, Lien JC, Peng SF, Fan MJ, Hsu FT, Chung JG. Benzyl isothiocyanate inhibits human brain glioblastoma multiforme GBM 8401 cell xenograft tumor in nude mice in vivo. ENVIRONMENTAL TOXICOLOGY 2018; 33:1097-1104. [PMID: 29972272 DOI: 10.1002/tox.22581] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
Benzyl isothiocyanate (BITC), a member of isothiocyanates (ITCs), has been shown to induce cell death in many human cancer cells, but there is no further report to show BITC suppresses glioblastoma multiforme cells in vivo. In the present study, we investigate the effects of BITC on the inhibition of GBM 8401/luc2 cell generated tumor on athymic nude mice. We established a luciferase expressing stable clone named as GBM 8401/luc2. Thirty male mice were inoculated subcutaneously with GBM 8401/luc2 cells to generate xenograft tumor mice model. Group I was treated with 110 μL phosphate-buffered solution plus 10 μL dimethyl sulfoxide, Group II-III with BITC (5 or 10 μmol/100 μL/day, relatively). Mice were given oral treatment of BITC by gavage for 21 days. Results showed that BITC did not affect the body weights. After anesthetized, the photons emitted from mice tumor were detected with Xenogen IVIS imaging system 200 and higher dose of BITC have low total photon flux than that of lower dose of BITC. Results also showed that higher dose of BITC have low total tumor volumes and weights than that of low dose of BITC. Isolated tumors were investigated by immunohistochemical analysis and results showed that BITC at both dose of treatment weakly stained with anti-MCL1 and -XIAP. However, both dose of BITC treatments have strong signals of caspase-3 and Bax. Overall, these data demonstrated that BITC suppressed tumor properties in vivo. Overall, based on these observations, BITC can be used against human glioblastoma multiforme in the future.
Collapse
Affiliation(s)
- Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Jen-Jyh Lin
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Chin-Chung Lin
- Department of Chinese Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Executive Yuan, Taichung, Taiwan
- General Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Jen Fan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Fei-Ting Hsu
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Radiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jing-Gung Chung
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Hasslacher S, Schneele L, Stroh S, Langhans J, Zeiler K, Kattner P, Karpel-Massler G, Siegelin MD, Schneider M, Zhou S, Grunert M, Halatsch ME, Nonnenmacher L, Debatin KM, Westhoff MA. Inhibition of PI3K signalling increases the efficiency of radiotherapy in glioblastoma cells. Int J Oncol 2018; 53:1881-1896. [PMID: 30132519 PMCID: PMC6192725 DOI: 10.3892/ijo.2018.4528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, the most common primary brain tumour, is also considered one of the most lethal cancers per se. It is highly refractory to therapeutic intervention, as highlighted by the mean patient survival of only 15 months, despite an aggressive treatment approach, consisting of maximal safe surgical resection, followed by radio- and chemotherapy. Radiotherapy, in particular, can have effects on the surviving fractions of tumour cells, which are considered adverse to the desired clinical outcome: It can induce increased cellular proliferation, as well as enhanced invasion. In this study, we established that differentiated glioblastoma cells alter their DNA repair response following repeated exposure to radiation and, therefore, high single-dose irradiation (SD-IR) is not a good surrogate marker for fractionated dose irradiation (FD-IR), as used in clinical practice. Integrating irradiation into a combination therapy approach, we then investigated whether the pharmacological inhibition of PI3K signalling, the most abundantly activated survival cascade in glioblastoma, enhances the efficacy of radiotherapy. Of note, treatment with GDC-0941, which blocks PI3K-mediated signalling, did not enhance cell death upon irradiation, but both treatment modalities functioned synergistically to reduce the total cell number. Furthermore, GDC-0941 not only prevented the radiation-induced increase in the motility of the differentiated cells, but further reduced their speed below that of untreated cells. Therefore, combining radiotherapy with the pharmacological inhibition of PI3K signalling is a potentially promising approach for the treatment of glioblastoma, as it can reduce the unwanted effects on the surviving fraction of tumour cells.
Collapse
Affiliation(s)
- Sebastian Hasslacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Lukas Schneele
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Sebastien Stroh
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Julia Langhans
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Katharina Zeiler
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Patricia Kattner
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Matthias Schneider
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Shaoxia Zhou
- Department of Clinical Chemistry, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Michael Grunert
- Department of Radiology, German Armed Forces Hospital of Ulm, D-89081 Ulm, Germany
| | - Marc-Eric Halatsch
- Department of Neurosurgery, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| |
Collapse
|
25
|
Yuan S, Qiao T, Li X, Zhuang X, Chen W, Chen X, Zhang Q. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol Lett 2018. [PMID: 29541253 DOI: 10.3892/ol.2018.7916] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Unmethylated cytosine-phosphorothioate-guanine (CpG)-containing oligodeoxynucleotides (ODNs) are synthetic DNA sequences that mimic bacterial DNA, and are known to serve as ligands for Toll-like receptor 9 (TLR9). The interaction between a CpG ODNs with TLR9 activates the complex downstream cascade that contributes to exerting its function. In the present study, the results of clonogenic assays demonstrated that the activation of TLR9 by CpG ODNs significantly increased the radiosensitivity of A549 lung cancer cells, with a sensitivity enhancement ratio (SER) of 1.28. When the expression of TLR9 was effectively silenced, CpG ODNs used alone were identified to produce SERs as low as 1.01. Flow cytometry demonstrated that the interaction between TLR9 and CpG ODN 7909 alone did not significantly affect the rate of apoptosis, but may significantly enhance the radiation-induced apoptosis of A549 cells. Western blot analysis revealed that TLR9 activation by CpG ODN 7909 increased the levels of mitogen-activated protein kinase 14, cellular tumor antigen p53, B-cell lymphoma 2 associated X protein and genome polyprotein, and decreased Bcl-2 expression levels, whereas these effects were not observed in CpG ODN 7909-treated cells in which TLR9 was knocked down. These results suggest that CpG ODN 7909 may enhance radiosensitivity through TLR9 activation, and partially via the p53 pathway in A549 lung cancer cells.
