1
|
Roshani F, Ahvar M, Ebrahimi A. Network analysis to identify driver genes and combination drugs in brain cancer. Sci Rep 2024; 14:18666. [PMID: 39134610 PMCID: PMC11319350 DOI: 10.1038/s41598-024-69705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Brain cancer is one of the deadliest diseases, although many efforts have been made to treat it, there is no comprehensive and effective treatment approach yet. In recent years, the use of network-based analysis to identify important biological genes and pathways involved in various complex diseases, including brain cancer, has attracted the attention of researchers. The goal of this manuscript is to perform a comprehensive analysis of the various results presented related to brain cancer. For this purpose, firstly, based on the CORMINE medical database, collected all the genes related to brain cancer with a valid P-value. Then the structural and functional relationships between the above gene sets have been identified based on the STRING database. Next, in the PPI network, hub centrality analysis was performed to determine the proteins that have many connections with other proteins. After the modularization of the network, the module with the most hub vertices is considered as the most relevant module to the formation and progression of brain cancer. Since the driver vertices play an important role in biological systems, the edges of the selected module were oriented, and by analyzing the controllability of complex networks, a set of five proteins with the highest control power has been identified. Finally, based on the drug-gene interaction, a set of drugs effective on each of the driver genes has been obtained, which can potentially be used as new combination drugs. Validation of the hub and driver proteins shows that they are mainly essential proteins in the biological processes related to the various cancers and therefore the drugs that affect them can be considered as new combination therapy. The presented procedure can be used for any other complex disease.
Collapse
Affiliation(s)
| | - Mobina Ahvar
- Department of Physics, Alzahra University, Tehran, Iran
| | - Ali Ebrahimi
- Department of Physics, Alzahra University, Tehran, Iran.
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| |
Collapse
|
2
|
Singh D, Singh L, Kaur S, Arora A. Nucleic acids based integrated macromolecular complexes for SiRNA delivery: Recent advancements. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-24. [PMID: 38693628 DOI: 10.1080/15257770.2024.2347499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
The therapeutic potential of small interfering RNA (siRNA) is monumental, offering a pathway to silence disease-causing genes with precision. However, the delivery of siRNA to target cells in-vivo remains a formidable challenge, owing to degradation by nucleases, poor cellular uptake and immunogenicity. This overview examines recent advancements in the design and application of nucleic acid-based integrated macromolecular complexes for the efficient delivery of siRNA. We dissect the innovative delivery vectors developed in recent years, including lipid-based nanoparticles, polymeric carriers, dendrimer complexes and hybrid systems that incorporate stimuli-responsive elements for targeted and controlled release. Advancements in bioconjugation techniques, active targeting strategies and nanotechnology-enabled delivery platforms are evaluated for their contribution to enhancing siRNA delivery. It also addresses the complex interplay between delivery system design and biological barriers, highlighting the dynamic progress and remaining hurdles in translating siRNA therapies from bench to bedside. By offering a comprehensive overview of current strategies and emerging technologies, we underscore the future directions and potential impact of siRNA delivery systems in personalized medicine.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, India
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Simranjeet Kaur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Akshita Arora
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
3
|
Ignatova TN, Chaitin HJ, Kukekov NV, Suslov ON, Dulatova GI, Hanafy KA, Vrionis FD. Gliomagenesis is orchestrated by the Oct3/4 regulatory network. J Neurosurg Sci 2024; 68:148-156. [PMID: 34342203 DOI: 10.23736/s0390-5616.21.05437-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a lethal brain tumor characterized by developmental hierarchical phenotypic heterogeneity, therapy resistance and recurrent growth. Neural stem cells (NSCs) from human central nervous system (CNS), and glioblastoma stem cells from patient-derived GBM (pdGSC) samples were cultured in both 2D well-plate and 3D monoclonal neurosphere culture system (pdMNCS). The pdMNCS model shows promise to establish a relevant 3D-tumor environment that maintains GBM cells in the stem cell phase within suspended neurospheres. METHODS Utilizing the pdMNCS, we examined GBM cell-lines for a wide spectrum of developmental cancer stem cell markers, including the early blastocyst inner-cell mass (ICM)-specific Nanog, Oct3/4,B, and CD133. RESULTS We observed that MNCS epigenotype is recapitulated using gliomasphere-derived cells. CD133, the marker of GSC is robustly expressed in 3D-gliomaspheres and localized within the plasma membrane compartment. Conversely, gliomasphere cultures grown in conventional 2D culture quickly lost CD133 expression, indicating its variable expression is dependent on cell-culture conditions. Incomplete differentiation of cytoskeleton microtubules and intermediate filaments (IFs) of patient derived cells, similar to commercially available GBM cell lines, was seen. Subsequently, in order to determine whether Oct3/4 it was necessary for CD133 expression and cancer stemness, we transfected 2D and 3D culture with siRNA against Oct3/4 and found a significant reduction in gliomasphere formation. CONCLUSIONS These results suggest that expression of Oct3/4,A- and CD133 suppress differentiation of GSCs.
