1
|
Deciphering of Adult Glioma Vulnerabilities through Expression Pattern Analysis of GABA, Glutamate and Calcium Neurotransmitter Genes. J Pers Med 2022; 12:jpm12040633. [PMID: 35455749 PMCID: PMC9030730 DOI: 10.3390/jpm12040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Adult infiltrating gliomas are highly aggressive tumors of the central nervous system with a dismal prognosis despite intensive multimodal therapy (chemotherapy and/or radiotherapy). In this study, we studied the expression, methylation and interacting miRNA profiles of GABA-, glutamate- and calcium-related genes in 661 adult infiltrating gliomas available through the TCGA database. Neurotransmitter-based unsupervised clustering identified three established glioma molecular subgroups that parallel major World Health Organization glioma subclasses (IDH-wildtype astrocytomas, IDH-mutant astrocytomas, IDH-mutant oligodendroglioma). In addition, this analysis also defined a novel, neurotransmitter-related glioma subgroup (NT-1), mostly comprised of IDH-mutated gliomas and characterized by the overexpression of neurotransmitter-related genes. Lower expression of neurotransmission-related genes was correlated with increased aggressivity in hypomethylated IDH-wildtype tumors. There were also significant differences in the composition of the tumor inflammatory microenvironment between neurotransmission-based tumor categories, with lower estimated pools of M2-phenotype macrophages in NT-1 gliomas. This multi-omics analysis of the neurotransmission expression landscape of TCGA gliomas—which highlights the existence of neurotransmission-based glioma categories with different expression, epigenetic and inflammatory profiles—supports the existence of operational neurotransmitter signaling pathways in adult gliomas. These findings could shed new light on potential vulnerabilities to exploit in future glioma-targeting drug therapies.
Collapse
|
2
|
Wei H, Ding C, Zhuang H, Hu W. TRIM47 Promotes the Development of Glioma by Ubiquitination and Degradation of FOXO1. Onco Targets Ther 2021; 13:13401-13411. [PMID: 33408486 PMCID: PMC7781021 DOI: 10.2147/ott.s264459] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/08/2020] [Indexed: 01/26/2023] Open
Abstract
Objective To investigate the effect of TRIM47 on glioma cells and further explore its underlying molecular mechanisms. Methods Mouse xenograft model was used in this study. The mRNA expression of TRIM47 was detected by qRT-PCR. The cell viability and proliferation activity was detected by MTT assay and colony formation assay. The migration and invasion of glioma cells were determined by Transwell assay. The protein levels of TRIM47, FOXO1, CyclinD1, C-myc, MMP-2 and TIMP-1 were assessed by Western-blotting. The interaction between TRIM47 and FOXO1 was measured by Co-immunoprecipitation (Co-IP) assay. Results In glioma tissues and cells, TRIM47 was significantly up-regulated. Silencing the expression of TRIM47 inhibited the cell viability and proliferation of cells A172 and U251, as well as their ability to invade and migrate. Among them, the expression levels of C-myc and CyclinD1 also decreased, and MMP-2 was down-regulated and TIMP-1 was up-regulated. Similarly, in vivo model, tumor volume and weight also decreased after TRIM47 knockout. Further research showed that TRIM47 inhibited FOXO1 expression by ubiquitination and degradation of FOXO1, thereby promoting glioma growth and progression. Conclusion In our study, we confirmed functional role of the TRIM47-FOXO1 axis in the progression of gliomas and provided a potential target for glioma treatment.