Collapse
Affiliation(s)
- Sujuan Yuan
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Tiankui Qiao
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xuan Li
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xibing Zhuang
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Wei Chen
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| | - Xue Chen
- Department of Radiology, Shanghai Cancer Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Qi Zhang
- Department of Oncology, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
26
|
Cell death-based treatment of glioblastoma. Cell Death Dis 2018; 9:121. [PMID: 29371590 PMCID: PMC5833770 DOI: 10.1038/s41419-017-0021-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/19/2017] [Accepted: 09/26/2017] [Indexed: 12/26/2022]
Abstract
Cancer cells including glioblastoma have typically evolved multiple mechanisms to escape programmed cell death in order to maintain their survival. Defects in cell death mechanisms not only facilitate tumorigenesis but also ensure resistance to current anticancer therapies. This emphasizes that targeting cell death pathways may provide a means to tackle one of the Achilles' heels of cancer. Over the last decades several approaches have been developed to selectively target cell death pathways for therapeutic purposes. Some of these concepts have already been transferred into clinical application in oncology and may open new perspectives for the treatment of cancer.
Collapse
|
27
|
Fulda S. Therapeutic opportunities based on caspase modulation. Semin Cell Dev Biol 2017; 82:150-157. [PMID: 29247787 DOI: 10.1016/j.semcdb.2017.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Caspases are a family of proteolytic enzymes that play a critical role in the regulation of programmed cell death via apoptosis. Activation of caspases is frequently impaired in human cancers, contributing to cancer formation, progression and therapy resistance. A better understanding of the molecular mechanisms regulating caspase activation in cancer cells is therefore highly important. Thus, targeted modulation of caspase activation and apoptosis represents a promising approach for the development of new therapeutic options to elucidate cancer cell death.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
28
|
Gilormini M, Malesys C, Armandy E, Manas P, Guy JB, Magné N, Rodriguez-Lafrasse C, Ardail D. Preferential targeting of cancer stem cells in the radiosensitizing effect of ABT-737 on HNSCC. Oncotarget 2017; 7:16731-44. [PMID: 26934442 PMCID: PMC4941347 DOI: 10.18632/oncotarget.7744] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/13/2016] [Indexed: 12/26/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are common human malignancies with poor clinical outcomes. The 5-year survival rates for patients with advanced stage HNSCC have not changed appreciably in the past few decades, underscoring a dire need for improved therapeutic options. HNSCC is frequently characterized by overexpression of anti-apoptotic Bcl-2 family members. Increased levels of these anti-apoptotic proteins have been associated with radio- and chemoresistance and poor clinical outcome. The aim of this study was to evaluate combined effects of radiation and ABT-737, a BH3-mimetic molecule, in HNSCC. Although ABT-737, as a single agent, was largely ineffective at promoting HNSCC cell death, we found that combining ABT-737 and radiation induced strong synergistic apoptosis in HNSCC cell lines and delayed tumoral growth in vivo. Moreover, we demonstrated for the first time that ABT-737, alone or in combination with radiation, can efficiently eliminate cancer stem cells (CSCs). Altogether, our results indicate that therapy targeting anti-apoptotic Bcl-2 family members could be a highly effective potential adjuvant to radiotherapy capable of targeting CSCs in HNSCC and therefore overcoming cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Marion Gilormini
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Céline Malesys
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Emma Armandy
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Patrick Manas
- UMS3444 BioSciences Gerland-Lyon Sud, PBES, Lyon, France
| | - Jean-Baptiste Guy
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Nicolas Magné
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Institut de Cancérologie L. Neuwirth, St Etienne, France
| | - Claire Rodriguez-Lafrasse
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Hospices-Civils-de-Lyon, CHLS, Pierre-Bénite, France
| | - Dominique Ardail
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Hospices-Civils-de-Lyon, CHLS, Pierre-Bénite, France
| |
Collapse
|
29
|
Tirapelli DPDC, Lustosa IL, Menezes SB, Franco IM, Rodrigues AR, Peria FM, Marinho AMDN, Serafini LN, Carlotti Jr CG, Tirapelli LF. High expression of XIAP and Bcl-2 may inhibit programmed cell death in glioblastomas. ARQUIVOS DE NEURO-PSIQUIATRIA 2017; 75:875-880. [DOI: 10.1590/0004-282x20170156] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/14/2017] [Indexed: 11/21/2022]
Abstract
ABSTRACT Glioblastoma (GBM) is the most malignant glioma and represents 29% of all brain tumors. Tumorigenesis is intimately connected with characteristics acquired in the physiologic pathway of cellular death. Objective: In the present study, the expression of anti-apoptotic (XIAP and Bcl-2) and apoptotic (cytochrome C, caspase 9, APAF-1), caspase 3 and the Smac/DIABLO genes related to the apoptosis pathway were evaluated in 30 samples of glioblastoma. Methods: The gene expression was evaluated in 30 glioblastomas (WHO grade IV) and compared to 10 white matter control samples with real-time PCR. Results and Conclusion: There were higher expressions of XIAP (p = 0.0032) and Bcl-2 (p = 0.0351) in the glioblastoma samples compared to the control samples of normal brain. These results raise the question of whether Bcl-2 and XIAP genes can be responsible for the inhibition of programmed cell death in glioblastomas. Moreover, they provide additional information capable of allowing the development of new target therapy strategies.