Collapse
Affiliation(s)
- Tatyana N Ignatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Hersh J Chaitin
- College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Nickolay V Kukekov
- Department of Pathology and Center for Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Oleg N Suslov
- McKnight Brain Institute, Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Galina I Dulatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
| | - Khalid A Hanafy
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Frank D Vrionis
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA -
| |
Collapse
|
4
|
Guo F, Ling G, Qiu J, Li J, Gan Y, Yu Y, Tang J, Mo L, Piao H. Juglone induces ferroptosis in glioblastoma cells by inhibiting the Nrf2-GPX4 axis through the phosphorylation of p38MAPK. Chin Med 2024; 19:52. [PMID: 38520025 PMCID: PMC10958923 DOI: 10.1186/s13020-024-00920-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Ferroptosis, a non-apoptotic form of cell death induced by accumulation of free iron ions and lipid peroxidation, its importance for cancer treatment is gradually being recognized. Research on the anti-cancer mechanism of juglone is accumulating. However, the specific mechanism by which it directs glioblastoma (GBM) to death is unknown. METHODS We used in vitro and in vivo experiments to explore the anti-GBM effect generated by juglone through the ferroptosis pathway. RESULTS Juglone mainly causes cell death by inducing ferroptosis. Mechanistically, juglone can significantly activate the phosphorylation of p38MAPK. According to transcriptome sequencing and protein interaction analysis, the Nrf2-GPX4 signaling pathway is identified as the primary pathway through which juglone mediates ferroptosis. In vitro and in vivo experiments further verified that juglone induces the ferroptosis of GBM by activating the phosphorylation of p38MAPK and negatively regulating the Nrf2-GPX4 signaling pathway. CONCLUSION Juglone induces ferroptosis and inhibits the growth of GBM by targeting the Nrf2/Gpx4 signaling pathway and thus holds promise as a novel ferroptosis inducer or anti-GBM drug.
Collapse
Affiliation(s)
- Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
- Graduate School, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Guoyuan Ling
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
- Graduate School, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jianting Qiu
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, 110042, Liaoning, China
| | - Jicheng Li
- Graduate School, China Medical University, Shenyang, 110042, Liaoning, China
| | - Yu Gan
- Graduate School, China Medical University, Shenyang, 110042, Liaoning, China
| | - YingYing Yu
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, 110042, Liaoning, China
| | - Jiamei Tang
- Graduate School, China Medical University, Shenyang, 110042, Liaoning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110801, Liaoning, China.
| |
Collapse
|
5
|
da Costa BHB, Becker AP, Neder L, Gonçalves PG, de Oliveira C, Polverini AD, Clara CA, Teixeira GR, Reis RM, Bidinotto LT. EGFL7 expression profile in IDH-wildtype glioblastomas is associated with poor patient outcome. J Pathol Transl Med 2022; 56:205-211. [PMID: 35698739 PMCID: PMC9288890 DOI: 10.4132/jptm.2022.04.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background Despite the advances in glioblastoma (GBM) treatment, the average life span of patients is 14 months. Therefore, it is urgent to identity biomarkers of prognosis, treatment response, or development of novel treatment strategies. We previously described the association of high epidermal growth factor-like domain multiple 7 (EGFL7) expression and unfavorable outcome of pilocytic astrocytoma patients. The present study aims to analyze the prognostic potential of EGFL7 in GBM isocitrate dehydrogenase (IDH)-wildtype, using immunohistochemistry and in silico approaches. Materials and Methods Spearman's correlation analysis of The Cancer Genome Atlas RNA sequencing data was performed. The genes strongly correlated to EGFL7 expression were submitted to enrichment gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Additionally, EGFL7 expression was associated with patient overall survival. The expression of EGFL7 was analyzed through immunohistochemistry in 74 GBM IDH-wildtype patients' samples, and was associated with clinicopathological data and overall survival. Results In silico analysis found 78 genes strongly correlated to EGFL7 expression. These genes were enriched in 40 biological processes and eight KEGG pathways, including angiogenesis/vasculogenesis, cell adhesion, and phosphoinositide 3-kinase-Akt, Notch, and Rap1 signaling pathways. The immunostaining showed high EGFL7 expression in 39 cases (52.7%). High immunolabelling was significantly associated with low Karnofsky Performance Status and poor overall survival. Cox analysis showed that GBMs IDH-wildtype with high EGFL7 expression presented a higher risk of death compared to low expression (hazard ratio, 1.645; 95% confidence interval, 1.021 to 2.650; p = .041). Conclusion This study gives insights regarding the genes that are correlated with EGFL7, as well as biological processes and signaling pathways, which should be further investigated in order to elucidate their role in glioblastoma biology.