Collapse
Affiliation(s)
- Huaming Wei
- Department of Neurology, Jiyang District People's Hospital of Jinan, Jinan, Shandong 251400, People's Republic of China
| | - Chonglan Ding
- Special Inspection Section, Shandong Zaozhuang Traditional Chinese Medicine Hospital, Zaozhuang, Shandong 277000, People's Republic of China
| | - Huanxia Zhuang
- Department of Neurology, Gaotang County People's Hospital, Gaotang, Shandong 252800, People's Republic of China
| | - WeiLi Hu
- Department of Neurology, Lianshui County People's Hospital, Lianshui, Jiangsu 223400, People's Republic of China
| |
Collapse
|
3
|
Mishra S, Verma SS, Rai V, Awasthee N, Arya JS, Maiti KK, Gupta SC. Curcuma raktakanda Induces Apoptosis and Suppresses Migration in Cancer Cells: Role of Reactive Oxygen Species. Biomolecules 2019; 9:biom9040159. [PMID: 31018580 PMCID: PMC6523773 DOI: 10.3390/biom9040159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Although over 100 species of Curcuma are reported, only Curcuma longa is extensively studied. Curcuma raktakanda, a poorly studied species, is most commonly distributed in the Kerala state of India. For the first time, we examined the efficacy of different fractions (acetone, hexane, and ethyl acetate) of C. raktakanda against glioma, cervical, and breast cancer cell lines. As determined by mitochondrial reductase activity assay, the viability of cancer cells was decreased in a concentration-dependent manner by the three fractions. The half maximal inhibitory concentration (IC-50) values after the treatment of C-6 glioma cells for 48 h was found to be 32.97 µg/mL (acetone extract), 40.63 µg/mL (hexane extract), and 51.65 µg/mL (ethyl acetate extract). Of the three fractions, the acetone fraction was more effective. The long-term colony formation of cancer cells was significantly suppressed by the acetone fraction. Analyses using DAPI (4',6-diamidino-2-phenylindole) staining, AO/PI (acridine orange/propidium iodide) staining, DNA laddering, and sub-G1 population revealed that the acetone extract induced apoptosis in glioma cells. The extract induced reactive oxygen species generation and suppressed the expression of cell survival proteins. The migration of cancer cells was also suppressed by the acetone extract. The gas chromatography-mass spectrometry (GC-MS) analysis indicated that tetracontane, dotriacontane, hexatriacontane, pentacosane, hexacosane, and eicosane are the major components in the acetone extract. Collectively, the extract from C. raktakanda exhibited anti-carcinogenic activities in cancer cells. We are exploring whether the phytoconstituents, individually, or collectively contribute to the anti-cancer activities of C. raktakanda.
Collapse
Affiliation(s)
- Shruti Mishra
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Sumit Singh Verma
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Vipin Rai
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Nikee Awasthee
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Jayadev S Arya
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Kaustabh K Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Subash C Gupta
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| |
Collapse
|
4
|
Xie CR, You CG, Zhang N, Sheng HS, Zheng XS. Epigallocatechin Gallate Preferentially Inhibits O6-Methylguanine DNA-Methyltransferase Expression in Glioblastoma Cells Rather than in Nontumor Glial Cells. Nutr Cancer 2018; 70:1339-1347. [PMID: 30558449 DOI: 10.1080/01635581.2018.1539189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Chao-Ran Xie
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Neurosurgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Chao-Guo You
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Neurosurgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Nu Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Han-Song Sheng
- Department of Neurosurgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Xue-Sheng Zheng