Collapse
|
30
|
Miranda A, Blanco-Prieto MJ, Sousa J, Pais A, Vitorino C. Breaching barriers in glioblastoma. Part II: Targeted drug delivery and lipid nanoparticles. Int J Pharm 2017; 531:389-410. [DOI: 10.1016/j.ijpharm.2017.07.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 02/07/2023]
|
31
|
Emery IF, Gopalan A, Wood S, Chow KH, Battelli C, George J, Blaszyk H, Florman J, Yun K. Expression and function of ABCG2 and XIAP in glioblastomas. J Neurooncol 2017; 133:47-57. [PMID: 28432589 PMCID: PMC5627495 DOI: 10.1007/s11060-017-2422-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/09/2017] [Indexed: 12/16/2022]
Abstract
Despite multimodal treatment that includes surgery, radiation and chemotherapy, virtually all glioblastomas (GBM) recur, indicating that these interventions are insufficient to eradicate all malignant cells. To identify potential new therapeutic targets in GBMs, we examined the expression and function of proteins that are associated with therapy resistance and cancer cell survival. We measured the expression of eight such proteins in 50 GBM samples by immunohistochemistry and analyzed patient survival. We report that GBM patients with high expression of ABCG2 (also called BCRP) or XIAP at the protein level had worse survival than those with low expression. The adjusted hazard ratio for ABCG2 was 2.35 and for XIAP was 2.65. Since glioma stem cells (GSCs) have been shown to be more resistant than bulk tumor cells to anti-cancer therapies and to express high levels of these proteins, we also sought to determine if ABCG2 and XIAP have functional roles in GSCs. We used small molecule inhibitors to treat patient-derived GBM tumorspheres in vitro and observed that inhibitors of ABCG2, Ko143 and fumitremorgin, significantly reduced self-renewal. These results suggest that ABCG2 and XIAP proteins may be useful indicators of patient survival and that inhibition of ABCG2 may be a promising therapeutic strategy in GBMs.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Brain Neoplasms/drug therapy
- Brain Neoplasms/metabolism
- Brain Neoplasms/mortality
- Brain Neoplasms/radiotherapy
- Cells, Cultured
- Dacarbazine/analogs & derivatives
- Dacarbazine/therapeutic use
- Diketopiperazines/pharmacology
- Female
- Follow-Up Studies
- Glioblastoma/drug therapy
- Glioblastoma/metabolism
- Glioblastoma/mortality
- Glioblastoma/radiotherapy
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Humans
- Indoles/pharmacology
- Male
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Middle Aged
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Neoplasm Transplantation
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Temozolomide
- X-Linked Inhibitor of Apoptosis Protein/metabolism
Collapse
Affiliation(s)
- Ivette F Emery
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA.
| | - Archana Gopalan
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Stephanie Wood
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Kin-Hoe Chow
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Chiara Battelli
- New England Cancer Specialists, 100 Campus Drive, Suite 108, Scarborough, ME, 04074, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Hagen Blaszyk
- Maine Medical Center Department of Pathology, 22 Bramhall Street, Portland, ME, 04102, USA
| | - Jeffrey Florman
- Maine Medical Center Neuroscience Institute, 22 Bramhall Street, Portland, ME, 04102, USA
| | - Kyuson Yun
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
- Peak Center for Brain and Pituitary Tumors, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Wang P, Yuan D, Guo F, Chen X, Zhu L, Zhang H, Wang C, Shao C. Chromatin remodeling modulates radiosensitivity of the daughter cells derived from cell population exposed to low- and high-LET irradiation. Oncotarget 2017; 8:52823-52836. [PMID: 28881774 PMCID: PMC5581073 DOI: 10.18632/oncotarget.17275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/27/2022] Open
Abstract
Radiation effects are dependent of linear energy transfer (LET), but it is still obscure whether the daughter cells (DCs) derived from irradiated population are radioresistance and much less the underlying mechanism. With the measurements of survival, proliferation and γH2AX foci, this study shows that the DCs from γ-ray irradiated cells (DCs-γ) became more radioresistant than its parent control without irradiation, but the radiosensitivity of DCs from α-particle irradiated cells (DCs-α) was not altered. After irradiation with equivalent doses of γ-rays and α-particles, the foci number of histone H3 lysine 9 dimethylation (H3K9me3) and the activity of histone deacetylase (HDAC) in DCs-γ was extensively higher than these in DCs-α and its parent control, indicating that a higher level of heterochromatin was formed in DCs-γ but not in DCs-α. Treatment of cells with SAHA (an inhibitor of HDAC) decreased the level of heterochromatin domains by inhibiting the expressions of H3K9m3 and HP-1a proteins and triggering the expression of acetylated core histone H3 (Ac-H3). When cells were treated with SAHA, the radioresistance phenotype of DCs-γ was eliminated so that the radiosensitivities of DCs-γ, DCs-α and their parent cells approached to same levels. Our current results reveal that γ-rays but not α-particles could induce chromatin remodeling and heterochromatinization which results in the occurrence of radioresistance of DCs, indicating that the combination treatment of irradiation and HDAC inhibitor could serve as a potential cancer therapy strategy, especially for the fraction radiotherapy of low-LET irradiation.