Collapse
Affiliation(s)
- Bruno Henrique Bressan da Costa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, São Paulo, Brazil
| | - Aline Paixão Becker
- The Ohio State University, Department of Radiation Oncology, Columbus, OH, USA
| | - Luciano Neder
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,Department of Pathology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Paola Gyuliane Gonçalves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,UNESP - Univ. Estadual Paulista, School of Medicine, Department of Pathology, Botucatu, São Paulo, Brazil
| | - Cristiane de Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,UNESP - Univ. Estadual Paulista, School of Medicine, Department of Pathology, Botucatu, São Paulo, Brazil
| | - Allan Dias Polverini
- Department of Neurosurgery, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Carlos Afonso Clara
- Department of Neurosurgery, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Gustavo Ramos Teixeira
- Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, São Paulo, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucas Tadeu Bidinotto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, São Paulo, Brazil.,UNESP - Univ. Estadual Paulista, School of Medicine, Department of Pathology, Botucatu, São Paulo, Brazil
| |
Collapse
|
6
|
Kolodziej MA, Gött H, Kopischke B, Bender MK, Weigand MA, Di Fazio P, Schwarm FP, Uhle F. Antiproliferative effect of GTS-21 in glioblastoma cells. Oncol Lett 2021; 22:759. [PMID: 34539863 PMCID: PMC8436335 DOI: 10.3892/ol.2021.13020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain tumour in adults. The poor prognosis and short median overall survival of patients with GBM is associated with resistance to therapy after surgical and adjuvant treatment. The expression of various acetylcholine receptors (AChR) in GBM has been widely reported. The present study aimed to investigate the expression of cholinergic system-related genes in primary GBM and to explore the antiproliferative effect of 3-(2,4-dimethoxybenzylidene) anabaseine (GTS-21) in GBM cell lines. Therefore, the expression of 28 genes associated with the cholinergic system was detected using a customized RT2 Profiler PCR Array in 44 GBM and 5 healthy control brain tissue samples. In addition, the activity of GTS-21, an alpha 7 subunit nicotinic AChR (α7 nAChR) agonist, and that of α-bungarotoxin (α-BTX), an α7 nAChR antagonist, was determined in primary and established GBM cells. Therefore, the A172, U87 and G28 cell lines and primary GBM cells were treated with GTS-21, ACh or nicotine. Cell viability was evaluated using MTT assay at 24, 48 and 72 h following cell treatment with the corresponding compounds. The results revealed that the expression of cholinergic system-related components was notably downregulated, except that of cholinergic receptor nicotinic alpha 7 subunit (CHRNA7), in primary GBM and U87 cells. However, the dominant-negative duplicate form of CHRNA7 was also downregulated. Furthermore, A172 and G28 cells exhibited a heterogeneous gene expression pattern. Additionally, GTS-21 inhibited the proliferation of GBM cells in a dose- and time-dependent manner. Interestingly, treatment with α-BTX restored the proliferation of U87 cells, but not that of A172 and G28 cells. Collectively, the findings of the present study suggested that GTS-21 may inhibit the proliferation of GBM cells and may therefore serve as a novel therapeutic approach to the treatment of GBM, which warrants further investigation.