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
5
|
Chen Z, Wei X, Shen L, Zhu H, Zheng X. 20(S)-ginsenoside-Rg3 reverses temozolomide resistance and restrains epithelial-mesenchymal transition progression in glioblastoma. Cancer Sci 2018; 110:389-400. [PMID: 30431207 PMCID: PMC6317960 DOI: 10.1111/cas.13881] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/11/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant human intracranial tumors. Temozolomide (TMZ) is the primary alkylating agent for GBM patients. However, many GBM patients are resistant to TMZ. Therefore, patients with GBM urgently need more effective therapeutic options. 20(S)‐ginsenoside‐Rg3 (20(S)‐Rg3) is a natural chemical with anti‐tumor effects, but at present there is little understanding of its functional mechanism. Several research reports have demonstrated that O6‐methylguanine DNA‐methyltransferase (MGMT) repairs damaged DNA and contributes to TMZ resistance in gliomas. In addition, recent studies have shown that MGMT gene expression could be regulated by the Wnt/β‐catenin pathway. However, whether 20(S)‐Rg3 inhibits MGMT expression and augments chemosensitivity to Temozolomide (TMZ) in glioma cells remains unclear. In this study, we explored the modulating effects of 20(S)‐Rg3 on MGMT. We used glioma cell lines, primary cell strain (including T98G, U118 and GBM‐XX; all of them are MGMT‐positive glioma cell lines) and xenograft glioma models to examine whether 20(S)‐Rg3 increased the sensitivity to TMZ and to reveal the underlying mechanisms. We found that the MGMT expression was effectively downregulated by 20(S)‐Rg3 via the Wnt/β‐catenin pathway in glioma cell lines, and TMZ resistance was significantly reversed by 20(S)‐Rg3. Meanwhile, 20(S)‐Rg3 shows no obvious cytotoxicity at its effective dose and is well tolerated in vivo. In addition, we found that 20(S)‐Rg3 significantly restrains the epithelial‐mesenchymal transition (EMT) progression of glioma cells. Taken together, these results indicate that 20(S)‐Rg3 may be a novel agent to use in treatment of GBM, especially in TMZ‐resistant GBM with high MGMT expression.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiangyu Wei
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lin Shen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Hanshuo Zhu
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuesheng Zheng
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
You CG, Sheng HS, Xie CR, Zhang N, Zheng XS. FM19G11 inhibits O 6 -methylguanine DNA-methyltransferase expression under both hypoxic and normoxic conditions. Cancer Med 2018; 7:3292-3300. [PMID: 29761922 PMCID: PMC6051152 DOI: 10.1002/cam4.1551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/14/2018] [Accepted: 04/19/2018] [Indexed: 12/20/2022] Open
Abstract
FM19G11 is a small molecular agent that inhibits hypoxia-inducible factor-1-alpha (HIF-1α) and other signaling pathways. In this study, we characterized the modulating effects of FM19G11 on O6 -methylguanine DNA-methyltransferase (MGMT), the main regulator of temozolomide (TMZ) resistance in glioblastomas. This study included 2 MGMT-positive cell lines (GBM-XD and T98G). MGMT promoter methylation status, mRNA abundance, and protein levels were determined before and after FM19G11 treatment, and the roles of various signaling pathways were characterized. Under hypoxic conditions, MGMT mRNA and protein levels were significantly downregulated by FM19G11 via the HIF-1α pathway in both GBM-XD and T98G cells. In normoxic culture, T98G cells were strongly positive for MGMT, and MGMT expression was substantially downregulated by FM19G11 via the NF-κB pathway. In addition, TMZ resistance was reversed by treatment with FM19G11. Meanwhile, FM19G11 has no cytotoxicity at its effective dose. FM19G11 could potentially be used to counteract TMZ resistance in MGMT-positive glioblastomas.