Collapse
Affiliation(s)
- Ping Wang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Dexiao Yuan
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Fei Guo
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Xiaoyan Chen
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Lin Zhu
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Hang Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chen Wang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|
33
|
Fulda S. Smac Mimetics to Therapeutically Target IAP Proteins in Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 330:157-169. [PMID: 28215531 DOI: 10.1016/bs.ircmb.2016.09.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Inhibitor of Apoptosis (IAP) proteins are overexpressed in a variety of human cancers. Therefore, they are considered as promising targets for the design of therapeutic strategies. Smac mimetics mimic the endogenous mitochondrial protein Smac that antagonizes IAP proteins upon its release into the cytosol. Multiple preclinical studies have documented the ability of Smac mimetics to either directly induce cell death of cancer cells or to prime them to agents that trigger cell death. At present, several Smac mimetics are being evaluated in early clinical trials. The current review provides an overview on the potential of Smac mimetics as cancer therapeutics to target IAP proteins for cancer therapy.
Collapse
Affiliation(s)
- S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
34
|
Rahmanian N, Hosseinimehr SJ, Khalaj A. The paradox role of caspase cascade in ionizing radiation therapy. J Biomed Sci 2016; 23:88. [PMID: 27923354 PMCID: PMC5142153 DOI: 10.1186/s12929-016-0306-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy alone or in combination with chemotherapy/surgery is widely used for treatment of cancers. It reduces tumor growth and prevents metastasis. While ionizing radiation activates caspase cascade resulted in apoptosis in cancer cells, it also stimulates tumor cell re-population that leads to reduce the effectiveness of the radiation therapy. This review describes the mechanisms for paradox role of caspase cascade in cancer therapy and discusses the logical and practical strategies for improvement the therapeutic index of radiotherapy through enhancement of radiosensitivity and decreasing the rate of tumor recurrence.
Collapse
Affiliation(s)
- Najmeh Rahmanian
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Khalaj
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Reactive oxygen species contribute toward Smac mimetic/temozolomide-induced cell death in glioblastoma cells. Anticancer Drugs 2016; 27:953-959. [PMID: 27669171 DOI: 10.1097/cad.0000000000000412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Small-molecule inhibitors of Inhibitor of Apoptosis proteins such as Smac mimetics have been reported to provide a promising tool to sensitize glioblastoma (GBM) cells to cytotoxic therapies including chemotherapeutic drugs. However, the underlying molecular mechanisms of action have not yet been fully unraveled. In the present study, we therefore investigated the role of reactive oxygen species (ROS) in the regulation of Smac mimetic/temozolomide (TMZ)-induced cell death in GBM cells. Here, we show that the Smac mimetic BV6 and TMZ act in concert to stimulate the production of both cytosolic and mitochondrial ROS. This accumulation of ROS contributes toward the activation of the proapoptotic factor BAX upon BV6/TMZ cotreatment as several ROS scavengers (i.e. N-acetyl-L-cysteine, MnTBAP, or α-tocopherol) protect GBM cells against BV6/TMZ-mediated BAX activation. In addition, ROS scavengers significantly rescue GBM cells from BV6/TMZ-triggered cell death, indicating that ROS generation is required for the induction of cell death. By showing that ROS play an important role in the regulation of Smac mimetic/TMZ-induced cell death, our work sheds light on the crucial role of the oxidative system in the cooperative antitumor activity of Smac mimetic/TMZ combination therapy against GBM cells.
Collapse
|
36
|
Abstract
Inhibitor of Apoptosis (IAP) proteins block programmed cell death and are expressed at high levels in various human cancers, thus making them attractive targets for cancer drug development. Second mitochondrial activator of caspases (Smac) mimetics are small-molecule inhibitors that mimic Smac, an endogenous antagonist of IAP proteins. Preclinical studies have shown that Smac mimetics can directly trigger cancer cell death or, even more importantly, sensitize tumor cells for various cytotoxic therapies, including conventional chemotherapy, radiotherapy, or novel agents. Currently, several Smac mimetics are under evaluation in early clinical trials as monotherapy or in rational combinations (i.e., GDC-0917/CUDC-427, LCL161, AT-406/Debio1143, HGS1029, and TL32711/birinapant). This review discusses the promise as well as some challenges at the translational interface of exploiting Smac mimetics as cancer therapeutics.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
37
|
Wang D, Berglund A, Kenchappa RS, Forsyth PA, Mulé JJ, Etame AB. BIRC3 is a novel driver of therapeutic resistance in Glioblastoma. Sci Rep 2016; 6:21710. [PMID: 26888114 PMCID: PMC4757860 DOI: 10.1038/srep21710] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
Genome-wide analysis of glioblastoma (GBM) reveals pervasive aberrations in apoptotic signaling pathways that collectively contribute to therapeutic resistance. Inhibitors of apoptosis proteins (IAP) exert critical control on the terminal segment of apoptosis leading to apoptosis evasion. In this study, we uncover a unique role for BIRC3, as an IAP that is critical in GBM in response to therapy. Using the TCGA dataset of 524 unique samples, we identify BIRC3 is the only IAP whose differential expression is associated with long-term survival in GBM patients. Using patient tissue samples we further show that BIRC3 expression increases with recurrence. When extrapolated to a preclinical model of a human GBM cell line, we find an increase in BIRC3 expression in response to irradiation (RT) and temozolomide (TMZ) treatment. More importantly, we mechanistically implicate STAT3 and PI3K signaling pathways as drivers of RT-induced up-regulation of BIRC3 expression. Lastly, we demonstrate that both in-vivo and in-vitro BIRC3 up-regulation results in apoptosis evasion and therapeutic resistance in GBM. Collectively, our study identifies a novel translational and targetable role for BIRC3 expression as a predictor of aggressiveness and therapeutic resistance to TMZ and RT mediated by STAT3 and PI3K signaling in GBM.