Collapse
Affiliation(s)
- Malgorzata A. Kolodziej
- Department of Neurosurgery, Justus-Liebig University Giessen, D-35392 Giessen, Germany
- Correspondence to: Dr Malgorzata Anna Kolodziej, Department of Neurosurgery, Justus-Liebig University Giessen, Klinikstrasse 33, D-35392 Giessen, Germany, E-mail:
| | - Hanna Gött
- Department of Neurosurgery, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Benjamin Kopischke
- Department of Neurosurgery, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Michael K.F. Bender
- Department of Neurosurgery, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Markus A. Weigand
- Department of Anaesthesiology, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, D-35033 Marburg, Germany
| | - Frank P. Schwarm
- Department of Neurosurgery, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Florian Uhle
- Department of Anaesthesiology, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| |
Collapse
|
7
|
Núñez-Carpintero I, Petrizzelli M, Zinovyev A, Cirillo D, Valencia A. The multilayer community structure of medulloblastoma. iScience 2021; 24:102365. [PMID: 33889829 PMCID: PMC8050854 DOI: 10.1016/j.isci.2021.102365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 01/20/2023] Open
Abstract
Multilayer networks allow interpreting the molecular basis of diseases, which is particularly challenging in rare diseases where the number of cases is small compared with the size of the associated multi-omics datasets. In this work, we develop a dimensionality reduction methodology to identify the minimal set of genes that characterize disease subgroups based on their persistent association in multilayer network communities. We use this approach to the study of medulloblastoma, a childhood brain tumor, using proteogenomic data. Our approach is able to recapitulate known medulloblastoma subgroups (accuracy >94%) and provide a clear characterization of gene associations, with the downstream implications for diagnosis and therapeutic interventions. We verified the general applicability of our method on an independent medulloblastoma dataset (accuracy >98%). This approach opens the door to a new generation of multilayer network-based methods able to overcome the specific dimensionality limitations of rare disease datasets. The molecular interpretation of rare diseases is a challenging task Multilayer networks allow patient stratification and explainability We identify subgroup-specific genes and multilayer associations in medulloblastoma Multilayer community analysis enables the molecular interpretation of rare diseases
Collapse
Affiliation(s)
| | - Marianyela Petrizzelli
- Institut Curie, PSL Research University, 75005 Paris, France
- INSERM, U900, 75005 Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, 75006 Paris, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, 75005 Paris, France
- INSERM, U900, 75005 Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, 75006 Paris, France
- Lobachevsky University, 603000 Nizhny Novgorod, Russia
| | - Davide Cirillo
- Barcelona Supercomputing Center (BSC), C/ Jordi Girona 29, 08034, Barcelona, Spain
- Corresponding author
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), C/ Jordi Girona 29, 08034, Barcelona, Spain
- ICREA - Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| |
Collapse
|
8
|
Osuka S, Zhu D, Zhang Z, Li C, Stackhouse CT, Sampetrean O, Olson JJ, Gillespie GY, Saya H, Willey CD, Van Meir EG. N-cadherin upregulation mediates adaptive radioresistance in glioblastoma. J Clin Invest 2021; 131:136098. [PMID: 33720050 PMCID: PMC7954595 DOI: 10.1172/jci136098] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is composed of heterogeneous tumor cell populations, including those with stem cell properties, termed glioma stem cells (GSCs). GSCs are innately less radiation sensitive than the tumor bulk and are believed to drive GBM formation and recurrence after repeated irradiation. However, it is unclear how GSCs adapt to escape the toxicity of repeated irradiation used in clinical practice. To identify important mediators of adaptive radioresistance in GBM, we generated radioresistant human and mouse GSCs by exposing them to repeat cycles of irradiation. Surviving subpopulations acquired strong radioresistance in vivo, which was accompanied by a reduction in cell proliferation and an increase in cell-cell adhesion and N-cadherin expression. Increasing N-cadherin expression rendered parental GSCs radioresistant, reduced their proliferation, and increased their stemness and intercellular adhesive properties. Conversely, radioresistant GSCs lost their acquired phenotypes upon CRISPR/Cas9-mediated knockout of N-cadherin. Mechanistically, elevated N-cadherin expression resulted in the accumulation of β-catenin at the cell surface, which suppressed Wnt/β-catenin proliferative signaling, reduced neural differentiation, and protected against apoptosis through Clusterin secretion. N-cadherin upregulation was induced by radiation-induced IGF1 secretion, and the radiation resistance phenotype could be reverted with picropodophyllin, a clinically applicable blood-brain-barrier permeable IGF1 receptor inhibitor, supporting clinical translation.
