651
|
Genetically Engineered Mouse Models of Gliomas: Technological Developments for Translational Discoveries. Cancers (Basel) 2019; 11:cancers11091335. [PMID: 31505839 PMCID: PMC6770673 DOI: 10.3390/cancers11091335] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 01/25/2023] Open
Abstract
The most common brain tumours, gliomas, have significant morbidity. Detailed biological and genetic understanding of these tumours is needed in order to devise effective, rational therapies. In an era generating unprecedented quantities of genomic sequencing data from human cancers, complementary methods of deciphering the underlying functional cancer genes and mechanisms are becoming even more important. Genetically engineered mouse models of gliomas have provided a platform for investigating the molecular underpinning of this complex disease, and new tools for such models are emerging that are enabling us to answer the most important questions in the field. Here, I discuss improvements to genome engineering technologies that have led to more faithful mouse models resembling human gliomas, including new cre/LoxP transgenic lines that allow more accurate cell targeting of genetic recombination, Sleeping Beauty and piggyBac transposons for the integration of transgenes and genetic screens, and CRISPR-cas9 for generating genetic knockout and functional screens. Applications of these technologies are providing novel insights into the functional genetic drivers of gliomagenesis, how these genes cooperate with one another, and the potential cells-of-origin of gliomas, knowledge of which is critical to the development of targeted treatments for patients in the clinic.
Collapse
|
652
|
Factorial Design as a Tool for the Optimization of PLGA Nanoparticles for the Co-Delivery of Temozolomide and O6-Benzylguanine. Pharmaceutics 2019; 11:pharmaceutics11080401. [PMID: 31405159 PMCID: PMC6722980 DOI: 10.3390/pharmaceutics11080401] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/23/2019] [Accepted: 08/08/2019] [Indexed: 12/07/2022] Open
Abstract
Poly(d,l-lactic-co-glycolic) (PLGA) nanoparticles (NPs) have been widely studied for several applications due to their advantageous properties, such as biocompatibility and biodegradability. Therefore, these nanocarriers could be a suitable approach for glioblastoma multiforme (GBM) therapy. The treatment of this type of tumours remains a challenge due to intrinsic resistance mechanisms. Thus, new approaches must be envisaged to target GBM tumour cells potentially providing an efficient treatment. Co-delivery of temozolomide (TMZ) and O6-benzylguanine (O6BG), an inhibitor of DNA repair, could provide good therapeutic outcomes. In this work, a fractional factorial design (FFD) was employed to produce an optimal PLGA-based nanoformulation for the co-loading of both molecules, using a reduced number of observations. The developed NPs exhibited optimal physicochemical properties for brain delivery (dimensions below 200 nm and negative zeta potential), high encapsulation efficiencies (EE) for both drugs, and showed a sustained drug release for several days. Therefore, the use of an FFD allowed for the development of a nanoformulation with optimal properties for the co-delivery of TMZ and O6BG to the brain.
Collapse
|
653
|
G3BP1 knockdown sensitizes U87 glioblastoma cell line to Bortezomib by inhibiting stress granules assembly and potentializing apoptosis. J Neurooncol 2019; 144:463-473. [DOI: 10.1007/s11060-019-03252-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022]
|
654
|
McCarthy D, Starke RM, Sheinberg D, Connolly ES. Treatment Synergy for Glioblastoma Multiforme with SynergySeq. Neurosurgery 2019; 85:E178-E179. [PMID: 30809656 DOI: 10.1093/neuros/nyz030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- David McCarthy
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - Robert M Starke
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - Dallas Sheinberg
- Department of Neurosurgery University of Miami Miller School of Medicine Miami, Florida
| | - E Sander Connolly
- Department of Neurological Surgery Columbia University College of Physicians and Surgeons New York, New York
| |
Collapse
|
655
|
DETERMINATION OF MOLECULAR GENETIC MARKERS IN PROGNOSIS OF THE EFFECTIVENESS OF TREATMENT OF MALIGNANT INTRACEREBRAL BRAIN TUMORS. EUREKA: HEALTH SCIENCES 2019. [DOI: 10.21303/2504-5679.2019.00949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intracerebral malignant brain tumors remain one of the most complex problems of neuro-oncology. Today, promising results of the use of targeted drugs have been received, which determine the important diagnostic and predictive value of molecular genetic markers of glial and metastatic brain tumors.
Aim: The study of the prevalence of MGMT (O6-methylguanine-DNA methyltransferase) and PTEN (phosphatase and tensin homologue deleted on chromosome 10) gene expression by real time polymerase chain reaction in tumor tissue of gliomas and brain metastases.
Materials and methods: From thirty patients were received tumor material (29 cases of glioma III-IV degree of anaplasia and one case of metastatic brain lesion of adenocarcinoma). The normalized expression of MGMT and PTEN genes was determined by real-time polymerase chain reaction.
Results: In all 30 (100 %) patients with tumor fragments, we determined normalized expression of MGMT and PTEN genes. In most cases, 53 % of the observations (16 out of 30 patients) showed a low normalized expression of MGMT gene (<40 c. u.) and a low normalized PTEN expression rate of 73 % (22 out of 30 patients) (<40 c. u.). The average expression level of the MGMT gene in the range from 40 to 100 c. u. (6/20 % of patients) was considered prognostic favourable for the response to temozolomide chemotherapy.
Conclusions: The study of MGMT gene expression, a chemotherapy marker for temozolomide, indicates a trend toward correlation between expression levels and therapeutic efficacy. The study of the expression of the PTEN gene, the blocker of the PI3K / AKT signal pathway, indicates a different degree of expression of this enzyme in the tumour samples studied. The predictive value of the indicator for target therapy is appropriate in comparison with the EGFR mutation. Further profound analysis of the results is required with increasing number of sampling and observation period.
Collapse
|
656
|
The synthesis of a novel Crizotinib heptamethine cyanine dye conjugate that potentiates the cytostatic and cytotoxic effects of Crizotinib in patient-derived glioblastoma cell lines. Bioorg Med Chem Lett 2019; 29:2617-2621. [PMID: 31378572 DOI: 10.1016/j.bmcl.2019.07.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 12/31/2022]
Abstract
We describe the synthesis of drug-dye conjugate 1 between anaplastic lymphoma kinase inhibitor Crizotinib and heptamethine cyanine dye IR-786. The drug-dye conjugate 1 was evaluated in three different patient-derived glioblastoma cell lines and showed potent cytotoxic activity with nanomolar potency (EC50: 50.9 nM). We also demonstrate evidence for antiproliferative activity of 1 with single digit nanomolar potency (IC50: 4.7 nM). Furthermore, the cytotoxic effects conveyed a dramatic, 110-fold improvement over Crizotinib. This improvement was even more pronounced (492-fold) when 1 was combined with Temozolomide, the standard drug for treatment for glioblastoma. This work lays the foundation for future exploration of similar tyrosine kinase inhibitor drug-dye conjugates for the treatment of glioblastoma.
Collapse
|
657
|
Kwon S, Yoo KH, Sym SJ, Khang D. Mesenchymal stem cell therapy assisted by nanotechnology: a possible combinational treatment for brain tumor and central nerve regeneration. Int J Nanomedicine 2019; 14:5925-5942. [PMID: 31534331 PMCID: PMC6681156 DOI: 10.2147/ijn.s217923] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) intrinsically possess unique features that not only help in their migration towards the tumor-rich environment but they also secrete versatile types of secretomes to induce nerve regeneration and analgesic effects at inflammatory sites. As a matter of course, engineering MSCs to enhance their intrinsic abilities is growing in interest in the oncology and regenerative field. However, the concern of possible tumorigenesis of genetically modified MSCs prompted the development of non-viral transfected MSCs armed with nanotechnology for more effective cancer and regenerative treatment. Despite the fact that a large number of successful studies have expanded our current knowledge in tumor-specific targeting, targeting damaged brain site remains enigmatic due to the presence of a blood–brain barrier (BBB). A BBB is a barrier that separates blood from brain, but MSCs with intrinsic features of transmigration across the BBB can efficiently deliver desired drugs to target sites. Importantly, MSCs, when mediated by nanoparticles, can further enhance tumor tropism and can regenerate the damaged neurons in the central nervous system through the promotion of axon growth. This review highlights the homing and nerve regenerative abilities of MSCs in order to provide a better understanding of potential cell therapeutic applications of non-genetically engineered MSCs with the aid of nanotechnology.
Collapse
Affiliation(s)
- Song Kwon
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Kwai Han Yoo
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Sun Jin Sym
- Department of Internal Medicine, Division of Hematology, School of Medicine, Gachon University Gil Medical Center, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea.,Department of Gachon Advanced Institute for Health Science & Technology (Gaihst), Gachon University, Incheon 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
658
|
Wen ZP, Zeng WJ, Chen YH, Li H, Wang JY, Cheng Q, Yu J, Zhou HH, Liu ZZ, Xiao J, Chen XP. Knockdown ATG4C inhibits gliomas progression and promotes temozolomide chemosensitivity by suppressing autophagic flux. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:298. [PMID: 31291988 PMCID: PMC6617611 DOI: 10.1186/s13046-019-1287-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Gliomas are the most common primary tumors in central nervous system. Despite advances in diagnosis and therapy, the prognosis of glioma remains gloomy. Autophagy is a cellular catabolic process that degrades proteins and damaged organelles, which is implicated in tumorigenesis and tumor progression. Autophagy related 4C cysteine peptidase (ATG4C) is an autophagy regulator responsible for cleaving of pro-LC3 and delipidation of LC3 II. This study was designed to investigate the role of ATG4C in glioma progression and temozolomide (TMZ) chemosensitivity. METHODS The association between ATG4C mRNA expression and prognosis of gliomas patients was analyzed using the TCGA datasets. The role of ATG4C in proliferation, apoptosis, autophagy, and TMZ chemosensitivity were investigated by silencing ATG4C in vivo. Ectopic xenograft nude mice model was established to investigate the effects of ATG4C on glioma growth in vivo. RESULTS The median overall survival (OS) time of patients with higher ATG4C expression was significantly reduced (HR: 1.48, p = 9.91 × 10- 7). ATG4C mRNA expression was evidently increased with the rising of glioma grade (p = 2.97 × 10- 8). Knockdown ATG4C suppressed glioma cells proliferation by inducing cell cycle arrest at G1 phase. ATG4C depletion suppressed autophagy and triggered apoptosis through ROS accumulation. Depletion of ATG4C suppressed TMZ-activated autophagy and promoted sensitivity of glioma cells to TMZ. Additionally, ATG4C knockdown suppressed the growth of glioma remarkably in nude mice. CONCLUSION ATG4C is a potential prognostic predictor for glioma patient. Targeting ATG4C may provide promising therapy strategies for gliomas treatment.