Collapse
Affiliation(s)
- Dapeng Wang
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA.,Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - Anders Berglund
- Department of Medical Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - Rajappa S Kenchappa
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA.,Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - Peter A Forsyth
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA.,Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - James J Mulé
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA.,Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - Arnold B Etame
- Department of Neuro-Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA.,Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612 USA
| |
Collapse
|
38
|
Zakaria Z, Tivnan A, Flanagan L, Murray DW, Salvucci M, Stringer BW, Day BW, Boyd AW, Kögel D, Rehm M, O'Brien DF, Byrne AT, Prehn JHM. Patient-derived glioblastoma cells show significant heterogeneity in treatment responses to the inhibitor-of-apoptosis-protein antagonist birinapant. Br J Cancer 2015; 114:188-98. [PMID: 26657652 PMCID: PMC4815807 DOI: 10.1038/bjc.2015.420] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/10/2015] [Indexed: 11/22/2022] Open
Abstract
Background: Resistance to temozolomide (TMZ) greatly limits chemotherapeutic effectiveness in glioblastoma (GBM). Here we analysed the ability of the Inhibitor-of-apoptosis-protein (IAP) antagonist birinapant to enhance treatment responses to TMZ in both commercially available and patient-derived GBM cells. Methods: Responses to TMZ and birinapant were analysed in a panel of commercial and patient-derived GBM cell lines using colorimetric viability assays, flow cytometry, morphological analysis and protein expression profiling of pro- and antiapoptotic proteins. Responses in vivo were analysed in an orthotopic xenograft GBM model. Results: Single-agent treatment experiments categorised GBM cells into TMZ-sensitive cells, birinapant-sensitive cells, and cells that were insensitive to either treatment. Combination treatment allowed sensitisation to therapy in only a subset of resistant GBM cells. Cell death analysis identified three principal response patterns: Type A cells that readily activated caspase-8 and cell death in response to TMZ while addition of birinapant further sensitised the cells to TMZ-induced cell death; Type B cells that readily activated caspase-8 and cell death in response to birinapant but did not show further sensitisation with TMZ; and Type C cells that showed no significant cell death or moderately enhanced cell death in the combined treatment paradigm. Furthermore, in vivo, a Type C patient-derived cell line that was TMZ-insensitive in vitro and showed a strong sensitivity to TMZ and TMZ plus birinapant treatments. Conclusions: Our results demonstrate remarkable differences in responses of patient-derived GBM cells to birinapant single and combination treatments, and suggest that therapeutic responses in vivo may be greatly affected by the tumour microenvironment.
Collapse
Affiliation(s)
- Z Zakaria
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,National Centre for Neurosurgery, Beaumont Hospital, Dublin 9, Ireland
| | - A Tivnan
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - L Flanagan
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - D W Murray
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - M Salvucci
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - B W Stringer
- Brain Cancer Research Unit, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - B W Day
- Brain Cancer Research Unit, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - A W Boyd
- Brain Cancer Research Unit, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - D Kögel
- Experimental Neurosurgery, Neuroscience Center, Frankfurt University Hospital, Frankfurt am Main, Germany
| | - M Rehm
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - D F O'Brien
- National Centre for Neurosurgery, Beaumont Hospital, Dublin 9, Ireland
| | - A T Byrne
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - J H M Prehn
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
39
|
Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, Siegelin MD, Fimognari C, Kumar NB, Dou QP, Yang H, Samadi AK, Russo GL, Spagnuolo C, Ray SK, Chakrabarti M, Morre JD, Coley HM, Honoki K, Fujii H, Georgakilas AG, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich WG, Yang X, Boosani CS, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Keith WN, Bilsland A, Halicka D, Nowsheen S, Azmi AS. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 2015; 35 Suppl:S78-S103. [PMID: 25936818 PMCID: PMC4720504 DOI: 10.1016/j.semcancer.2015.03.001] [Citation(s) in RCA: 596] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022]
Abstract
Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.