Collapse
Affiliation(s)
- Chao-Guo You
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China.,Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Han-Song Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chao-Ran Xie
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China.,Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue-Sheng Zheng
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Diao B, Liu Y, Zhang Y, Yu J, Xie J, Xu GZ. IQGAP1‑siRNA inhibits proliferation and metastasis of U251 and U373 glioma cell lines. Mol Med Rep 2017; 15:2074-2082. [PMID: 28259970 PMCID: PMC5365011 DOI: 10.3892/mmr.2017.6257] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
Abstract
IQ motif containing GTPase activating protein 1 (IQGAP1) is a scaffold protein, which is aberrantly expressed in several tumor types and is closely associated with the development, metastasis and prognosis of cancer. Several studies have demonstrated that IQGAP1 has broad prospects in the basic and clinical research of tumors. The present study aimed to explore the effects of IQGAP1‑small interfering (si) NA on the proliferation and metastasis of U251 and U373 glioma cell lines, which markedly expressed IQGAP1. The human glioma cell lines (U251 and U373) were transfected with siRNA and transfection efficacy was confirmed by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis. Cell proliferation was detected using the Cell Counting kit‑8, and cell metastasis capabilities were detected using cell adhesion, migration and invasion assays. In addition, the expression levels of several tumor‑associated genes were determined by RT‑qPCR and western blotting. The results indicated that IQGAP1 was expressed at higher levels in glioma tissues compared with in normal brain tissues. IQGAP1‑siRNA significantly inhibited cell proliferation, and cell adhesion, migration and invasion. Furthermore, the expression levels of matrix metalloproteinase (MMP)2, Snail, MMP9, fibronectin 1 and Twist were suppressed, and E‑cadherin was upregulated in response to siRNA‑IQGAP1. The present study identified the function of IQGAP1 in glioma cell biology, and indicated that it may be considered a novel target for glioma treatment.
Collapse
Affiliation(s)
- Bo Diao
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Command and Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei 430070, P.R. China
| | - Ying Liu
- Department of Clinical Laboratory, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Yi Zhang
- Department of Clinical Experiment, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Jing Yu
- Department of Clinical Experiment, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Jun Xie
- Department of Science and Training, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Guo-Zheng Xu
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Command and Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
8
|
Bache M, Rot S, Keßler J, Güttler A, Wichmann H, Greither T, Wach S, Taubert H, Söling A, Bilkenroth U, Kappler M, Vordermark D. mRNA expression levels of hypoxia-induced and stem cell-associated genes in human glioblastoma. Oncol Rep 2015; 33:3155-61. [PMID: 25963717 DOI: 10.3892/or.2015.3932] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/24/2015] [Indexed: 11/05/2022] Open
Abstract
The roles of hypoxia-induced and stem cell-associated genes in the development of malignancy and tumour progression are well known. However, there are a limited number of studies analysing the impact of mRNA expression levels of hypoxia-induced and stem cell-associated genes in the tissues of brain tumours and glioblastoma patients. In this study, tumour tissues from patients with glioblastoma multiforme and tumour adjacent tissues were analysed. We investigated mRNA expression levels of hypoxia-inducible factor-1α (HIF-1α), hypoxia-inducible factor-2α (HIF-2α), carbonic anhydrase 9 (CA9), vascular endothelial growth factor (VEGF), glucose transporter-1 (GLUT-1) and osteopontin (OPN), and stem cell-associated genes survivin, epidermal growth factor receptor (EGFR), human telomerase reverse transcriptase (hTERT), Nanog and octamer binding transcription factor 4 (OCT4) using quantitative real-time polymerase chain reaction (qRT-PCR). Our data revealed higher mRNA expression levels of hypoxia-induced and stem cell-associated genes in tumour tissue than levels in the tumour adjacent tissues in patients with glioblastoma multiforme. A strong positive correlation between the mRNA expression levels of HIF-2α, CA9, VEGF, GLUT-1 and OPN suggests a specific hypoxia-associated profile of mRNA expression in glioblastoma multiforme. Additionally, the results indicate the role of stem-cell-related genes in tumour hypoxia. Kaplan-Maier analysis revealed that high mRNA expression levels of hypoxia-induced markers showed a trend towards shorter overall survival in glioblastoma patients (P=0.061). Our data suggest that mRNA expression levels of hypoxia-induced genes are important tumour markers in patients with glioblastoma multiforme.