Collapse
Affiliation(s)
- Zhi-Peng Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Wen-Jing Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Yan-Hong Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Medical Genetic Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, Hunan, People's Republic of China
| | - He Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jie-Ya Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jing Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Zheng-Zheng Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan province, China.
| | - Jian Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China. .,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, 410078, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
659
|
Tang Z, He Z. TIGAR promotes growth, survival and metastasis through oxidation resistance and AKT activation in glioblastoma. Oncol Lett 2019; 18:2509-2517. [PMID: 31402948 PMCID: PMC6676722 DOI: 10.3892/ol.2019.10574] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma has a poor prognosis and is one of the most lethal types of cancer in the world. TP53 induced glycolysis regulatory phosphatase (TIGAR) is upregulated in various types of cancer. Therefore, the present study investigated the role of TIGAR in glioblastoma. TIGAR expression was measured in glioma samples and cell lines using immunohistochemistry and western blotting. Reduced nicotinamide adenine dinucleotide phosphate (NADPH), glutathione, malondialdehyde and intracellular reactive oxygen species levels were detected to measure oxidative stress in U-87MG cells following short hairpin RNA (shRNA)-mediated knockdown of TIGAR. Cell viability was determined using an MTT assay for TIGAR-overexpression vector- and TIGAR-shRNA-transfected U-87MG cells. Apoptosis was assessed to evaluate whether TIGAR knockdown sensitized cells to the antitumor effects of temozolomide (TMZ). Migration, invasion and epithelial-mesenchymal transition (EMT) were further assessed using Transwell and western blotting assays. A co-immunoprecipitation assay was used to detect the interaction between TIGAR and protein kinase B (AKT). The results of the present study revealed that TIGAR was positively associated with poor survival and was upregulated in glioblastoma. TIGAR knockdown significantly increased oxidative stress, decreased cell proliferation and exacerbated TMZ-induced apoptosis in U-87MG cells. Additionally, TIGAR knockdown decreased migration, invasion and EMT, and treatment of TIGAR-shRNA-transfected cells with NADPH had no effect on metastasis. In addition, TIGAR promoted AKT activation and bound to AKT. In conclusion, the present study demonstrated that TIGAR may promote glioblastoma growth and progression through oxidation resistance and AKT activation.
Collapse
Affiliation(s)
- Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhengwen He
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
660
|
Jiang X, Tan J, Wen Y, Liu W, Wu S, Wang L, Wangou S, Liu D, Du C, Zhu B, Xie D, Ren C. MSI2-TGF-β/TGF-β R1/SMAD3 positive feedback regulation in glioblastoma. Cancer Chemother Pharmacol 2019; 84:415-425. [PMID: 31250154 DOI: 10.1007/s00280-019-03892-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE Glioblastoma is the most malignant glioma tumors with inevitable relapse and resistance to chemotherapy; however, the mechanisms driving chemoresistance remain to be fully elucidated. This study is to explore the molecular and cellular mechanisms involving in the chemoresistance of glioblastoma. METHODS The expression of musashi (MSI) RNA-binding protein in the tumor tissues and cells of glioblastoma was measured. The effects of MSI2 in epithelial-to-mesenchymal transition (EMT), resistance to temozolomide (TMZ), tumor cell invasion, migration, and proliferation and associated signaling were evaluated. RESULTS High MSI2 expression was observed in the glioblastoma tissues. Silencing or overexpression of MSI2 significantly affected tumor cells invasion, migration, and proliferation. Silencing of MSI2 expression significantly inhibited O6-methylguanine-DNA methyltransferase (MGMT) expression and tumor growth, and reversed resistance to TMZ in xenograft tumor models. MSI2 expression regulated EMT through activating the transcription factors Snail and the TGFβ R1/SMAD3 signaling. CONCLUSIONS Our study demonstrated a positive feedback loop of MSI2-TGFβ/SMAD3 signaling which activates the EMT and MGMT which may contribute to chemoresistance in glioblastoma. This study also highlights that MSI2 could be a new target for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Xingjun Jiang
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China.
| | - Jun Tan
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Yin Wen
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Weidong Liu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Shuyu Wu
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Lei Wang
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Siyi Wangou
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Dingyang Liu
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Can Du
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Bin Zhu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Caiping Ren
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
661
|
Mukhtar YM, Wang K, Li R, Deng W, Adu-Frimpong M, Zhang H, Zhang K, Gu C, Xu X, Yu J. Novel N-arylamide derivatives of ( S)-perillic acid (( S)-PA): in vitro and in vivo cytotoxicity and antitumor evaluation. RSC Adv 2019; 9:19973-19982. [PMID: 35514731 PMCID: PMC9065563 DOI: 10.1039/c9ra03382c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/10/2019] [Indexed: 11/21/2022] Open
Abstract
Hepatocellular carcinoma (HC) and glioblastoma (GBA) are the most commonly aggressive malignant liver and brain tumors. Based on an established method for the synthesis of amide, two novel analogues (4 and 5) of (S)-perillic acid were synthesized and their structures were affirmed using nuclear magnetic resonance spectroscopic analysis. An MTT cytotoxic assay showed that our derivatives (4 and 5) demonstrated a substantial anti-proliferative effect against HC (HepG2) and GBA (U251) cell lines. Particularly, compound 5 showed growth inhibitory (IC50) effects on U251 (IC50 = 3.10 ± 0.12 μg mL-1) and HepG2 cells (IC50 = 1.49 ± 0.43 μg mL-1), which fall within the acceptable standard recommended by the National institute of cancer (Bethesda, MD, USA) for the selection of anticancer drug candidates. Consequently, we assessed the in vivo antitumor and organ/tissue toxicity of 4, 5 and 5-fluorouracil (5-FU) in hepatoma H22-inoculated mice. The results obtained indicated remarkable tumor growth inhibition with no substantial toxicological effects on the mice and the organs/tissues in the treated groups compared well with the control.
Collapse
Affiliation(s)
- Yusif Mohammed Mukhtar
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Kaili Wang
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Ran Li
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Wenwen Deng
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
- Department of Basic and Biomedical Sciences, College of Health and Well-Being P. O. Box 9, Kintampo Ghana
| | - Huiyun Zhang
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Kangyi Zhang
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Chenlu Gu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Ximing Xu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| | - Jiangnan Yu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University 301 Xuefu Road Zhenjiang 212001 P. R. China
| |
Collapse
|
662
|
Bonafé GA, Dos Santos JS, Ziegler JV, Umezawa K, Ribeiro ML, Rocha T, Ortega MM. Growth Inhibitory Effects of Dipotassium Glycyrrhizinate in Glioblastoma Cell Lines by Targeting MicroRNAs Through the NF-κB Signaling Pathway. Front Cell Neurosci 2019; 13:216. [PMID: 31191251 PMCID: PMC6546822 DOI: 10.3389/fncel.2019.00216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
It has been shown that nuclear factor kappa-B (NF-κB) is constitutively activated in glioblastoma (GBM), suggesting that the pathway could be a therapeutic target. Glycyrrhetic acid (GA), a compound isolated from licorice (Glycyrrhiza glabra), has been shown to decrease cell viability and increases apoptosis in human cancer cell lines by NF-κB signaling pathway suppression. Dipotassium glycyrrhizinate (DPG), a dipotassium salt of GA, has anti-inflammatory properties without toxicity. The current study examined the effectiveness of DPG as an anti-tumor in U87MG and T98G GBM cell lines. Additionally, we assessed DPG as a candidate for combinational therapy in GBM with temozolomide (TMZ). Our results demonstrated that the viability of U87MG and T98G cells significantly decreased in a time- and dose-dependent manner after DPG treatment, and the apoptotic ratio of DPG-treated groups was significantly higher than that of control groups. In addition, DPG in combination with TMZ revealed synergistic effects. Furthermore, the expression of NF-κB-luciferase-reporter in transfected GBM cell lines was remarkably reduced after DPG exposure by up-regulating miR16 and miR146a, which down-regulate its target genes, IRAK2 and TRAF6. A reduced neuro-sphere formation was also observed after DPG in both GBM cells. In conclusion, DPG presented anti-tumoral effect on GBM cell lines through a decrease on proliferation and an increase on apoptosis. In addition, our data also suggest that DPG anti-tumoral effect is related to NF-κB suppression, where IRAK2- and TRAF6-mediating miR16 and miR146a, respectively, might be a potential therapeutic target of DPG.
Collapse
Affiliation(s)
- Gabriel Alves Bonafé
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| | - Jéssica Silva Dos Santos
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| | - Jussara Vaz Ziegler
- Multidisciplinary Research Laboratory, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Marcelo Lima Ribeiro
- Clinical Pharmacology and Gastroenterology Unit, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| | - Thalita Rocha
- Multidisciplinary Research Laboratory, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, Post Graduate Program in Health Science, Universidade São Francisco (USF), Bragança Paulista, Brazil
| |
Collapse
|
663
|
Basu B, Ghosh MK. Extracellular Vesicles in Glioma: From Diagnosis to Therapy. Bioessays 2019; 41:e1800245. [PMID: 31188499 DOI: 10.1002/bies.201800245] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/11/2019] [Indexed: 01/08/2023]
Abstract
Increasing evidence indicates that extracellular vesicles (EVs) secreted from tumor cells play a key role in the overall progression of the disease state. EVs such as exosomes are secreted by a wide variety of cells and transport a varied population of proteins, lipids, DNA, and RNA species within the body. Gliomas constitute a significant proportion of all primary brain tumors and majority of brain malignancies. Glioblastoma multiforme (GBM) represents grade IV glioma and is associated with very poor prognosis despite the cumulative advances in diagnostic procedures and treatment strategies. Here, the authors describe the progress in understanding the role of EVs, especially exosomes, in overall glioma progression, and how new research is unraveling the utilities of exosomes in glioma diagnostics and development of next-generation therapeutic systems. Finally, based on an understanding of the latest scientific literature, a model for the possible working of therapeutic exosomes in glioma treatment is proposed.