Collapse
Affiliation(s)
- Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Clement Yedjou
- C-SET, [Jackson, #229] State University, Jackson, MS, United States
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, United States
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Italy
| | - Nagi B Kumar
- Moffit Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Departments of Pharmacology and Pathology, Karmanos Cancer Institute, Detroit MI, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | | | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - James D Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, United Arab Emirates; Faculty of Science, Cairo University, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, United Arab Emirates
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine Creighton University, Omaha NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
40
|
Yoon TM, Kim SA, Cho WS, Lee DH, Lee JK, Park YL, Lee KH, Lee JH, Kweon SS, Chung IJ, Lim SC, Joo YE. SOX4 expression is associated with treatment failure and chemoradioresistance in oral squamous cell carcinoma. BMC Cancer 2015; 15:888. [PMID: 26555193 PMCID: PMC4641419 DOI: 10.1186/s12885-015-1875-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/30/2015] [Indexed: 01/01/2023] Open
Abstract
Background In humans, sex-determining region-Y (SRY) related high-mobility-group box 4 (SOX4) is linked to development and tumorigenesis. SOX4 is over-expressed in several cancers and has prognostic significance. This study evaluated whether SOX4 affects oncogenic behavior and chemoradiotherapy response in head and neck squamous cell carcinoma (HNSCC) cells, and documented the relationship between its expression and prognosis in oral squamous cell carcinoma (OSCC). Methods We used small interfering RNA in HNSCC cells to evaluate the effect of SOX4 on cell proliferation, apoptosis, chemoradiation-induced apoptosis, invasion, and migration. SOX4 expression in OSCC tissues was investigated by immunohistochemistry. Results SOX4 knockdown (KO) decreased cell proliferation and induced apoptosis by activating caspases-3 and −7, and poly-ADP ribose polymerase and suppressing X-linked inhibitor of apoptosis protein in HNSCC cells; it also enhanced radiation/cisplatin-induced apoptosis; and suppressed tumor cell invasion and migration. Immunostaining showed SOX4 protein was significantly increased in OSCC tissues compared with adjacent normal mucosa. SOX4 expression was observed in 51.8 % of 85 OSCC tissues, and was significantly correlated with treatment failure (P = 0.032) and shorter overall survival (P = 0.036) in patients with OSCC. Conclusions SOX4 may contribute to oncogenic phenotypes of HNSCC cells by promoting cell survival and causing chemoradioresistance. It could be a potential prognostic marker for OSCC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1875-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tae Mi Yoon
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Sun-Ae Kim
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Wan Seok Cho
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Dong Hoon Lee
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Joon Kyoo Lee
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Young-Lan Park
- Departments of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Kyung-Hwa Lee
- Departments of Pathology, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Jae Hyuk Lee
- Departments of Pathology, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Sun-Seog Kweon
- Departments of Preventive Medicine, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Ik-Joo Chung
- Departments of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Sang Chul Lim
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| | - Young-Eun Joo
- Departments of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, 8 Hak-Dong, Dong-Ku, Gwangju, 501-757, South Korea.
| |
Collapse
|
41
|
Smac mimetic-induced upregulation of interferon-β sensitizes glioblastoma to temozolomide-induced cell death. Cell Death Dis 2015; 6:e1888. [PMID: 26379193 PMCID: PMC4650438 DOI: 10.1038/cddis.2015.235] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/09/2015] [Accepted: 07/20/2015] [Indexed: 12/13/2022]
Abstract
Inhibitor of apoptosis (IAP) proteins are frequently expressed at high levels in cancer cells and represent attractive therapeutic targets. We previously reported that the Smac (second mitochondria-derived activator of caspases) mimetic BV6, which antagonizes IAP proteins, sensitizes glioblastoma cells to temozolomide (TMZ)-induced cell death in a nuclear factor-κB (NF-κB)-dependent manner. However, BV6-induced NF-κB target genes responsible for this synergistic interaction have remained elusive. Using whole-genome gene expression profiling, we here identify BV6-stimulated, NF-κB-dependent transcriptional upregulation of interferon-β (IFNβ) and IFN-mediated proapoptotic signaling as critical events that mediate BV6/TMZ-induced apoptosis. Knockdown of IFNβ significantly rescues cells from BV6/TMZ-induced cell death. Similarly, silencing of the corresponding receptor IFNα/β receptor (IFNAR) confers a significant protection against apoptosis, demonstrating that IFNβ and IFN signaling are required for BV6/TMZ-mediated cell death. Moreover, BV6 and TMZ cooperate to transcriptionally upregulate the proapoptotic B-cell lymphoma 2 family proteins Bax (Bcl-2-associated X protein) or Puma (p53-upregulated modulator of apoptosis). Knockdown of Bax or Puma significantly decreases BV6/TMZ-induced apoptosis, showing that both proteins are necessary for apoptosis. By identifying IFNβ as a key mediator of BV6/TMZ-induced apoptosis, our study provides novel insights into the underlying molecular mechanisms of Smac mimetic-mediated chemosensitization with important implications for the development of novel treatment strategies for glioblastoma.
Collapse
|
42
|
Wang YH, Scadden DT. Harnessing the apoptotic programs in cancer stem-like cells. EMBO Rep 2015; 16:1084-98. [PMID: 26253117 DOI: 10.15252/embr.201439675] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 06/19/2015] [Indexed: 12/12/2022] Open
Abstract
Elimination of malignant cells is an unmet challenge for most human cancer types even with therapies targeting specific driver mutations. Therefore, a multi-pronged strategy to alter cancer cell biology on multiple levels is increasingly recognized as essential for cancer cure. One such aspect of cancer cell biology is the relative apoptosis resistance of tumor-initiating cells. Here, we provide an overview of the mechanisms affecting the apoptotic process in tumor cells emphasizing the differences in the tumor-initiating or stem-like cells of cancer. Further, we summarize efforts to exploit these differences to design therapies targeting that important cancer cell population.