Collapse
Affiliation(s)
- Satoru Osuka
- Department of Neurosurgery, School of Medicine and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Dan Zhu
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Zhaobin Zhang
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Chaoxi Li
- Department of Neurosurgery, School of Medicine and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Christian T. Stackhouse
- Department of Neurosurgery, School of Medicine and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, USA
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Jeffrey J. Olson
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - G. Yancey Gillespie
- Department of Neurosurgery, School of Medicine and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Christopher D. Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, USA
| | - Erwin G. Van Meir
- Department of Neurosurgery, School of Medicine and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Bender K, Träger M, Wahner H, Onken J, Scheel M, Beck M, Ehret F, Budach V, Kaul D. What is the role of the subventricular zone in radiotherapy of glioblastoma patients? Radiother Oncol 2021; 158:138-145. [PMID: 33636228 DOI: 10.1016/j.radonc.2021.02.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/28/2021] [Accepted: 02/13/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND PURPOSE Current glioblastoma (GBM) therapies prolong survival, but overall prognosis is still poor. Irradiation of the subventricular zone (SVZ) has recently been discussed as a promising concept as this tissue harbors stem cells which seem to play a role in the initiation and recurrence of GBM. In this study, we retrospectively examined the relationship of SVZ irradiation dose and survival in a large, homogeneous GBM patient cohort. MATERIALS AND METHODS We included 200 GBM patients who had been treated at our institution with trimodal therapy (surgery, radiotherapy and chemotherapy) between 2009 and 2020. The SVZ was delineated, and dose-volume histograms were calculated and extracted. Tumors were classified according to their contact with the SVZ. The Kaplan-Meier method was used for survival analysis, and univariable and multivariable Cox regression (MVA) were used to determine prognostic effects on progression-free survival (PFS) and overall survival (OS). RESULTS Median PFS of the study group was 7.2 months; median OS was 15.1 months. In MVA (with mean dose to the ipsilateral SVZ as a continuous covariable), PFS was significantly lower for patients with a Karnofsky performance status (KPS) < 70% and without MGMT promoter methylation. Factors prognostic for shorter OS were old age, lower KPS, unmethylated MGMT status, SVZ contact and biopsy instead of subtotal- or gross total resection. There was no significant correlation between survival and SVZ dose. CONCLUSION In this cohort, an increased mean dose to the ipsilateral or contralateral SVZ did not correlate with improved survival in irradiated GBM patients in MVA. Patients whose tumor directly involved the SVZ showed worse OS in MVA.
Collapse
Affiliation(s)
- Katja Bender
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Malte Träger
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Helena Wahner
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; German Cancer Consortium (DKTK), partner site Berlin, Germany
| | - Michael Scheel
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Marcus Beck
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Felix Ehret
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Volker Budach
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - David Kaul
- Department of Radiation Oncology Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; German Cancer Consortium (DKTK), partner site Berlin, Germany.
| |
Collapse
|
10
|
Vieira de Castro J, S. Gonçalves C, P. Martins E, Miranda-Lorenzo I, T. Cerqueira M, Longatto-Filho A, A. Pinto A, L. Reis R, Sousa N, Heeschen C, M. Costa B. Intracellular Autofluorescence as a New Biomarker for Cancer Stem Cells in Glioblastoma. Cancers (Basel) 2021; 13:828. [PMID: 33669350 PMCID: PMC7920313 DOI: 10.3390/cancers13040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 11/16/2022] Open
Abstract
The identification of cancer stem cells (CSCs), which are implicated in tumor initiation, progression, therapy resistance, and relapse, is of great biological and clinical relevance. In glioblastoma (GBM), this is still a challenge, as no single marker is able to universally identify populations of GBM cancer stem cells (GSCs). Indeed, there is still controversy on whether biomarker-expressing cells fulfill the functional criteria of bona fide GSCs, despite being widely used. Here, we describe a novel subpopulation of autofluorescent (Fluo+) cells in GBM that bear all the functional characteristics of GSCs, including higher capacity to grow as neurospheres, long-term self-renewal ability, increased expression of stem cell markers, and enhanced in vivo tumorigenicity. Mechanistically, the autofluorescent phenotype is largely due to the intracellular accumulation of riboflavin, mediated by the ABC transporter ABCG2. In summary, our work identifies an intrinsic cellular autofluorescent phenotype enriched in GBM cells with functional stem cells features that can be used as a novel, simple and reliable biomarker to target these highly malignant tumors, with implications for GBM biological and clinical research.
Collapse
Affiliation(s)
- Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
| | - Eduarda P. Martins
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
| | - Irene Miranda-Lorenzo
- Stem Cells and Cancer Group, Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (I.M.-L.); (C.H.)
| | - Mariana T. Cerqueira
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
- Medical Laboratory of Medical Investigation (LIM) 14, Department of Pathology, Medical School, University of São Paulo, São Paulo 01246-903, Brazil
| | - Afonso A. Pinto
- Department of Neurosurgery, Hospital de Braga, 4710-243 Braga, Portugal;
| | - Rui L. Reis
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
| | - Christopher Heeschen
- Stem Cells and Cancer Group, Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (I.M.-L.); (C.H.)