Collapse
Affiliation(s)
- Bhaskar Basu
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata, 700091, & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata, 700091, & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| |
Collapse
|
664
|
miR-126-3p sensitizes glioblastoma cells to temozolomide by inactivating Wnt/β-catenin signaling via targeting SOX2. Life Sci 2019; 226:98-106. [DOI: 10.1016/j.lfs.2019.04.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/27/2022]
|
665
|
Rajesh Y, Biswas A, Kumar U, Das S, Banerjee I, Banik P, Bharti R, Nayak S, Ghosh SK, Mandal M. Targeting NFE2L2, a transcription factor upstream of MMP-2: A potential therapeutic strategy for temozolomide resistant glioblastoma. Biochem Pharmacol 2019; 164:1-16. [DOI: 10.1016/j.bcp.2019.03.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/14/2019] [Indexed: 12/30/2022]
|
666
|
Witusik-Perkowska M, Zakrzewska M, Jaskolski DJ, Liberski PP, Szemraj J. Artificial microenvironment of in vitro glioblastoma cell cultures changes profile of miRNAs related to tumor drug resistance. Onco Targets Ther 2019; 12:3905-3918. [PMID: 31190889 PMCID: PMC6535444 DOI: 10.2147/ott.s190601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/09/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose: The in vitro environment can influence not only the molecular background of glioblastoma drug-resistance and treatment efficiency, but also the mechanisms and pathways of cell death. Both crucial molecular pathways and the deregulation of miRNAs are thought to participate in tumor therapy-resistance. The aim of our study is to examine the potential influence of ex vivo conditions on the expression of miRNAs engaged in the machinery of tumor-drug resistance, since in vitro models are commonly used for testing new therapeutics. Methods: Glioblastoma-derived cells, cultured under three different sets of conditions, were used as experimental models in vitro. The expression of 84 miRNAs relevant to brain tumorigenesis was evaluated by multi-miRNA profiling for initial tumors and their corresponding cultures. Finally, the expression of selected miRNAs related to temozolomide-resistance (miR-125b, miR-130a, miR-21, miR-221, miR-222, miR-31, miR-149, miR-210, miR-181a) was assessed by real-time PCR for each tumor and neoplastic cells in cultures. Results: Our results demonstrate significant discrepancies in the expression of several miRNAs between tumor cells in vivo and in vitro, with miR-130a, miR-221, miR-31, miR-21, miR-222, miR-210 being the most marked. Also differences were observed between particular models in vitro. The results of computational analysis revealed the interplay between examined miRNAs and their targets involved in processes of glioblastoma chemosensitivity, including the genes relevant to temozolomide response (MGMT, PTEN, MDM2, TP53, BBC3A). Conclusion: The artificial environment may influence the selective proliferation of cell populations carrying specific patterns of miRNAs and/or the phenotype of neoplastic cells (eg differentiation) by the action of molecular events including miRNAs. These phenomena may influence the tumor-responsiveness to particular drugs, disturbing the evaluation of their efficacy in vitro, with unpredictable results caused by the interdependency of molecular pathways.
Collapse
Affiliation(s)
| | - Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Dariusz J Jaskolski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Lodz, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
667
|
McConnell DD, Carr SB, Litofsky NS. Potential effects of nicotine on glioblastoma and chemoradiotherapy: a review. Expert Rev Neurother 2019; 19:545-555. [PMID: 31092064 DOI: 10.1080/14737175.2019.1617701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Glioblastoma multiforme (GBM) has a poor prognosis despite maximal surgical resection with subsequent multi-modal radiation and chemotherapy. Use of tobacco products following diagnosis and during the period of treatment for non-neural tumors detrimentally affects treatment and prognosis. Approximately, 16-28% of patients with glioblastoma continue to smoke after diagnosis and during treatment. The literature is sparse for information-pertaining effects of smoking and nicotine on GBM treatment and prognosis. Areas covered: This review discusses cellular pathways involved in GBM progression that might be affected by nicotine, as well as how nicotine may contribute to resistance to treatment. Similarities of GBM pathways to those in non-neural tumors are investigated for potential effects by nicotine. English language papers were identified using PubMed, Medline and Scopus databases using a combination of keywords including but not limited to the following: nicotine, vaping, tobacco, e-cigarettes, smoking, vaping AND glioblastoma or brain cancer OR/AND temozolomide, carmustine, methotrexate, procarbazine, lomustine, vincristine, and neural tumor cell lines. Expert opinion: Understanding the impact of nicotine on treatment and resistance to chemotherapeutics should allow physicians to educate their patients with GBM with evidence-based recommendations about the effects of continuing to use nicotine-containing products after diagnosis and during treatment.
Collapse
Affiliation(s)
- Diane D McConnell
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - Steven B Carr
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - N Scott Litofsky
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| |
Collapse
|
668
|
Liang H, Chen Z, Sun L. Inhibition of cyclin E1 overcomes temozolomide resistance in glioblastoma by Mcl-1 degradation. Mol Carcinog 2019; 58:1502-1511. [PMID: 31045274 DOI: 10.1002/mc.23034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is one of the major causes of brain cancer-related mortality worldwide. Temozolomide (TMZ) is an important agent against GBM. Acquired TMZ-resistance severely limits the chemotherapeutic effect and leads to poor GBM patient survival. To study the underlying mechanism of drug resistance, two TMZ resistant GBM cell lines, A172 and U87, were generated. In this study, the TMZ resistant cells have less apoptosis and cell-cycle change in response to the TMZ treatment. Western blot results revealed that cyclin E1 was upregulation in TMZ resistant cells. Inhibition or depletion of cyclin E1 re-sensitized the resistant cells to the TMZ treatment, which indicated the induction of cyclin E1 is the cause of TMZ resistance in GBM cells. Furthermore, we also found the expression of cyclin E1 stabilized the expression of Mcl-1, which contributes to the TMZ resistance in GBM cells. Finally, our in vivo xenograft data showed that the combination of flavopiridol, a cyclin E1/CDK2 inhibitor, overcomes the TMZ resistant by inducing higher apoptosis. Overall, our data provided a rationale to overcome the TMZ resistant in GBM treatment by inhibiting the cyclin E1 activity.
Collapse
Affiliation(s)
- Huaxin Liang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhuo Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Libo Sun
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
669
|
Forte I, Indovina P, Iannuzzi C, Cirillo D, Di Marzo D, Barone D, Capone F, Pentimalli F, Giordano A. Targeted therapy based on p53 reactivation reduces both glioblastoma cell growth and resistance to temozolomide. Int J Oncol 2019; 54:2189-2199. [DOI: 10.3892/ijo.2019.4788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/23/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Iris Forte
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Carmelina Iannuzzi
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Donatella Cirillo
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Domenico Di Marzo
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Daniela Barone
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Francesca Capone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Francesca Pentimalli
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
670
|
Assessment of Early Therapeutic Response to Nitroxoline in Temozolomide-Resistant Glioblastoma by Amide Proton Transfer Imaging: A Preliminary Comparative Study with Diffusion-weighted Imaging. Sci Rep 2019; 9:5585. [PMID: 30944404 PMCID: PMC6447588 DOI: 10.1038/s41598-019-42088-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/25/2019] [Indexed: 01/19/2023] Open
Abstract
Amide proton transfer (APT) imaging is a novel molecular MRI technique to detect endogenous mobile proteins and peptides through chemical exchange saturation transfer. In this preliminary study, the purpose was to evaluate the feasibility of APT imaging in monitoring the early therapeutic response to nitroxoline (NTX) in a temozolomide (TMZ)-resistant glioblastoma multiforme (GBM) mouse model, which was compared with diffusion-weighted imaging (DWI). Here, we prepared TMZ-resistant GBM mouse model (n = 12), which were treated with 100 mg/kg/day of NTX (n = 4) or TMZ (n = 4), or saline (n = 4) for 7 days for the evaluation of short-term treatment by using APT imaging and DWI sequentially. The APT signal intensities and apparent diffusion coefficient (ADC) values were calculated and compared before and after treatment. Moreover, immunohistological analysis was also employed for the correlation between APT imaging and histopathology. The association between the APT value and Ki-67 labeling index was evaluated by using simple linear regression analysis. The short-term NTX treatment resulted in significant decrease in APT value as compared to untreated and TMZ group, in which APT signals were increased. However, we did not observe significantly increased mean ADC value following short-term NTX treatment. The Ki-67 labeling index shows a correlation with APT value. APT imaging could show the earlier response to NTX treatment as compared to ADC values in a TMZ-resistant mouse model. We believe that APT imaging can be a useful imaging biomarker for the early therapeutic evaluation in GBM patients.
Collapse
|
671
|
Doan P, Musa A, Candeias NR, Emmert-Streib F, Yli-Harja O, Kandhavelu M. Alkylaminophenol Induces G1/S Phase Cell Cycle Arrest in Glioblastoma Cells Through p53 and Cyclin-Dependent Kinase Signaling Pathway. Front Pharmacol 2019; 10:330. [PMID: 31001122 PMCID: PMC6454069 DOI: 10.3389/fphar.2019.00330] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 03/19/2019] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most common type of malignant brain tumor in adults. We show here that small molecule 2-[(3,4-dihydroquinolin-1(2H)-yl)(p-tolyl)methyl]phenol (THTMP), a potential anticancer agent, increases the human glioblastoma cell death. Its mechanism of action and the interaction of selective signaling pathways remain elusive. Three structurally related phenolic compounds were tested in multiple glioma cell lines in which the potential activity of the compound, THTMP, was further validated and characterized. Upon prolonged exposer to THTMP, all glioma cell lines undergo p53 and cyclin-dependent kinase mediated cell death with the IC50 concentration of 26.5 and 75.4 μM in LN229 and Snb19, respectively. We found that THTMP strongly inhibited cell growth in a dose and in time dependent manner. THTMP treatment led to G1/S cell cycle arrest and apoptosis induction of glioma cell lines. Furthermore, we identified 3,714 genes with significant changes at the transcriptional level in response to THTMP. Further, a transcriptional analysis (RNA-seq) revealed that THTMP targeted the p53 signaling pathway specific genes causing DNA damage and cell cycle arrest at G1/S phase explained by the decrease of cyclin-dependent kinase 1, cyclin A2, cyclin E1 and E2 in glioma cells. Consistently, THTMP induced the apoptosis by regulating the expression of Bcl-2 family genes and reactive oxygen species while it also changed the expression of several anti-apoptotic genes. These observations suggest that THTMP exerts proliferation activity inhibition and pro-apoptosis effects in glioma through affecting cell cycle arrest and intrinsic apoptosis signaling. Importantly, THTMP has more potential at inhibiting GBM cell proliferation compared to TMZ, the current chemotherapy treatment administered to GBM patients; thus, we propose that THTMP may be an alternative therapeutic option for glioblastoma.
Collapse
Affiliation(s)
- Phuong Doan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tampere, Finland.,Institute of Biosciences and Medical Technology, Tampere, Finland
| | - Aliyu Musa
- Institute of Biosciences and Medical Technology, Tampere, Finland.,Predictive Medicine and Data Analytics Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tampere, Finland
| | - Nuno R Candeias
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Frank Emmert-Streib
- Institute of Biosciences and Medical Technology, Tampere, Finland.,Predictive Medicine and Data Analytics Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tampere, Finland
| | - Olli Yli-Harja
- Institute of Biosciences and Medical Technology, Tampere, Finland.,Computaional Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tampere, Finland.,Institute for Systems Biology, Seattle, WA, United States
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tampere, Finland.,Institute of Biosciences and Medical Technology, Tampere, Finland
| |
Collapse
|
672
|
Shafi A, Nguyen T, Peyvandipour A, Nguyen H, Draghici S. A Multi-Cohort and Multi-Omics Meta-Analysis Framework to Identify Network-Based Gene Signatures. Front Genet 2019; 10:159. [PMID: 30941158 PMCID: PMC6434849 DOI: 10.3389/fgene.2019.00159] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/14/2019] [Indexed: 12/20/2022] Open
Abstract
Although massive amounts of condition-specific molecular profiles are being accumulated in public repositories every day, meaningful interpretation of these data remains a major challenge. In an effort to identify the biomarkers that describe the key biological phenomena for a given condition, several approaches have been developed over the past few years. However, the majority of these approaches either (i) do not consider the known intermolecular interactions, or (ii) do not integrate molecular data of multiple types (e.g., genomics, transcriptomics, proteomics, epigenomics, etc.), and thus potentially fail to capture the true biological changes responsible for complex diseases (e.g., cancer). In addition, these approaches often ignore the heterogeneity and study bias present in independent molecular cohorts. In this manuscript, we propose a novel multi-cohort and multi-omics meta-analysis framework that overcomes all three limitations mentioned above in order to identify robust molecular subnetworks that capture the key dynamic nature of a given biological condition. Our framework integrates multiple independent gene expression studies, unmatched DNA methylation studies, and protein-protein interactions to identify methylation-driven subnetworks. We demonstrate the proposed framework by constructing subnetworks related to two complex diseases: glioblastoma and low-grade gliomas. We validate the identified subnetworks by showing their ability to predict patients' clinical outcome on multiple independent validation cohorts.