Collapse
Affiliation(s)
- Ying-Hua Wang
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
43
|
Opel D, Schnaiter A, Dodier D, Jovanovic M, Gerhardinger A, Idler I, Mertens D, Bullinger L, Stilgenbauer S, Fulda S. Targeting inhibitor of apoptosis proteins by Smac mimetic elicits cell death in poor prognostic subgroups of chronic lymphocytic leukemia. Int J Cancer 2015; 137:2959-70. [PMID: 26096065 DOI: 10.1002/ijc.29650] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/20/2015] [Accepted: 05/29/2015] [Indexed: 12/21/2022]
Abstract
Inhibitor of apoptosis (IAP) proteins are highly expressed in chronic lymphocytic leukemia (CLL) cells and contribute to evasion of cell death and poor therapeutic response. Here, we report that Smac mimetic BV6 dose-dependently induces cell death in 28 of 51 (54%) investigated CLL samples, while B-cells from healthy donors are largely unaffected. Importantly, BV6 is significantly more effective in prognostic unfavorable cases with, e.g., non-mutated VH status and TP53 mutation than samples with unknown or favorable prognosis. The majority of cases with 17p deletion (10/12) and Fludarabine refractory cases respond to BV6, indicating that BV6 acts independently of p53. BV6 also triggers cell death under survival conditions mimicking the microenvironment, e.g., by adding CD40 ligand or conditioned medium. Gene expression profiling identifies cell death, NF-κB and redox signaling among the top pathways regulated by BV6 not only in CLL but also in core-binding factor (CBF) acute myeloid leukemia (AML). Consistently, BV6 stimulates production of reactive oxygen species (ROS), which are contributing to BV6-induced cell death, since antioxidants reduce cell death. While BV6 causes degradation of cellular inhibitor of apoptosis (cIAP)1 and cIAP2 and nuclear factor-kappaB (NF-κB) pathway activation in primary CLL samples, BV6 induces cell death independently of caspase activity, receptor-interacting protein (RIP)1 activity or tumor necrosis factor (TNF)α, as zVAD.fmk, necrostatin-1 or TNFα-blocking antibody Enbrel fail to inhibit cell death. Together, these novel insights into BV6-regulated cell death in CLL have important implications for developing new therapeutic strategies to overcome cell death resistance especially in poor prognostic CLL subgroups.
Collapse
Affiliation(s)
- Daniela Opel
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Andrea Schnaiter
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Dagmar Dodier
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Marjana Jovanovic
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Irina Idler
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Daniel Mertens
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Lars Bullinger
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, , Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
44
|
Steinwascher S, Nugues AL, Schoeneberger H, Fulda S. Identification of a novel synergistic induction of cell death by Smac mimetic and HDAC inhibitors in acute myeloid leukemia cells. Cancer Lett 2015; 366:32-43. [PMID: 26028172 DOI: 10.1016/j.canlet.2015.05.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/17/2015] [Accepted: 05/22/2015] [Indexed: 10/23/2022]
Abstract
Inhibitor of Apoptosis (IAP) proteins are expressed at high levels in acute myeloid leukemia (AML) and contribute to resistance to programmed cell death. Here, we report that inhibition of IAP proteins by the small-molecule Smac mimetic BV6 acts together with histone deacetylase (HDAC) inhibitors (HDACIs) such as MS275 or SAHA to trigger cell death in AML cell lines in a synergistic manner, as underscored by calculation of combination index (CI). Also, BV6 and HDACIs cooperate to trigger DNA fragmentation, a marker of apoptotic cell death, and to suppress long-term clonogenic survival of AML cells. In contrast, equimolar concentrations of BV6 and MS275 or SAHA do not synergize to elicit cell death in normal peripheral blood lymphocytes (PBLs), emphasizing some tumor cell selectivity of this combination treatment. Addition of the tumor necrosis factor (TNF)α-blocking antibody Enbrel significantly reduces BV6/MS275-induced cell death in the majority of AML cell lines, indicating that autocrine/paracrine TNFα signaling contributes to cell death. Remarkably, the broad-range caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (zVAD.fmk) fails to rescue MV4-11, Molm13 and OCI-AML3 cells and even enhances BV6/MS275-mediated cell death, whereas zVAD.fmk reduces BV6/MS275-induced cell death in NB4 cells. Annexin-V/propidium iodide (PI) double staining reveals that BV6/MS275 cotreatment predominately increases the percentage of double-positive cells. Of note, the Receptor-Interacting Protein (RIP)1 inhibitor necrostatin-1 (Nec-1) or the Mixed Lineage Kinase Domain-Like protein (MLKL) inhibitor necrosulfonamide (NSA) significantly reduce BV6/MS275-induced cell death in the presence of zVAD.fmk, suggesting that BV6/MS275 cotreatment triggers necroptosis when caspases are inhibited. Thus, BV6 acts in concert with HDACIs to induce cell death in AML cells and can bypass apoptosis resistance, at least in several AML cell lines, by engaging necroptosis as an alternative route of regulated cell death. The identification of a novel synergism of BV6 and HDACIs has important implications for the development of new treatment strategies for AML.