Collapse
Affiliation(s)
- Matthias Bache
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Swetlana Rot
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jacqueline Keßler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Antje Güttler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Henri Wichmann
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Thomas Greither
- Center for Reproductive Medicine and Andrology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sven Wach
- Clinic of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Helge Taubert
- Clinic of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ariane Söling
- Department of Pediatrics, University of Göttingen, Göttingen, Germany
| | | | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dirk Vordermark
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
9
|
Temozolomide may induce cell cycle arrest by interacting with URG4/URGCP in SH-SY5Y neuroblastoma cells. Tumour Biol 2015; 36:6765-72. [DOI: 10.1007/s13277-015-3373-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/24/2015] [Indexed: 11/26/2022] Open
|
10
|
Bache M, Bernhardt S, Passin S, Wichmann H, Hein A, Zschornak M, Kappler M, Taubert H, Paschke R, Vordermark D. Betulinic acid derivatives NVX-207 and B10 for treatment of glioblastoma--an in vitro study of cytotoxicity and radiosensitization. Int J Mol Sci 2014; 15:19777-90. [PMID: 25361208 PMCID: PMC4264138 DOI: 10.3390/ijms151119777] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023] Open
Abstract
Betulinic acid (BA), a pentacyclic triterpene, represents a new therapeutic substance that has potential benefits for treating glioblastoma. Recently, new strategies for producing BA derivatives with improved properties have evolved. However, few studies have examined the combination of BA or BA derivatives using radiotherapy. The effects of two BA derivatives, NVX-207 and B10, on cellular and radiobiological behavior were analyzed using glioblastoma cell lines (U251MG, U343MG and LN229). Based on IC50 values under normoxic conditions, we detected a 1.3-2.9-fold higher cytotoxicity of the BA derivatives B10 and NVX-207, respectively, compared to BA. Incubation using both BA derivatives led to decreased cell migration, cleavage of PARP and decreased protein expression levels of Survivin. Weak radiation sensitivity enhancement was observed in U251MG cells after treatment with both BA derivatives. The enhancement factors at an irradiation dose of 6 Gy after treatment with 5 µM NVX-207 and 5 µM B10 were 1.32 (p=0.029) and 1.55 (p=0.002), respectively. In contrast to BA, neither NVX-207 nor B10 had additional effects under hypoxic conditions. Our results suggest that the BA derivatives NVX-207 and B10 improve the effects of radiotherapy on human malignant glioma cells, particularly under normoxic conditions.
Collapse
Affiliation(s)
- Matthias Bache
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Stephan Bernhardt
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Sarina Passin
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Henri Wichmann
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Anja Hein
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Martin Zschornak
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| | - Helge Taubert
- Clinic of Urology, Friedrich Alexander University Hospital Erlangen, Hartmann Str. 14, D-91054 Erlangen, Germany.
| | - Reinhard Paschke
- Biozentrum, Martin Luther Universität Halle-Wittenberg, Weinbergweg 22, D-06120 Halle, Germany.
| | - Dirk Vordermark
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst Grube Straße 40, D-06120 Halle, Germany.
| |
Collapse
|
11
|
Multiscale design of cell-type-specific pharmacokinetic/pharmacodynamic models for personalized medicine: application to temozolomide in brain tumors. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e112. [PMID: 24785551 PMCID: PMC4017092 DOI: 10.1038/psp.2014.9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/18/2014] [Indexed: 12/31/2022]
Abstract
Optimizing anticancer therapeutics needs to account for variable drug responses in heterogeneous cell populations within the tumor as well as in organs of toxicity. To address cell heterogeneity, we propose a multiscale modeling approach—from in vitro to preclinical and clinical studies—to develop cell-type–specific pharmacokinetic–pharmacodynamic (PK-PD) models. A physiologically based mechanistic modeling approach integrating data from aqueous solutions, U87 glioma cells, mice, and cancer patients was utilized to characterize the brain disposition of temozolomide (TMZ), the cornerstone of chemotherapy against glioblastoma multiforme. The final model represented intracellular normal brain and brain tumor compartments in which TMZ pH-dependent conversion to the DNA-alkylating species leads to the formation of DNA adducts that serve as an entry point for a PD model. This multiscale protocol can be extended to account for TMZ PK-PD in different cell populations, thus providing a critical tool to personalize TMZ-based chemotherapy on a cell-type–specific basis.