- Center for Single-Cell Omics & State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.); (E.P.M.); (A.L.-F.); (N.S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal; (M.T.C.); (R.L.R.)
| |
Collapse
|
11
|
Matsumoto T, Chino H, Akiya M, Hashimura M, Yokoi A, Tochimoto M, Nakagawa M, Jiang Z, Saegusa M. Requirements of LEFTY and Nodal overexpression for tumor cell survival under hypoxia in glioblastoma. Mol Carcinog 2020; 59:1409-1419. [PMID: 33111989 DOI: 10.1002/mc.23265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor β (TGF-β) superfamily, have glycogen synthase kinase 3β (GSK-3β) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3β, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3β, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3β, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-β1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3β inactivation, with or without cooperation of the TGF-β1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.
Collapse
Affiliation(s)
- Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiromi Chino
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masashi Akiya
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Zesong Jiang
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
12
|
Abstract
Even though the treatment of childhood cancer has evolved significantly in recent decades, aggressive central nervous system (CNS) tumors are still a leading cause of morbidity and mortality in this population. Consequently, the identification of molecular targets that can be incorporated into diagnostic practice, effectively predict prognosis, follow treatment response, and materialize into potential targeted therapeutic approaches are still warranted. Since the first evidence of the participation of miRNAs in cancer development and progression 20 years ago, notable progress has been made in the basic understanding of the contribution of their dysregulation as epigenetic driver of tumorigenesis. Nevertheless, among the plethora of articles in the literature, microRNA profiling of pediatric tumors are scarce. This article gives an overview of the recent advances in the diagnostic/prognostic potential of miRNAs in a selection of pediatric CNS tumors: medulloblastoma, ependymoma, pilocytic astrocytoma, glioblastoma, diffuse intrinsic pontine glioma, atypical teratoid/rhabdoid tumors, and choroid plexus tumors.
Collapse
|
13
|
Chew MT, Nisbet A, Suzuki M, Matsufuji N, Murakami T, Jones B, Bradley DA. Potential lethal damage repair in glioblastoma cells irradiated with ion beams of various types and levels of linear energy transfer. JOURNAL OF RADIATION RESEARCH 2019; 60:59-68. [PMID: 30452663 PMCID: PMC6373669 DOI: 10.1093/jrr/rry081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/06/2018] [Indexed: 06/09/2023]
Abstract
Glioblastoma (GBM), a Grade IV brain tumour, is a well-known radioresistant cancer. To investigate one of the causes of radioresistance, we studied the capacity for potential lethal damage repair (PLDR) of three altered strains of GBM: T98G, U87 and LN18, irradiated with various ions and various levels of linear energy transfer (LET). The GBM cells were exposed to 12C and 28Si ion beams with LETs of 55, 100 and 200 keV/μm, and with X-ray beams of 1.7 keV/μm. Mono-energetic 12C ions and 28Si ions were generated by the Heavy Ion Medical Accelerator at the National Institute of Radiological Science, Chiba, Japan. Clonogenic assays were used to determine cell inactivation. The ability of the cells to repair potential lethal damage was demonstrated by allowing one identical set of irradiated cells to repair for 24 h before subplating. The results show there is definite PLDR with X-rays, some evidence of PLDR at 55 keV/μm, and minimal PLDR at 100 keV/μm. There is no observable PLDR at 200 keV/μm. This is the first study, to the authors' knowledge, demonstrating the capability of GBM cells to repair potential lethal damage following charged ion irradiations. It is concluded that a GBM's PLDR is dependent on LET, dose and GBM strain; and the more radioresistant the cell strain, the greater the PLDR.