Collapse
Affiliation(s)
- Adib Shafi
- Department of Computer Science, Wayne State University, Detroit, MI, United States
| | - Tin Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV, United States
| | - Azam Peyvandipour
- Department of Computer Science, Wayne State University, Detroit, MI, United States
| | - Hung Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV, United States
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, MI, United States.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
673
|
Zembrzuska K, Ostrowski RP, Matyja E. Hyperbaric oxygen increases glioma cell sensitivity to antitumor treatment with a novel isothiourea derivative in vitro. Oncol Rep 2019; 41:2703-2716. [PMID: 30896865 PMCID: PMC6448092 DOI: 10.3892/or.2019.7064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/04/2019] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor. Tumor hypoxia is a pivotal factor responsible for the progression of this malignant glioma, and its resistance to radiation and chemotherapy. Thus, improved tumor tissue oxygenation may promote greater sensitivity to anticancer treatment. Protein kinase D1 (PKD1) protects cells from oxidative stress, and its abnormal activity serves an important role in multiple malignancies. The present study examined the effects of various oxygen conditions on the cytotoxic potential of the novel isothiourea derivate N,N′-dimethyl-S-(2,3,4,5,6-pentabromobenzyl)- isothiouronium bromide (ZKK-3) against the T98G GBM cell line. ZKK-3 was applied at concentrations of 10, 25 and 50 µM, and cells were maintained under conditions of normoxia, anoxia, hypoxia, hyperbaric oxygen (HBO), hypoxia/hypoxia and hypoxia/HBO. The proliferation and viability of neoplastic cells, and protein expression levels of hypoxia-inducible factor 1α (HIF-1α), PKD1, phosphorylated (p)PKD1 (Ser 916) and pPKD1 (Ser 744/748) kinases were evaluated. Oxygen deficiency, particularly regarding hypoxia, could diminish the cytotoxic effect of ZKK-3 at 25 and 50 µM and improve T98G cell survival compared with normoxia. HBO significantly reduced cell proliferation and increased T98G cell sensitivity to ZKK-3 when compared with normoxia. HIF-1α expression levels were increased under hypoxia compared with normoxia and decreased under HBO compared with hypoxia/hypoxia at 0, 10 and 50 µM ZKK-3, suggesting that HBO improved oxygenation of the cells. ZKK-3 exhibited inhibitory activity against pPKD1 (Ser 916) kinase; however, the examined oxygen conditions did not appear to significantly influence the expression of this phosphorylated form in cells treated with the tested compound. Regarding pPKD1 (Ser 744/748), a significant difference in expression was observed only for cells treated with 10 µM ZKK-3 and hypoxia/hypoxia compared with normoxia. However, there were significant differences in the expression levels of both phosphorylated forms of PKD1 under different oxygen conditions in the controls. In conclusion, the combination of isothiourea derivatives and hyperbaric oxygenation appears to be a promising therapeutic approach for malignant glioma treatment.
Collapse
Affiliation(s)
- Katarzyna Zembrzuska
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| | - Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| | - Ewa Matyja
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| |
Collapse
|
674
|
Gonçalves RM, Agnes JP, Delgobo M, de Souza PO, Thomé MP, Heimfarth L, Lenz G, Moreira JCF, Zanotto-Filho A. Late autophagy inhibitor chloroquine improves efficacy of the histone deacetylase inhibitor SAHA and temozolomide in gliomas. Biochem Pharmacol 2019; 163:440-450. [PMID: 30878553 DOI: 10.1016/j.bcp.2019.03.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/12/2019] [Indexed: 01/20/2023]
Abstract
Glioblastoma multiforme is the most aggressive type of primary brain tumor associated with few therapeutic opportunities and poor prognosis. In this study, we evaluated the efficacy of combining temozolomide (TMZ) with suberoylanilide hydroxamic acid (SAHA) - a specific histone deacetylases inhibitor - in glioma models in vitro and in vivo. In glioma cell lines, combined TMZ/SAHA promoted more cytotoxicity, G2/M arrest and apoptosis than either drugs alone. G2/M arrest was detected as soon as 24 h post drug exposure and preceded apoptosis, which occurred from 72 h treatment. TMZ and SAHA, alone or combined, also stimulated autophagy as evaluated by means of acridine orange staining and immunodetection of LC3I-II conversion and p62/SQSTM1 degradation. Time-course of autophagy accompanied G2/M arrest and preceded apoptosis, and blockage of late steps of autophagy with chloroquine (CQ) augmented SAHA/TMZ toxicity leading to apoptosis. In orthotopic gliomas in vivo, combined SAHA/TMZ showed better antitumor efficacy than either drugs alone, and adding CQ to the regimen improved antiglioma effects of SAHA and TMZ monotherapies without further benefit on combined SAHA/TMZ. In summary, the herein presented data suggest that autophagy acts as a protective response that impairs efficacy of SAHA and TMZ. Inhibiting autophagy termination with CQ may offer means to improve antitumor effects of SAHA and TMZ in gliomas and possibly other cancers.
Collapse
Affiliation(s)
- Rosângela Mayer Gonçalves
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jonathan Paulo Agnes
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Marina Delgobo
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Marcos P Thomé
- Departamento de Biofísica and Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luana Heimfarth
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Guido Lenz
- Departamento de Biofísica and Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Alfeu Zanotto-Filho
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
675
|
Philbrick BD, Adamson DC. Early clinical trials of Toca 511 and Toca FC show a promising novel treatment for recurrent malignant glioma. Expert Opin Investig Drugs 2019; 28:207-216. [PMID: 30676111 DOI: 10.1080/13543784.2019.1572112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Glioblastoma and anaplastic astrocytoma are two of the most aggressive and common glioma malignancies in adults. These high-grade gliomas (HGG) universally recur despite aggressive treatment modalities and have a median overall survival (mOS) of approximately 14 months from initial diagnosis. Upon recurrence, there is no standard of care and these patients have a dismal prognosis of around 9 months at time of recurrence. Areas covered: In this article, we assess the newly published phase I data of Toca 511 and Toca FC, a two-drug combination therapy for recurrent HGG (rHGG) tumors, for effectiveness and safety. Expert opinion: These early studies provide very encouraging results for Toca 511 and Toca FC in rHGG. This therapy had a response rate of 11.3% and a mOS of 11.9 months in 56 patients, an improvement compared to historical controls. Furthermore, all responders were complete responses after extended follow-up. The drug is well tolerated for most patients. Responders tended to be young and have high-performance scores prior to beginning therapy, but more studies are necessary to understand the patient profile that receives the most benefit. Randomized-controlled trials are warranted for Toca 511 and Toca FC to confirm drug efficacy.
Collapse
Affiliation(s)
- Brandon D Philbrick
- a Department of Neurosurgery , Emory University School of Medicine , Atlanta , GA , USA
| | - D Cory Adamson
- a Department of Neurosurgery , Emory University School of Medicine , Atlanta , GA , USA.,b Neurosurgery Section , Atlanta VA Medical Center , Decatur , GA , USA
| |
Collapse
|
676
|
Zhang D, Liu X, Xu X, Xu J, Yi Z, Shan B, Liu B. HPCAL1 promotes glioblastoma proliferation via activation of Wnt/β-catenin signalling pathway. J Cell Mol Med 2019; 23:3108-3117. [PMID: 30843345 PMCID: PMC6484330 DOI: 10.1111/jcmm.14083] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/22/2018] [Accepted: 11/16/2018] [Indexed: 01/11/2023] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignancy of the central nervous system with obvious aggressiveness, and is associated with poor clinical outcome. Studies have indicated that calcium ion (Ca2+) can positively regulate the initiation of malignancy with regard to GBM by modulating quiescence, proliferation, migration and maintenance. Hippocalcin like‐1 protein (HPCAL1) serves as a sensor of Ca2+. However, the understanding of HPCAL1 activity in GBM is limited. The present study revealed that the gene HPCAL1 was up‐regulated by Ca2+ in the tissues and cells of GBM. Ectopic expression of HPCAL1 promoted proliferation of cells. Exhaustion of HPCAL1 inhibited cell growth not only in vivo, but also in vitro. In addition, HPCAL1 enhanced the Wnt pathway by stimulating β‐catenin accumulation and nuclear translocation in GBM cells, while β‐catenin silencing significantly inhibited the proliferation and growth of the GBM cells. Our results showed that Ser9 phosphorylation of GSK3β was significantly decreased after HPCAL1 knockdown in GBM cells, and knockdown of the gene GSK3β in GBM cells enhanced cell proliferation and promoted transcription of the genes CCND1 and c‐Myc. Furthermore, the phosphorylation of ERK was decreased in the cells with HPCAL1 knockdown, while it was promoted via overexpression of HPCAL1. The suppression or depletion of the gene ERK decreased proliferation triggered by overexpression of HPCAL1 and impaired transcription of the genes c‐Myc and CCND1. These studies elucidate the tumour‐promoting activity of HPCAL1. They also offer an innovative therapeutic strategy focusing on the HPCAL1‐Wnt/β‐catenin axis to regulate proliferation and development of GBM.
Collapse
Affiliation(s)
- Dongming Zhang
- Department of Neurosurgery, Dongying People's Hospital, Dongying, Shandong, China
| | - Xidong Liu
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong, China
| | - Xuebin Xu
- Department of Neurosurgery, Dongying People's Hospital, Dongying, Shandong, China
| | - Jianmeng Xu
- Department of Neurosurgery, Dongying District People's Hospital, Dongying, Shandong, China
| | - Zhongjun Yi
- Department of Neurology, Dongying District People's Hospital, Dongying, Shandong, China
| | - Baochang Shan
- Department of Neurosurgery, Dongying People's Hospital, Dongying, Shandong, China
| | - Bing Liu
- Department of Neurosurgery, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
677
|
Kosgodage US, Uysal-Onganer P, MacLatchy A, Mould R, Nunn AV, Guy GW, Kraev I, Chatterton NP, Thomas EL, Inal JM, Bell JD, Lange S. Cannabidiol Affects Extracellular Vesicle Release, miR21 and miR126, and Reduces Prohibitin Protein in Glioblastoma Multiforme Cells. Transl Oncol 2019; 12:513-522. [PMID: 30597288 PMCID: PMC6314156 DOI: 10.1016/j.tranon.2018.12.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/10/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of primary malignant brain tumor in adults, with poor prognosis. Extracellular vesicles (EVs) are key-mediators for cellular communication through transfer of proteins and genetic material. Cancers, such as GBM, use EV release for drug-efflux, pro-oncogenic signaling, invasion and immunosuppression; thus the modulation of EV release and cargo is of considerable clinical relevance. As EV-inhibitors have been shown to increase sensitivity of cancer cells to chemotherapy, and we recently showed that cannabidiol (CBD) is such an EV-modulator, we investigated whether CBD affects EV profile in GBM cells in the presence and absence of temozolomide (TMZ). Compared to controls, CBD-treated cells released EVs containing lower levels of pro-oncogenic miR21 and increased levels of anti-oncogenic miR126; these effects were greater than with TMZ alone. In addition, prohibitin (PHB), a multifunctional protein with mitochondrial protective properties and chemoresistant functions, was reduced in GBM cells following 1 h CBD treatment. This data suggests that CBD may, via modulation of EVs and PHB, act as an adjunct to enhance treatment efficacy in GBM, supporting evidence for efficacy of cannabinoids in GBM.