Collapse
Affiliation(s)
- Sofie Steinwascher
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, Frankfurt 60528, Germany
| | - Anne-Lucie Nugues
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, Frankfurt 60528, Germany
| | - Hannah Schoeneberger
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, Frankfurt 60528, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, Frankfurt 60528, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
45
|
Fulda S. Targeting IAP proteins in combination with radiotherapy. Radiat Oncol 2015; 10:105. [PMID: 25927408 PMCID: PMC4436972 DOI: 10.1186/s13014-015-0399-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/01/2015] [Indexed: 01/30/2023] Open
Abstract
The efficacy of radiotherapy critically depends on the activation of intrinsic cell death programs in cancer cells. This implies that evasion of cell death, a hallmark of human cancers, can contribute to radioresistance. Therefore, novel strategies to reactivate cell death programs in cancer cells are required in order to overcome resistance to radiotherapy. Since Inhibitor of Apoptosis (IAP) proteins are expressed at high levels in multiple cancers and block cell death induction at a central point, therapeutic targeting of IAP proteins represents a promising approach to potentiate the efficacy of radiotherapy. The current review discusses the concept of targeting IAP proteins in combination with radiotherapy.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
46
|
N-acetylcysteine relieves oxidative stress and protects hippocampus of rat from radiation-induced apoptosis by inhibiting caspase-3. Biomed Pharmacother 2015; 70:1-6. [DOI: 10.1016/j.biopha.2014.12.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 12/11/2014] [Indexed: 12/11/2022] Open
|
47
|
Yang Y, Xu H, Huang W, Ding M, Xiao J, Yang D, Li H, Liu XY, Chu L. Targeting lung cancer stem-like cells with TRAIL gene armed oncolytic adenovirus. J Cell Mol Med 2015; 19:915-23. [PMID: 25683371 PMCID: PMC4420595 DOI: 10.1111/jcmm.12397] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/18/2014] [Indexed: 12/11/2022] Open
Abstract
Lung cancer stem cell (LCSC) is critical in cancer initiation, progression, drug resistance and relapse. Disadvantages showed in conventional lung cancer therapy probably because of its existence. In this study, lung cancer cell line A549 cells propagated as spheroid bodies (named as A549 sphere cells) in growth factors-defined serum-free medium. A549 sphere cells displayed CSC properties, including chemo-resistance, increased proportion of G0/G1 cells, slower proliferation rate, ability of differentiation and enhanced tumour formation ability in vivo. Oncolytic adenovirus ZD55 carrying EGFP gene, ZD55-EGFP, infected A549 sphere cells and inhibited cell growth. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) armed oncolytic adenovirus, ZD55-TRAIL, exhibited enhanced cytotoxicity and induced A549 sphere cells apoptosis through mitochondrial pathway. Moreover, small molecules embelin, LY294002 and resveratrol improved the cytotoxicity of ZD55-TRAIL. In the A549 sphere cells xenograft models, ZD55-TRAIL significantly inhibited tumour growth and improved survival status of mice. These results suggested that gene armed oncolytic adenovirus is a potential approach for lung cancer therapy through targeting LCSCs.
Collapse
Affiliation(s)
- Yu Yang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Maccalli C, De Maria R. Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother 2015; 64:91-7. [PMID: 25104304 PMCID: PMC11028671 DOI: 10.1007/s00262-014-1592-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/25/2014] [Indexed: 01/05/2023]
Abstract
Cells with "stemness" and tumor-initiating properties have been isolated from both hematological and solid tumors. These cells denominated as cancer stem cells (CSCs), representing rare populations within tumors, have the ability to metastasize and are resistant to standard therapies and immunotherapy. Heterogeneity and plasticity in the phenotype of CSCs have been described in relation to their tissue origin. Few definitive markers have been isolated for CSCs from human solid tumors, limiting their usage for in vivo identification of these cells. Nevertheless, progress in the emerging CSCs concept has been achieved gaining, at least for some type of tumors, their biological and immunological characterization. The recent identification of molecules and signaling pathways that are up-regulated or aberrantly induced in CSCs allowed the development of small agents for specifically targeting of CSCs. A general low immunogenic profile has been reported for CSCs with, in some cases, the identification of the mechanisms responsible of the impairment of cell-mediated immune responses. These concepts are discussed in the context of this review. Although CSCs still need to be fully characterized, potential candidate markers and/or signaling pathways, to be exploited for the design of novel CSC-targeting therapeutic strategies, are described in this review.
Collapse
Affiliation(s)
- Cristina Maccalli
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors, San Raffaele Foundation Centre, Milan, Italy,
| | | |
Collapse
|
49
|
Abstract
As the Inhibitor of Apoptosis (IAP) proteins are expressed at high levels in human cancers, they represent promising targets for therapeutic intervention. Small-molecule inhibitors of IAP proteins mimicking the endogenous IAP antagonist Smac, called Smac mimetics, neutralize IAP proteins and thereby promote the induction of cell death. Smac mimetics have been shown in preclinical models of human cancer to directly trigger cancer cell death or to sensitize for cancer cell death induced by a variety of cytotoxic stimuli. Smac mimetics are currently undergoing clinical evaluation in phase I/II trials, demonstrating that therapeutic targeting of IAP proteins has reached the clinical stage.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
50
|
Ionizing radiations sustain glioblastoma cell dedifferentiation to a stem-like phenotype through survivin: possible involvement in radioresistance. Cell Death Dis 2014; 5:e1543. [PMID: 25429620 PMCID: PMC4260760 DOI: 10.1038/cddis.2014.509] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022]
Abstract
Glioblastomas (GBM) are some bad prognosis brain tumors despite a conventional
treatment associating surgical resection and subsequent radio-chemotherapy. Among
these heterogeneous tumors, a subpopulation of chemo- and radioresistant GBM
stem-like cells appears to be involved in the systematic GBM recurrence. Moreover,
recent studies showed that differentiated tumor cells may have the ability to
dedifferentiate and acquire a stem-like phenotype, a phenomenon also called
plasticity, in response to microenvironment stresses such as hypoxia. We hypothesized
that GBM cells could be subjected to a similar dedifferentiation process after
ionizing radiations (IRs), then supporting the GBM rapid recurrence after
radiotherapy. In the present study we demonstrated that subtoxic IR exposure of
differentiated GBM cells isolated from patient resections potentiated the long-term
reacquisition of stem-associated properties such as the ability to generate primary
and secondary neurospheres, the expression of stemness markers and an increased
tumorigenicity. We also identified during this process an upregulation of the
anti-apoptotic protein survivin and we showed that its specific downregulation led to
the blockade of the IR-induced plasticity. Altogether, these results demonstrated
that irradiation could regulate GBM cell dedifferentiation via a survivin-dependent
pathway. Targeting the mechanisms associated with IR-induced plasticity will likely
contribute to the development of some innovating pharmacological strategies for an
improved radiosensitization of these aggressive brain cancers.
Collapse
|