Collapse
Affiliation(s)
- Ming Tsuey Chew
- Sunway University, Centre for Biomedical Physics, School of Healthcare and Medical Sciences, No 5, Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Department of Physics, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK
| | - Andrew Nisbet
- Sunway University, Centre for Biomedical Physics, School of Healthcare and Medical Sciences, No 5, Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Department of Physics, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK
- The Department of Medical Physics, Royal Surrey County Hospital, Egerton Road, Guildford, UK
| | - Masao Suzuki
- Department of Basic Medical Sciences for Radiation Damages; National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, 4–9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, Japan
| | - Naruhiro Matsufuji
- Radiation Effect Research Team, Department of Accelerator and Medical Physics, NIRS, National Institutes for Quantum and Radiological Science and Technology, 4–9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, Japan
| | - Takeshi Murakami
- Heavy-Ion Radiotherapy Promotion Unit & Department of Accelerator and Medical Physics, NIRS, National Institutes for Quantum and Radiological Science and Technology, 4–9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, Japan
| | - Bleddyn Jones
- Gray Laboratory, CRUK/MRC Oxford Oncology Institute, University of Oxford, ORCRB-Roosevelt Drive, Oxford, UK
| | - David A Bradley
- Sunway University, Centre for Biomedical Physics, School of Healthcare and Medical Sciences, No 5, Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Department of Physics, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
14
|
Ghosh D, Nandi S, Bhattacharjee S. Combination therapy to checkmate Glioblastoma: clinical challenges and advances. Clin Transl Med 2018; 7:33. [PMID: 30327965 PMCID: PMC6191404 DOI: 10.1186/s40169-018-0211-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022] Open
Abstract
Combination therapy is increasingly becoming the cornerstone of current day antitumor therapy. Glioblastoma multiforme is an aggressive brain tumor with a dismal median survival post diagnosis and a high rate of disease recurrence. The poor prognosis can be attributed to unique treatment limitations, which include the infiltrative nature of tumor cells, failure of anti-glioma drugs to cross the blood-brain barrier, tumor heterogeneity and the highly metastatic and angiogenic nature of the tumor making cells resistant to chemotherapy. Combination therapy approach is being developed against glioblastoma with new innovative combination drug regimens being tested in preclinical and clinical trials. In this review, we discuss the pathophysiology of glioblastoma, diagnostic markers, therapeutic targeting strategies, current treatment limitations, novel combination therapies in the context of current treatment options and the ongoing clinical trials for glioblastoma therapy.
Collapse
Affiliation(s)
- Debarati Ghosh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Saikat Nandi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| | | |
Collapse
|
15
|
Smith SJ, Diksin M, Chhaya S, Sairam S, Estevez-Cebrero MA, Rahman R. The Invasive Region of Glioblastoma Defined by 5ALA Guided Surgery Has an Altered Cancer Stem Cell Marker Profile Compared to Central Tumour. Int J Mol Sci 2017; 18:E2452. [PMID: 29156557 PMCID: PMC5713419 DOI: 10.3390/ijms18112452] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma, a WHO grade IV astrocytoma, is a highly aggressive and heterogeneous tumour that infiltrates deeply into surrounding brain parenchyma, making complete surgical resection impossible. Despite chemo-radiotherapy, the residual cell population within brain parenchyma post-surgery causes inevitable recurrence. Previously, the tumour core has been the focus of research and the basis for targeted therapeutic regimes, which have failed to improve survival in clinical trials. Here, we focus on the invasive margin as defined by the region with 5-aminolevulinic acid (5ALA) (GliolanTM) fluorescence at surgery beyond the T1 enhancing region on magnetic resonance imaging (MRI). This area is hypothesized to constitute unique microenvironmental pressures, and consequently be molecularly distinct to tumour core and enhancing rim regions. We conducted hematoxylin and eosin (H&E), array real time polymerase chain reaction (PCR), and immunohistochemistry staining on various intra-tumour regions of glioblastoma to determine molecular heterogeneity between regions. We analyzed 73 tumour samples from 21 patients and compared cellular density, cell proliferation, and the degree of vascularity. There is a statistically significant difference between the core, invasive margin and other regions for cell density (p < 0.001), cell proliferation (p = 0.029), and vascularity (p = 0.007). Aldehyde dehydrogenase 1 (ALDH1) and Nestin immunohistochemistry were used as a measure of stem-like properties, showing significantly decreased Nestin expression (p < 0.0001) in the invasive margin. Array PCR of the core, rim, and invasive regions showed significantly increased fibroblast growth factor (FGF) and ALDH1 expression in the invasive zone, with elevated hypoxia inducing factor 1-alpha (HIF1α) in the rim region, adjacent to the hypoxic core. The influence of varying microenvironments in the intra-tumour regions is a major key to understanding intra-tumour heterogeneity. This study confirms the distinct molecular composition of the heterogeneous invasive margin and cautions against purported therapy strategies that target candidate glioblastoma stem-like genes that are predominantly expressed in the tumour core. Full characterization of tumour cells in the invasive margin is critical, as these cells may more closely resemble the residual cell population responsible for tumour recurrence. Their unique nature should be considered when developing targeted agents for residual glioblastoma multiforme (GBM).