Collapse
Affiliation(s)
- Uchini S Kosgodage
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, London, UK.
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London, UK.
| | - Amy MacLatchy
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK.
| | - Rhys Mould
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK.
| | - Alistair V Nunn
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK.
| | - Geoffrey W Guy
- GW Research, Sovereign House, Vision Park, Cambridge, CB24 9BZ, UK.
| | - Igor Kraev
- The Open University, Walton Hall, Milton Keynes, UK.
| | | | - E Louise Thomas
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK.
| | - Jameel M Inal
- Extracellular Vesicle Research Unit and Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, UK.
| | - Jimmy D Bell
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK.
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London, UK.
| |
Collapse
|
678
|
Pellosi DS, Paula LB, de Melo MT, Tedesco AC. Targeted and Synergic Glioblastoma Treatment: Multifunctional Nanoparticles Delivering Verteporfin as Adjuvant Therapy for Temozolomide Chemotherapy. Mol Pharm 2019; 16:1009-1024. [PMID: 30698450 DOI: 10.1021/acs.molpharmaceut.8b01001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite advances in cancer therapies, glioblastoma multiforme treatment remains inefficient due to the brain-blood barrier (BBB) inhibitory activity and to the low temozolomide (TMZ) chemotherapeutic selectivity. To improve therapeutic outcomes, in this work we propose two strategies, (i) photodynamic therapy (PDT) as adjuvant treatment and (ii) engineering of multifunctional theranostic/targeted nanoparticles ( m-NPs) that integrate biotin as a targeting moiety with rhodamine-B as a theranostic agent in pluronic P85/F127 copolymers. These smart m-NPs can surmount the BBB and coencapsulate multiple cargoes under optimized conditions. Overall, the present study conducts a rational m-NP design, characterization, and optimizes the formulation conditions. Confocal microscopy studies on T98-G, U87-MG, and U343 glioblastoma cells and on NIH-3T3 normal fibroblast cells show that the m-NPs and the encapsulated drugs are selectively taken up by tumor cells presenting a broad intracellular distribution. The formulations display no toxicity in the absence of light and are not toxic to healthy cells, but they exert a robust synergic action in cancer cells in the case of concomitant PDT/TMZ treatment, especially at low TMZ concentrations and higher light doses, as demonstrated by nonlinear dose-effect curves based on the Chou-Talalay method. The results evidenced different mechanisms of action related to the disjoint cell cycle phases at the optimal PDT/TMZ ratio. This effect favors synergism between the PDT and the chemotherapy with TMZ, enhances the antiproliferative effect, and overcomes cross-resistance mechanisms. These results point out that m-NP-based PDT adjuvant therapy is a promising strategy to improve TMZ-based glioblastoma multiforme treatments.
Collapse
Affiliation(s)
- Diogo S Pellosi
- Laboratory of Hybrid Materials, Department of Chemistry , Federal University of São Paulo , Diadema 04021-001 , Brazil.,Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Leonardo B Paula
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Maryanne T de Melo
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Antonio C Tedesco
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| |
Collapse
|
679
|
Tomé M, Tchorz J, Gassmann M, Bettler B. Constitutive activation of Notch2 signalling confers chemoresistance to neural stem cells via transactivation of fibroblast growth factor receptor-1. Stem Cell Res 2019; 35:101390. [PMID: 30763736 DOI: 10.1016/j.scr.2019.101390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/28/2018] [Accepted: 01/23/2019] [Indexed: 12/01/2022] Open
Abstract
Notch signalling regulates neural stem cell (NSC) proliferation, differentiation and survival for the correct development and functioning of the central nervous system. Overactive Notch2 signalling has been associated with poor prognosis of aggressive brain tumours, such as glioblastoma multiforme (GBM). We recently reported that constitutive expression of the Notch2 intracellular domain (N2ICD) enhances proliferation and gliogenesis in NSCs. Here, we investigated the mechanism by which Notch2 promotes resistance to apoptosis of NSCs to cytotoxic insults. We performed ex vivo studies using NSC cultures from transgenic mice constitutively expressing N2ICD. These NSCs expressed increased levels of pro-survival factors and lack an apoptotic response to the topoisomerase inhibitor etoposide, not showing neither mitochondrial damage nor caspase activation. Interestingly, Notch2 signalling also regulated chemoresistance of human GBM cells to etoposide. We also identified a signalling crosstalk with FGF signalling pathway involved in this resistance to apoptosis of NSCs. Aberrant Notch2 expression enhances fibroblast growth factor receptor-1 (FGFR1) activity to specifically target the AKT-GSK3 signalling pathway to block apoptosis. These results have implications for understanding molecular changes involved in both tumorigenesis and therapy resistance.
Collapse
Affiliation(s)
- Mercedes Tomé
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland.
| | - Jan Tchorz
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
680
|
Su BC, Pan CY, Chen JY. Antimicrobial Peptide TP4 Induces ROS-Mediated Necrosis by Triggering Mitochondrial Dysfunction in Wild-Type and Mutant p53 Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11020171. [PMID: 30717309 PMCID: PMC6406555 DOI: 10.3390/cancers11020171] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Antimicrobial peptide tilapia piscidin 4 (TP4) from Oreochromis niloticus exhibits potent bactericidal and anti-tumorigenic effects. In a variety of cancers, the mutation status of p53 is a decisive factor for therapeutic sensitivity. Therefore, we investigated the impact of p53 status on TP4-induced cytotoxicity in glioblastoma cell lines and the molecular mechanisms that govern cytotoxic effects. Both U87MG (wild-type/WT p53) and U251 (mutant p53) glioblastoma cell lines were sensitive to TP4-induced cytotoxicity. The necrosis inhibitors Necrostatin-1 and GSK’872 attenuated TP4-induced cytotoxicity, and TP4 treatment induced the release of cyclophilin A, a biomarker of necrosis. Moreover, TP4 induced mitochondrial hyperpolarization and dysfunction, which preceded the elevation of intracellular reactive oxygen species, DNA damage, and necrotic cell death in both U87MG and U251 glioblastoma cells. p38 was also activated by TP4, but did not contribute to cytotoxicity. SB202190, a specific p38 inhibitor, enhanced TP4-induced oxidative stress, mitochondrial dysfunction, and cytotoxicity, suggesting a protective role of p38. Furthermore, TP4-induced cytotoxicity, oxidative stress, phosphorylation of p38, and DNA damage were all attenuated by the mitochondrial-targeted reactive oxygen species (ROS) scavenger MitoTEMPO, or the reactive oxygen species scavenger N-acetyl-L-cysteine. Based on these data, we conclude that TP4 induces necrosis in both WT and mutant p53 glioblastoma cells through a mitochondrial ROS-dependent pathway.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan.
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| |
Collapse
|
681
|
Jovanović N, Mitrović T, Cvetković VJ, Tošić S, Vitorović J, Stamenković S, Nikolov V, Kostić A, Vidović N, Krstić M, Jevtović-Stoimenov T, Pavlović D. The Impact of MGMT Promoter Methylation and Temozolomide Treatment in Serbian Patients with Primary Glioblastoma. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E34. [PMID: 30717206 PMCID: PMC6409652 DOI: 10.3390/medicina55020034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 01/29/2019] [Indexed: 01/25/2023]
Abstract
Background and objective: Despite recent advances in treatment, glioblastoma (GBM) remains the most lethal and aggressive brain tumor. A continuous search for a reliable molecular marker establishes the methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) gene promoter as a key prognostic factor in primary glioblastoma. The aim of our study was to screen Serbian patients with primary glioblastoma for an MGMT promoter hypermethylation and to evaluate its associations with overall survival (OS) and sensitivity to temozolomide (TMZ) treatment. Materials and methods: A cohort of 30 Serbian primary glioblastoma patients treated with radiation therapy and chemotherapy were analyzed for MGMT promoter methylation and correlated with clinical data. Results: MGMT methylation status was determined in 25 out of 30 primary glioblastomas by methylation-specific PCR (MSP). MGMT promoter hypermethylation was detected in 12 out of 25 patients (48%). The level of MGMT promoter methylation did not correlate with patients' gender (p = 0.409), age (p = 0.536), and OS (p = 0.394). Treatment with TMZ significantly prolonged the median survival of a patient (from 5 to 15 months; p < 0.001). Conclusions: Due to a small cohort of primary GBM patients, our study is not sufficient for definitive conclusions regarding the prognostic value of MGMT methylation for the Serbian population. Our preliminary data suggest a lack of association between MGMT promoter methylation and overall survival and a significant correlation of TMZ treatment with overall survival. Further population-based studies are needed to assess the prognostic value of the MGMT promoter methylation status for patients with primary glioblastoma.