Collapse
|
12
|
Shen W, Hu JA, Zheng JS. Mechanism of temozolomide-induced antitumour effects on glioma cells. J Int Med Res 2013; 42:164-72. [PMID: 24326954 DOI: 10.1177/0300060513501753] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To investigate the mechanisms of action of the tumoricidal effects of temozolomide against the human glioma cell line U251 in vitro, and to provide preclinical proof-of-concept studies of the effects of temozolomide-containing regimens. METHODS U251 cells were exposed to 100 µmol/l temozolomide. Morphological alterations were monitored by light microscopy. Cell viability was measured using the 3 -(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell cycle analysis and the rate of apoptosis were determined using flow cytometry and the number of acidic vesicular organelles stained with acridine orange were analysed by fluorescence microscopy. The scratch recovery test was used to measure cell migration. RESULTS U251 cells that were treated with temozolomide displayed morphological alterations indicative of a rounder shape and impaired cellular adhesion to the cell culture plate compared with control U251 cells. Temozolomide reduced cell viability as measured by the MTT assay, caused cell cycle arrest in the gap 2/mitosis phase, inhibited cell migration and promoted autophagy in U251 cells. CONCLUSION Temozolomide induced autophagic, but not apoptotic processes, in U251 cells and thus reduced their viability and migration.
Collapse
Affiliation(s)
- Wei Shen
- Department of Neurosurgery, Beilun People's Hospital, Ningbo, Zhejiang Province, China
| | | | | |
Collapse
|
13
|
MicroRNA-650 expression in glioma is associated with prognosis of patients. J Neurooncol 2013; 115:375-80. [PMID: 24062138 DOI: 10.1007/s11060-013-1243-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 09/06/2013] [Indexed: 12/20/2022]
Abstract
MicroRNAs are known as non-coding RNAs that regulate the expression of target mRNA. Accumulating evidence has indicated that microRNA expression in human malignancies can be utilized as a prognostic marker for patients. However, the prognostic value of miR-650 in human glioma has not been investigated yet. In the present investigation, we have recruited 168 cases glioma specimens and 21 normal control brain specimens. Quantitative real-time PCR was carried out to investigate the expression of miR-650. Kaplan-Meier analysis and Cox's proportional hazards model was used to evaluate the association of miR-650 with prognosis of glioma patients. Results showed that miR-650 expression was increased in glioma compared with normal control specimens (P < 0.001). It was also found that miR-650 expression was related to World Health Organization grade and Karnofsky performance score (KPS) for high expression was more frequently detected in glioma of high grade or low KPS score (P < 0.001). The prognosis of glioma with high miR-650 expression was significantly worse compared with that of glioma with low miR-650 expression. These results proved that miR-650 expression was a significant prognostic indicator in glioma, which may suggest new management of human glioma.
Collapse
|
14
|
Dahlrot RH, Kristensen BW, Hjelmborg J, Herrstedt J, Hansen S. A population-based study of high-grade gliomas and mutated isocitrate dehydrogenase 1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2012; 6:31-40. [PMID: 23236540 PMCID: PMC3515987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 11/10/2012] [Indexed: 06/01/2023]
Abstract
UNLABELLED High-grade gliomas have a dismal prognosis, and prognostic factors are needed to optimize treatment algorithms. In this study we identified clinical prognostic factors as well as the prognostic value of isocitrate dehydrogenase 1 (IDH1) status in a population-based group of patients with high-grade gliomas. Using the Danish Cancer Registry and the Danish Pathology Databank we identified 359 patients: 234 had WHO grade IV gliomas, 58 had WHO grade III gliomas, and 67 were diagnosed clinically. Mutated IDH1 was predominantly observed in oligodendroglial tumors (WHO grade III). Patients with mutated IDH1 had a significantly better outcome than patients with wildtype IDH1: 2-year OS 59% and 18%, respectively (HR 0.38, 95% CI 0.21-0.68). However, when adjusting for other prognostic factors, IDH1 status was not a significant independent prognostic factor (HR=0.58, 95% CI 0.32-1.07). Young age, absence of neurological deficit, performance status 0-1, tumor not crossing the midline, and receiving post-surgical treatment were significant independent indicators of a good prognosis in multivariate analysis. IN CONCLUSION This population-based study could not demonstrate IDH1 status to be an independent prognostic factor in high-grade gliomas when adjusting for the effect of classic prognostic factors.