Collapse
Affiliation(s)
- Nikola Jovanović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Tatjana Mitrović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Vladimir J Cvetković
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Svetlana Tošić
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Jelena Vitorović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Slaviša Stamenković
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Vesna Nikolov
- University of Niš, Faculty of Medicine, Clinic of Neurosurgery, Clinical Center, 18000 Niš, Serbia.
| | - Aleksandar Kostić
- University of Niš, Faculty of Medicine, Clinic of Neurosurgery, Clinical Center, 18000 Niš, Serbia.
| | - Nataša Vidović
- University of Niš, Faculty of Medicine, Pathology and Pathological Anatomy Center, 18000 Niš, Serbia.
| | - Miljan Krstić
- University of Niš, Faculty of Medicine, Pathology and Pathological Anatomy Center, 18000 Niš, Serbia.
| | | | - Dušica Pavlović
- University of Niš, Faculty of Medicine, Institute of Biochemistry, 18000 Niš, Serbia.
| |
Collapse
|
682
|
Sarisozen C, Tan Y, Liu J, Bilir C, Shen L, Filipczak N, Porter TM, Torchilin VP. MDM2 antagonist-loaded targeted micelles in combination with doxorubicin: effective synergism against human glioblastoma via p53 re-activation. J Drug Target 2019; 27:624-633. [DOI: 10.1080/1061186x.2019.1570518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- C. Sarisozen
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| | - Y. Tan
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| | - J. Liu
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| | - C. Bilir
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
- Department of Medical Oncology, Sakarya University School of Medicine, Adapazarı, Turkey
| | - L. Shen
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| | - N. Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| | - T. M. Porter
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - V. P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine Northeastern University, Boston, MA, USA
| |
Collapse
|
683
|
de Moura Sperotto ND, Deves Roth C, Rodrigues-Junior VS, Ev Neves C, Reisdorfer Paula F, da Silva Dadda A, Bergo P, Freitas de Freitas T, Souza Macchi F, Moura S, Duarte de Souza AP, Campos MM, Valim Bizarro C, Santos DS, Basso LA, Machado P. Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer. J Med Chem 2019; 62:1231-1245. [DOI: 10.1021/acs.jmedchem.8b01305] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | - Fávero Reisdorfer Paula
- Laboratório de Desenvolvimento e Controle de Qualidade em Medicamentos, Universidade Federal do Pampa, 97508-000 Uruguaiana, RS, Brazil
| | | | | | | | | | - Sidnei Moura
- Laboratório de Produtos Naturais e Sintéticos, Instituto de Biotecnologia, Universidade de Caxias do Sul, 95070-560 Caxias do Sul, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
684
|
Peptidylarginine Deiminases Post-Translationally Deiminate Prohibitin and Modulate Extracellular Vesicle Release and MicroRNAs in Glioblastoma Multiforme. Int J Mol Sci 2018; 20:ijms20010103. [PMID: 30597867 PMCID: PMC6337164 DOI: 10.3390/ijms20010103] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of adult primary malignant brain tumour with poor prognosis. Extracellular vesicles (EVs) are a key-mediator through which GBM cells promote a pro-oncogenic microenvironment. Peptidylarginine deiminases (PADs), which catalyze the post-translational protein deimination of target proteins, are implicated in cancer, including via EV modulation. Pan-PAD inhibitor Cl-amidine affected EV release from GBM cells, and EV related microRNA cargo, with reduced pro-oncogenic microRNA21 and increased anti-oncogenic microRNA126, also in combinatory treatment with the chemotherapeutic agent temozolomide (TMZ). The GBM cell lines under study, LN18 and LN229, differed in PAD2, PAD3 and PAD4 isozyme expression. Various cytoskeletal, nuclear and mitochondrial proteins were identified to be deiminated in GBM, including prohibitin (PHB), a key protein in mitochondrial integrity and also involved in chemo-resistance. Post-translational deimination of PHB, and PHB protein levels, were reduced after 1 h treatment with pan-PAD inhibitor Cl-amidine in GBM cells. Histone H3 deimination was also reduced following Cl-amidine treatment. Multifaceted roles for PADs on EV-mediated pathways, as well as deimination of mitochondrial, nuclear and invadopodia related proteins, highlight PADs as novel targets for modulating GBM tumour communication.
Collapse
|
685
|
Llaguno-Munive M, Romero-Piña M, Serrano-Bello J, Medina LA, Uribe-Uribe N, Salazar AM, Rodríguez-Dorantes M, Garcia-Lopez P. Mifepristone Overcomes Tumor Resistance to Temozolomide Associated with DNA Damage Repair and Apoptosis in an Orthotopic Model of Glioblastoma. Cancers (Basel) 2018; 11:cancers11010016. [PMID: 30583528 PMCID: PMC6356343 DOI: 10.3390/cancers11010016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
The standard treatment for glioblastoma multiforme (GBM) is surgery followed by chemo/radiotherapy. A major limitation on patient improvement is the high resistance of tumors to drug treatment, likely responsible for their subsequent recurrence and rapid progression. Therefore, alternatives to the standard therapy are necessary. The aim of the present study was to evaluate whether mifepristone, an antihormonal agent, has a synergistic effect with temozolomide (used in standard therapy for gliomas). Whereas the mechanism of temozolomide involves damage to tumor DNA leading to apoptosis, tumor resistance is associated with DNA damage repair through the O6-methylguanine-DNA-methyltransferase (MGMT) enzyme. Temozolomide/mifepristone treatment, herein examined in Wistar rats after orthotopically implanting C6 glioma cells, markedly reduced proliferation. This was evidenced by a decreased level of the following parameters: a proliferation marker (Ki-67), a tumor growth marker (18F-fluorothymidine uptake, determined by PET/CT images), and the MGMT enzyme. Increased apoptosis was detected by the relative expression of related proteins, (e.g. Bcl-2 (B-cell lymphoma 2), Bax (bcl-2-like protein 4) and caspase-3). Thus, greater apoptosis of tumor cells caused by their diminished capacity to repair DNA probably contributed significantly to the enhanced activity of temozolomide. The results suggest that mifepristone could possibly act as a chemo-sensitizing agent for temozolomide during chemotherapy for GBM.
Collapse
Affiliation(s)
- Monserrat Llaguno-Munive
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico.
- Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico.
| | - Mario Romero-Piña
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico.
| | - Janeth Serrano-Bello
- Facultad de Odontología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico.
| | - Luis A Medina
- Instituto de Física, Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Ciudad de México, 14080, Mexico.
| | - Norma Uribe-Uribe
- Instituto Nacional de Ciencias Médicas y de la Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico.
| | - Ana Maria Salazar
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico.
| | | | - Patricia Garcia-Lopez
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico.
| |
Collapse
|
686
|
A New Patient-Derived Metastatic Glioblastoma Cell Line: Characterisation and Response to Sodium Selenite Anticancer Agent. Cancers (Basel) 2018; 11:cancers11010012. [PMID: 30583471 PMCID: PMC6356827 DOI: 10.3390/cancers11010012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiform (GBM) tumors are very heterogeneous, organized in a hierarchical pattern, including cancer stem cells (CSC), and are responsible for development, maintenance, and cancer relapse. Therefore, it is relevant to establish new GBM cell lines with CSC characteristics to develop new treatments. A new human GBM cell line, named R2J, was established from the cerebro-spinal fluid (CSF) of a patient affected by GBM with leptomeningeal metastasis. R2J cells exhibits an abnormal karyotype and form self-renewable spheres in a serum-free medium. Original tumor, R2J, cultured in monolayer (2D) and in spheres showed a persistence expression of CD44, CD56 (except in monolayer), EGFR, Ki67, Nestin, and vimentin. The R2J cell line is tumorigenic and possesses CSC properties. We tested in vitro the anticancer effects of sodium selenite (SS) compared to temozolomide TMZ. SS was absorbed by R2J cells, was cytotoxic, induced an oxidative stress, and arrested cell growth in G2M before inducing both necrosis and apoptosis via caspase-3. SS also modified dimethyl-histone-3-lysine-9 (H3K9m2) levels and decreased histone deacetylase (HDAC) activity, suggesting anti-invasiveness potential. This study highlights the value of this new GBM cell line for preclinical modeling of clinically relevant, patient specific GBM and opens a therapeutic window to test SS to target resistant and recurrent GBM.
Collapse
|
687
|
Tapeinos C, Marino A, Battaglini M, Migliorin S, Brescia R, Scarpellini A, De Julián Fernández C, Prato M, Drago F, Ciofani G. Stimuli-responsive lipid-based magnetic nanovectors increase apoptosis in glioblastoma cells through synergic intracellular hyperthermia and chemotherapy. NANOSCALE 2018; 11:72-88. [PMID: 30357214 PMCID: PMC6336008 DOI: 10.1039/c8nr05520c] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/01/2018] [Indexed: 05/20/2023]
Abstract
In this study, taking into consideration the limitations of current treatments of glioblastoma multiforme, we fabricated a biomimetic lipid-based magnetic nanovector with a good loading capacity and a sustained release profile of the encapsulated chemotherapeutic drug, temozolomide. These nanostructures demonstrated an enhanced release after exposure to an alternating magnetic field, and a complete release of the encapsulated drug after the synergic effect of low pH (4.5), increased concentration of hydrogen peroxide (50 μM), and increased temperature due to the applied magnetic field. In addition, these nanovectors presented excellent specific absorption rate values (up to 1345 W g-1) considering the size of the magnetic component, rendering them suitable as potential hyperthermia agents. The presented nanovectors were progressively internalized in U-87 MG cells and in their acidic compartments (i.e., lysosomes and late endosomes) without affecting the viability of the cells, and their ability to cross the blood-brain barrier was preliminarily investigated using an in vitro brain endothelial cell-model. When stimulated with alternating magnetic fields (20 mT, 750 kHz), the nanovectors demonstrated their ability to induce mild hyperthermia (43 °C) and strong anticancer effects against U-87 MG cells (scarce survival of cells characterized by low proliferation rates and high apoptosis levels). The optimal anticancer effects resulted from the synergic combination of hyperthermia chronic stimulation and the controlled temozolomide release, highlighting the potential of the proposed drug-loaded lipid magnetic nanovectors for treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Christos Tapeinos
- Smart Bio-Interfaces
, Istituto Italiano di Tecnologia
,
Pontedera (Pisa)
, 56025 Italy
.
;
;
| | - Attilio Marino
- Smart Bio-Interfaces
, Istituto Italiano di Tecnologia
,
Pontedera (Pisa)
, 56025 Italy
.
;
;
| | - Matteo Battaglini
- Smart Bio-Interfaces
, Istituto Italiano di Tecnologia
,
Pontedera (Pisa)
, 56025 Italy
.
;
;
- The Biorobotics Institute
, Scuola Superiore Sant'Anna
,
Pontedera (Pisa)
, 56025 Italy
| | - Simone Migliorin
- Department of Mechanical and Aerospace Engineering
, Politecnico di Torino
,
Torino
, 10129 Italy
| | - Rosaria Brescia
- Electron Microscopy Facility
, Istituto Italiano di Tecnologia
,
Genova
, 16163 Italy
| | - Alice Scarpellini
- Electron Microscopy Facility
, Istituto Italiano di Tecnologia
,
Genova
, 16163 Italy
| | - César De Julián Fernández
- Istituto dei Materiali per l'Elettronica e il Magnetismo
, Consiglio Nazionale delle Ricerche – CNR
,
Parma
, 43124 Italy
| | - Mirko Prato
- Materials Characterization Facility
, Istituto Italiano di Tecnologia
,
Genova
, 16163 Italy
| | - Filippo Drago
- Nanochemistry Department
, Istituto Italiano di Tecnologia
,
Genova
, 16163 Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces
, Istituto Italiano di Tecnologia
,
Pontedera (Pisa)
, 56025 Italy
.
;
;
- Department of Mechanical and Aerospace Engineering
, Politecnico di Torino
,
Torino
, 10129 Italy
| |
Collapse
|
688
|
Drug and disease signature integration identifies synergistic combinations in glioblastoma. Nat Commun 2018; 9:5315. [PMID: 30552330 PMCID: PMC6294341 DOI: 10.1038/s41467-018-07659-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary adult brain tumor. Despite extensive efforts, the median survival for GBM patients is approximately 14 months. GBM therapy could benefit greatly from patient-specific targeted therapies that maximize treatment efficacy. Here we report a platform termed SynergySeq to identify drug combinations for the treatment of GBM by integrating information from The Cancer Genome Atlas (TCGA) and the Library of Integrated Network-Based Cellular Signatures (LINCS). We identify differentially expressed genes in GBM samples and devise a consensus gene expression signature for each compound using LINCS L1000 transcriptional profiling data. The SynergySeq platform computes disease discordance and drug concordance to identify combinations of FDA-approved drugs that induce a synergistic response in GBM. Collectively, our studies demonstrate that combining disease-specific gene expression signatures with LINCS small molecule perturbagen-response signatures can identify preclinical combinations for GBM, which can potentially be tested in humans. Inherent or acquired resistance to treatment of glioblastoma (GBM) is nearly universal. Here, the authors introduce a platform to identify synergistic drug combinations for patient-specific treatment of GBM based on gene expression signatures and small molecule perturbation-response profiles.
Collapse
|
689
|
Kapçak E, Şatana-Kara EH. Development and Full Validation of a Stability-indicating HPLC Method for the Determination of the Anticancer Drug Temozolomide in Pharmaceutical Form. Turk J Pharm Sci 2018; 15:271-277. [PMID: 32454670 DOI: 10.4274/tjps.43265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/07/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVES In the present study, an accurate, precise and simple method has been developed for the determination of TMZ in its pharmaceutical form by using HPLC. MATERIALS AND METHODS An HPLC method with a DAD was validated according to ICH guidelines. A C18 column (150x4.6 mm. i.d., 5 µm particle size) and an aqueous acetate buffer (0.02 M)-acetonitrile (90:10, v/v) (pH 4.5) as a mobile phase were used. RESULTS The linear range and LOD value were 5-100 µg/mL and 0.02 µg/mL, respectively. The accuracy of the method was determined using a recovery test and found as 98.8-100.3%. In addition, forced degradation studies of the drug were also performed in bulk drug samples to demonstrate the specificity and stability-indicating. Degradation studies under acidic, basic, oxidative, and thermal degradation conditions were applied. CONCLUSION The proposed method could be applied successfully for the determination and identification of the degradation of the drug.
Collapse
Affiliation(s)
- Evin Kapçak
- Gazi University, Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, Turkey
| | | |
Collapse
|
690
|
Ganguly D, Sims M, Cai C, Fan M, Pfeffer LM. Chromatin Remodeling Factor BRG1 Regulates Stemness and Chemosensitivity of Glioma Initiating Cells. Stem Cells 2018; 36:1804-1815. [PMID: 30171737 DOI: 10.1002/stem.2909] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/23/2018] [Accepted: 08/18/2018] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor that is refractory to existing therapeutic regimens, which reflects the presence of stem-like cells, termed glioma-initiating cells (GICs). The complex interactions between different signaling pathways and epigenetic regulation of key genes may be critical in the maintaining GICs in their stem-like state. Although several signaling pathways have been identified as being dysregulated in GBM, the prognosis of GBM patients remains miserable despite improvements in targeted therapies. In this report, we identified that BRG1, the catalytic subunit of the SWI/SNF chromatin remodeling complex, plays a fundamental role in maintaining GICs in their stem-like state. In addition, we identified a novel mechanism by which BRG1 regulates glycolysis genes critical for GICs. BRG1 downregulates the expression of TXNIP, a negative regulator of glycolysis. BRG1 knockdown also triggered the STAT3 pathway, which led to TXNIP activation. We further identified that TXNIP is an STAT3-regulated gene. Moreover, BRG1 suppressed the expression of interferon-stimulated genes, which are negatively regulated by STAT3 and regulate tumorigenesis. We further demonstrate that BRG1 plays a critical role in the drug resistance of GICs and in GIC-induced tumorigenesis. By genetic and pharmacological means, we found that inhibiting BRG1 can sensitize GICs to chemotherapeutic drugs, temozolomide and carmustine. Our studies suggest that BRG1 may be a novel therapeutic target in GBM. The identification of the critical role that BRG1 plays in GIC stemness and chemosensitivity will inform the development of better targeted therapies in GBM and possibly other cancers. Stem Cells 2018;36:1806-12.
Collapse
Affiliation(s)
- Debolina Ganguly
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Michelle Sims
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Chun Cai
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Meiyun Fan
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
691
|
Ward S, Skinner M, Saha B, Emrick T. Polymer-Temozolomide Conjugates as Therapeutics for Treating Glioblastoma. Mol Pharm 2018; 15:5263-5276. [PMID: 30354145 PMCID: PMC6220362 DOI: 10.1021/acs.molpharmaceut.8b00766] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/20/2023]
Abstract
A series of polymer-drug conjugates based on 2-methacryloyloxyethyl phosphorylcholine (MPC) was prepared with the glioblastoma drug temozolomide (TMZ) as pendent groups. Random and block copolymers were synthesized by reversible addition-fragmentation chain-transfer (RAFT) polymerization using a TMZ-containing methacrylate monomer. The solution properties of the polyMPC-TMZ copolymers were investigated by dynamic light scattering and transmission electron microscopy, revealing well-defined nanostructures from the block copolymers. Conjugation of TMZ to polyMPC enhanced drug stability, with decomposition half-life values ranging from 2- to 19-times longer than that of free TMZ. The cytotoxicity of polyMPC-TMZ was evaluated in both chemosensitive (U87MG) and chemoresistant (T98G) glioblastoma cell lines. Furthermore, the polyMPC-TMZ platform was expanded considerably by the preparation of redox-sensitive polyMPC-TMZ copolymers utilizing disulfides as the polymer-to-drug linker.
Collapse
Affiliation(s)
| | | | - Banishree Saha
- Polymer Science and Engineering Department, University of Massachusetts, 120 Governors Drive, Amherst, Massachusetts 01003, United States
| | - Todd Emrick
- Polymer Science and Engineering Department, University of Massachusetts, 120 Governors Drive, Amherst, Massachusetts 01003, United States
| |
Collapse
|
692
|
Ciechomska IA, Marciniak MP, Jackl J, Kaminska B. Pre-treatment or Post-treatment of Human Glioma Cells With BIX01294, the Inhibitor of Histone Methyltransferase G9a, Sensitizes Cells to Temozolomide. Front Pharmacol 2018; 9:1271. [PMID: 30450051 PMCID: PMC6224489 DOI: 10.3389/fphar.2018.01271] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is a malignant, primary brain tumor, highly resistant to conventional therapies. Temozolomide (TMZ) is a first line therapeutic agent in GBM patients, however, survival of such patients is poor. High level of DNA repair protein, O6-methylguanine-DNA-methyltransferase (MGMT) and occurrence of glioma stem-like cells contribute to GBM resistance to the drug. Here, we explored a possibility of epigenetic reprograming of glioma cells to increase sensitivity to TMZ and restore apoptosis competence. We combined TMZ treatment with BIX01294, an inhibitor of histone methyltransferase G9a, known to be involved in cancerogenesis. Two treatment combinations were tested: BIX01294 was administered to human LN18 and U251 glioma cell cultures 48 h before TMZ or 48 h after TMZ treatment. Despite their different status of the MGMT gene promoter, there was no correlation with the response to TMZ. The analyses of cell viability, appearance of apoptotic alterations in morphology of cells and nuclei, and markers of apoptosis, such as levels of cleaved caspase 3, caspase 7 and PARP, revealed that both pre-treatment and post-treatment with BIX01294 sensitize glioma cells to TMZ. The additive effect was stronger in LN18 cells. Moreover, BIX01294 enhanced the cytotoxic effect of TMZ on glioma stem-like cells, although it was not associated with modulation of the pluripotency markers (NANOG, SOX2, CD133) expression or methylation of NANOG and SOX2 gene promoters. Accordingly, knockdown of methyltransferase G9a augments TMZ-induced cell death in LN18 cells. We found the significant increases of the LC3-II levels in LN18 cells treated with BIX01294 alone and with drug combination that suggests involvement of autophagy in enhancement of anti-tumor effect of TMZ. Treatment with BIX01294 did not affect methylation of the MGMT gene promoter. Altogether, our results suggest that G9a is a potential therapeutic target in malignant glioma and the treatment with the G9a inhibitor reprograms glioma cells and glioma stem-like cells to increase sensitivity to TMZ and restore apoptosis competence.
Collapse
Affiliation(s)
- Iwona Anna Ciechomska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marta Patrycja Marciniak
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Judyta Jackl
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
693
|
Kadari A, Pooja D, Gora RH, Gudem S, Kolapalli VRM, Kulhari H, Sistla R. Design of multifunctional peptide collaborated and docetaxel loaded lipid nanoparticles for antiglioma therapy. Eur J Pharm Biopharm 2018; 132:168-179. [DOI: 10.1016/j.ejpb.2018.09.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022]
|
694
|
Akay M, Hite J, Avci NG, Fan Y, Akay Y, Lu G, Zhu JJ. Drug Screening of Human GBM Spheroids in Brain Cancer Chip. Sci Rep 2018; 8:15423. [PMID: 30337660 PMCID: PMC6194126 DOI: 10.1038/s41598-018-33641-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), an extremely invasive and high-grade (grade IV) glioma, is the most common and aggressive form of brain cancer. It has a poor prognosis, with a median overall survival of only 11 months in the general GBM population and 14.6 to 21 months in clinical trial participants with standard GBM therapies, including maximum safe craniotomy, adjuvant radiation, and chemotherapies. Therefore, new approaches for developing effective treatments, such as a tool for assessing tumor cell drug response before drug treatments are administered, are urgently needed to improve patient survival. To address this issue, we developed an improved brain cancer chip with a diffusion prevention mechanism that blocks drugs crossing from one channel to another. In the current study, we demonstrate that the chip has the ability to culture 3D spheroids from patient tumor specimen-derived GBM cells obtained from three GBM patients. Two clinical drugs used to treat GBM, temozolomide (TMZ) and bevacizumab (Avastin, BEV), were applied and a range of relative concentrations was generated by the microfluidic channels in the brain cancer chip. The results showed that TMZ works more effectively when used in combination with BEV compared to TMZ alone. We believe that this low-cost brain cancer chip could be further developed to generate optimal combination of chemotherapy drugs tailored to individual GBM patients.