Collapse
Affiliation(s)
- Rikke H Dahlrot
- Department of Oncology, Odense University HospitalOdense, Denmark
- Institute of Clinical Research, University of Southern DenmarkOdense, Denmark
| | - Bjarne W Kristensen
- Department of Pathology, Odense University HospitalOdense, Denmark
- Institute of Clinical Research, University of Southern DenmarkOdense, Denmark
| | - Jacob Hjelmborg
- Department of Biostatistics, Institute of Public Health, University of Southern DenmarkOdense, Denmark
| | - Jørn Herrstedt
- Department of Oncology, Odense University HospitalOdense, Denmark
- Institute of Clinical Research, University of Southern DenmarkOdense, Denmark
| | - Steinbjørn Hansen
- Department of Oncology, Odense University HospitalOdense, Denmark
- Institute of Clinical Research, University of Southern DenmarkOdense, Denmark
| |
Collapse
|
15
|
da Silva AC, Cabral FR, Mamani JB, Malheiros JM, Polli RS, Tannus A, Vidoto E, Martins MJ, Sibov TT, Pavon LF, Miyaki LAM, Cárdenas WHZ, Malheiros SMF, Brandt RA, Amaro Júnior E, Gamarra LF. Tumor growth analysis by magnetic resonance imaging of the C6 glioblastoma model with prospects for the assessment of magnetohyperthermia therapy. EINSTEIN-SAO PAULO 2012; 10:11-5. [PMID: 23045819 DOI: 10.1590/s1679-45082012000100004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The objective was to establish a pattern of tumor growth of the C6 model of glioblastoma multiform in Wistar rats via magnetic resonance imaging (MRI) for the subsequent verification of tumor volume reduction due to magnetic hyperthermia therapy. METHODS Young male Wistar rats weighing between 250 and 300 g were used for the C6 model. After the rats were anesthetized (55 mg/ kg ketamine and 11 mg/kg xylazine), C6 lineage tumorigenic cells suspended in culture medium (10(5) cells in 10 microl) were stereotaxically injected into the right frontal cortex (bregma coordinates: 2.0 mm anteroposterior, 3.0 mm laterolateral, and 2.5 mm depth) of the rats using a Hamilton syringe. For the control group, the rats were injected with culture medium without cells. MRI scans were performed at 14, 21, and 28 d after the injection using a 2.0 T MRI scanner (Bruker BioSpec, Germany). The animals were anesthetized with 55 mg/kg ketamine and 11 mg/kg xylazine before being examined. Coronal multilayers were acquired using a standard spin echo sequence with the following parameters: repetition/echo time = 4.000 ms/67.1 ms, field of view = 3.50, matrix = 192, slice thickness = 0.4 mm, and slice separation = 0 mm. RESULTS The MRI analysis enabled a clear visualization of the tumor mass, and it was possible to establish the tumor volume parameters on the various days that were examined. The volume at 14 d after induction was 13.7 +/- 2.5 mm3. On days 21 and 28, the tumor volumes were 31.7 +/- 6.5 mm3 and 122.1 +/- 11.8 mm3, respectively. CONCLUSION These results demonstrated that it is possible to evaluate the C6 model tumor volume in rats, which will allow for the future implementation and verification of magnetic hyperthermia therapy.
Collapse
Affiliation(s)
- André César da Silva
- Brain Institute, Hospital Israelita Albert Einstein - HIAE, São Paulo (SP), Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|