Collapse
Affiliation(s)
- Metin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA.
| | - John Hite
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Naze Gul Avci
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yantao Fan
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yasemin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Guangrong Lu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| | - Jay-Jiguang Zhu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| |
Collapse
|
695
|
JIAPAER S, FURUTA T, TANAKA S, KITABAYASHI T, NAKADA M. Potential Strategies Overcoming the Temozolomide Resistance for Glioblastoma. Neurol Med Chir (Tokyo) 2018; 58:405-421. [PMID: 30249919 PMCID: PMC6186761 DOI: 10.2176/nmc.ra.2018-0141] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a highly malignant type of primary brain tumor with a high mortality rate. Although the current standard therapy consists of surgery followed by radiation and temozolomide (TMZ), chemotherapy can extend patient's post-operative survival but most cases eventually demonstrate resistance to TMZ. O6-methylguanine-DNA methyltransferase (MGMT) repairs the main cytotoxic lesion, as O6-methylguanine, generated by TMZ, can be the main mechanism of the drug resistance. In addition, mismatch repair and BER also contribute to TMZ resistance. TMZ treatment can induce self-protective autophagy, a mechanism by which tumor cells resist TMZ treatment. Emerging evidence also demonstrated that a small population of cells expressing stem cell markers, also identified as GBM stem cells (GSCs), contributes to drug resistance and tumor recurrence owing to their ability for self-renewal and invasion into neighboring tissue. Some molecules maintain stem cell properties. Other molecules or signaling pathways regulate stemness and influence MGMT activity, making these GCSs attractive therapeutic targets. Treatments targeting these molecules and pathways result in suppression of GSCs stemness and, in highly resistant cases, a decrease in MGMT activity. Recently, some novel therapeutic strategies, targeted molecules, immunotherapies, and microRNAs have provided new potential treatments for highly resistant GBM cases. In this review, we summarize the current knowledge of different resistance mechanisms, novel strategies for enhancing the effect of TMZ, and emerging therapeutic approaches to eliminate GSCs, all with the aim to produce a successful GBM treatment and discuss future directions for basic and clinical research to achieve this end.
Collapse
Affiliation(s)
| | - Takuya FURUTA
- Department of Pathology, Kurume University, Kurume, Fukuoka, Japan
| | - Shingo TANAKA
- Department of Neurosurgery, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | - Mitsutoshi NAKADA
- Department of Neurosurgery, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
696
|
Da Ros M, Iorio AL, De Gregorio V, Fantappiè O, Laffi G, de Martino M, Pisano C, Genitori L, Sardi I. Aldoxorubicin and Temozolomide combination in a xenograft mice model of human glioblastoma. Oncotarget 2018; 9:34935-34944. [PMID: 30405885 PMCID: PMC6201851 DOI: 10.18632/oncotarget.26183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/15/2018] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is still an incurable disease. The front-line Temozolomide (TMZ)-based therapy suffers from poor efficacy, underlining the need of new therapies. Preclinically, Aldoxorubicin (Aldox), a novel prodrug of Doxorubicin (Dox), has been successfully tested against GBM, encouraging the study of its association with other agents. For the first time, we evaluated the effectiveness of Aldox combined to TMZ in preclinical models of GBM. Our in vitro results demonstrated that the anti–glioma effect of Aldox was more marked than TMZ and their combination increased the killing effect of the anthracycline in TMZ-resistant GBM cells. Moreover, unlike Dox, Aldox was able to accumulate in P-glycoprotein (P-gp)-overexpressed cells due to a negative regulation of the P-gp function. We also compared efficacy and safety of weekly administrations of Aldox (16 mg/kg), with or without TMZ (0.9 mg/kg, daily injections), in the U87 xenograft mouse model. Aldox therapy induced a moderate tumor volume inhibition (TVI) and an increased survival rate (+12.5% vs vehicle). On the other hand, when combined to TMZ, Aldox caused a significant TVI (P=0.0175 vs vehicle) and delayed the mortality during the experimental period, although TVI and endpoint survival percentage (+37.5% vs vehicle) were not significantly different from TMZ alone. Our preliminary data showed that Aldox exerts anti–glioma effects in vitro and in vivo. It also enhances its antitumor activity when combined with TMZ, resulting in a superior efficacy compared to the single agents, without adverse side effects.
Collapse
Affiliation(s)
- Martina Da Ros
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Anna Lisa Iorio
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Veronica De Gregorio
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Ornella Fantappiè
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giacomo Laffi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maurizio de Martino
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | | | - Lorenzo Genitori
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| |
Collapse
|
697
|
Lam P, Lin R, Steinmetz NF. Delivery of mitoxantrone using a plant virus-based nanoparticle for the treatment of glioblastomas. J Mater Chem B 2018; 6:5888-5895. [PMID: 30923616 PMCID: PMC6433411 DOI: 10.1039/c8tb01191e] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitoxatrone (MTO), an antineoplastic chemotherapeutic, has potent activity against the most common and agressive type of primary brain tumor, glioblastoma multiforme (GBM). However, its poor penetration through the blood brain barrier, and cardiotoxic side effects from systemic delivery limit its effectiveness for clinical treatment. To address these limitations, we utilize a plant virus-based nanoparticle, cowpea mosaic virus (CPMV), to deliver MTO to treat GBM. In this work, we loaded MTO into the interior cavity of CPMV (CPMV-MTO) through diffusion through its pores. We report the uptake of CPMV-MTO in glioma cells and demonstrate its cytotoxic effects in vitro as a solo therapy, and in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). These results reveal the potential for this plant virus-based nanoparticle platform for the treatment of GBM.
Collapse
Affiliation(s)
- Patricia Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Richard Lin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
698
|
Zhang I, Beus M, Stochaj U, Le PU, Zorc B, Rajić Z, Petrecca K, Maysinger D. Inhibition of glioblastoma cell proliferation, invasion, and mechanism of action of a novel hydroxamic acid hybrid molecule. Cell Death Discov 2018; 4:41. [PMID: 30302275 PMCID: PMC6158288 DOI: 10.1038/s41420-018-0103-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/10/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme is one of the most aggressive brain tumors and current therapies with temozolomide or suberoylanilide hydroxamic acid (SAHA, vorinostat) show considerable limitations. SAHA is a histone deacetylase (HDAC) inhibitor that can cause undesirable side effects due to the lack of selectivity. We show here properties of a novel hybrid molecule, sahaquine, which selectively inhibits cytoplasmic HDAC6 at nanomolar concentrations without markedly suppressing class I HDACs. Inhibition of HDAC6 leads to significant α-tubulin acetylation, thereby impairing cytoskeletal organization in glioblastoma cells. The primaquine moiety of sahaquine reduced the activity of P-glycoprotein, which contributes to glioblastoma multiforme drug resistance. We propose the mechanism of action of sahaquine to implicate HDAC6 inhibition together with suppression of epidermal growth factor receptor and downstream kinase activity, which are prominent therapeutic targets in glioblastoma multiforme. Sahaquine significantly reduces the viability and invasiveness of glioblastoma tumoroids, as well as brain tumor stem cells, which are key to tumor survival and recurrence. These effects are augmented with the combination of sahaquine with temozolomide, the natural compound quercetin or buthionine sulfoximine, an inhibitor of glutathione biosynthesis. Thus, a combination of agents disrupting glioblastoma and brain tumor stem cell homeostasis provides an effective anti–cancer intervention.
Collapse
Affiliation(s)
- Issan Zhang
- 1Department of Pharmacology and Therapeutics, McGill University, Montreal, QC Canada
| | - Maja Beus
- 1Department of Pharmacology and Therapeutics, McGill University, Montreal, QC Canada.,2Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Ursula Stochaj
- 3Department of Physiology, McGill University, Montreal, QC Canada
| | - Phuong Uyen Le
- 4Brain Tumour Research Centre, Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada
| | - Branka Zorc
- 2Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Zrinka Rajić
- 2Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Kevin Petrecca
- 4Brain Tumour Research Centre, Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada
| | - Dusica Maysinger
- 1Department of Pharmacology and Therapeutics, McGill University, Montreal, QC Canada
| |
Collapse
|
699
|
Vecera M, Sana J, Lipina R, Smrcka M, Slaby O. Long Non-Coding RNAs in Gliomas: From Molecular Pathology to Diagnostic Biomarkers and Therapeutic Targets. Int J Mol Sci 2018; 19:ijms19092754. [PMID: 30217088 PMCID: PMC6163683 DOI: 10.3390/ijms19092754] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the most common malignancies of the central nervous system. Because of tumor localization and the biological behavior of tumor cells, gliomas are characterized by very poor prognosis. Despite significant efforts that have gone into glioma research in recent years, the therapeutic efficacy of available treatment options is still limited, and only a few clinically usable diagnostic biomarkers are available. More and more studies suggest non-coding RNAs to be promising diagnostic biomarkers and therapeutic targets in many cancers, including gliomas. One of the largest groups of these molecules is long non-coding RNAs (lncRNAs). LncRNAs show promising potential because of their unique tissue expression patterns and regulatory functions in cancer cells. Understanding the role of lncRNAs in gliomas may lead to discovery of the novel molecular mechanisms behind glioma biological features. It may also enable development of new solutions to overcome the greatest obstacles in therapy of glioma patients. In this review, we summarize the current knowledge about lncRNAs and their involvement in the molecular pathology of gliomas. A conclusion follows that these RNAs show great potential to serve as powerful diagnostic, prognostic, and predictive biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Marek Vecera
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
| | - Jiri Sana
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Radim Lipina
- Department of Neurosurgery, University Hospital Ostrava, 70852 Ostrava, Czech Republic.
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| |
Collapse
|
700
|
Beberok A, Rzepka Z, Respondek M, Rok J, Sierotowicz D, Wrześniok D. GSH depletion, mitochondrial membrane breakdown, caspase-3/7 activation and DNA fragmentation in U87MG glioblastoma cells: New insight into the mechanism of cytotoxicity induced by fluoroquinolones. Eur J Pharmacol 2018; 835:94-107. [PMID: 30086267 DOI: 10.1016/j.ejphar.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022]
Abstract
Fluoroquinolones are a known synthetic group of antibiotics that have been the subject of many research interests. This class of antibiotics was shown to be cytotoxic towards various cancer cell lines, thus representing a potentially important source of new anticancer agents. The present study was designed to examine the effect of ciprofloxacin and moxifloxacin on cell viability, redox balance and apoptosis in U87MG glioblastoma cells. Herein, we found that both fluoroquinolones decrease the viability and exert an anti-proliferative effect on U87MG cells. The EC50 values were found to be as 0.75 µmol/ml, 0.57 µmol/ml, 0.53 µmol/ml for ciprofloxacin and 24, 48, 72 h incubation time, respectively, and 0.48 µmol/ml, 0.22 µmol/ml, 0.15 µmol/ml for moxifloxacin and 24, 48, 72 h incubation time, respectively. Ciprofloxacin and moxifloxacin have also induced the intracellular GSH depletion and apoptosis as shown by externalization of phosphatidylserine, caspase-3/7 activation, S and sub-G1 cell cycle arrest, nuclear morphological changes induction and DNA fragmentation. The mechanism of apoptosis was related to the loss of mitochondrial membrane potential suggesting activation of the intrinsic mitochondrial pathway. This is the first study that may provide the basis for understanding potential cellular and molecular mechanism underlying ciprofloxacin and moxifloxacin cytotoxic and pro-apoptotic effect towards U87MG glioblastoma cells, suggesting that these fluoroquinolone derivatives may have value for the development as anti-glioma agents.
Collapse
Affiliation(s)
- Artur Beberok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland.
| | - Zuzanna Rzepka
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Michalina Respondek
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Jakub Rok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Daniel Sierotowicz
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| |
Collapse
|