Collapse
Affiliation(s)
- Stuart J Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Mohammed Diksin
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Saachi Chhaya
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Shwetha Sairam
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Maria A Estevez-Cebrero
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
16
|
Junca A, Villalva C, Tachon G, Rivet P, Cortes U, Guilloteau K, Balbous A, Godet J, Wager M, Karayan-Tapon L. Crizotinib targets in glioblastoma stem cells. Cancer Med 2017; 6:2625-2634. [PMID: 28960893 PMCID: PMC5673924 DOI: 10.1002/cam4.1167] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma stem cells (GSCs) are believed to be involved in the mechanisms of tumor resistance, therapeutic failures, and recurrences after conventional glioblastoma therapy. Therefore, elimination of GSCs might be a prerequisite for the development of successful therapeutic strategies. ALK, ROS1, and MET are targeted by Crizotinib, a tyrosine kinase inhibitor which has been approved for treatment of ALK-rearranged non-small-cell lung cancer. In this study we investigated ALK, ROS1, and MET status in nine glioblastoma stem cell lines and tumors from which they arise. Fluorescent in situ hybridization (FISH), Sanger's direct sequencing, and immunohistochemistry were used to screen genomic rearrangements (or amplifications), genomic mutations, and protein expression, respectively. The immunohistochemical and FISH studies revealed no significant dysregulation of ROS1 in GSCs and associated tumors. Neither amplification nor polysomy of ALK was observed in GSC, but weak overexpression was detected by IHC in three of nine GSCs. Similarly, no MET amplification was found by FISH but three GSCs presented significant immunohistochemical staining. No ALK or MET mutation was found by Sanger's direct sequencing. In this study, we show no molecular rearrangement of ALK, ROS1, and MET that would lead us not to propose, as a valid strategy, the use of crizotinib to eradicate GSCs. However, MET was overexpressed in all GSCs with mesenchymal subtype and three GSCs presented an overexpression of ALK. Therefore, our study corroborates the idea that MET and ALK may assume a role in the tumorigenicity of GSC.
Collapse
Affiliation(s)
- Audelaure Junca
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,Department of Pathology, University Hospital of Poitiers, Poitiers, F-86021, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Claire Villalva
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Gaëlle Tachon
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Pierre Rivet
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Ulrich Cortes
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Karline Guilloteau
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Anaïs Balbous
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Julie Godet
- Department of Pathology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Michel Wager
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France.,Department of Neurosurgery, University of Poitiers, Poitiers, F-86021, France
| | - Lucie Karayan-Tapon
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| |
Collapse
|
17
|
Lai YJ, Tsai JC, Tseng YT, Wu MS, Liu WS, Lam HI, Yu JH, Nozell SE, Benveniste EN. Small G protein Rac GTPases regulate the maintenance of glioblastoma stem-like cells in vitro and in vivo. Oncotarget 2017; 8:18031-18049. [PMID: 28160553 PMCID: PMC5392305 DOI: 10.18632/oncotarget.14949] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/03/2017] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma is the most common and aggressive malignant brain tumor in adults. The existence of glioblastoma stem cells (GSCs) or stem-like cells (stemloids) may account for its invasiveness and high recurrence. Rac proteins belong to the Rho small GTPase subfamily which regulates cell movement, proliferation, and survival. To investigate whether Rac proteins can serve as therapeutic targets for glioblastoma, especially for GSCs or stemloids, we examined the potential roles of Rac1, Rac2 and Rac3 on the properties of tumorspheres derived from glioblastoma cell lines. Tumorspheres are thought to be glioblastoma stem-like cells. We showed that Rac proteins promote the STAT3 and ERK activation and enhance cell proliferation and colony formation of glioblastoma stem-like cells. Knockdown of Rac proteins reduces the expression of GSC markers, such as CD133 and Sox2. The in vivo effects of Rac proteins in glioblastoma were further studied in zebrafish and in the mouse xenotransplantation model. Knocking-down Rac proteins abolished the angiogenesis effect induced by the injected tumorspheres in zebrafish model. In the CD133+-U373-tumorsphere xenotransplanted mouse model, suppression of Rac proteins decreased the incidence of tumor formation and inhibited the tumor growth. Moreover, knockdown of Rac proteins reduced the sphere forming efficiency of cells derived from these tumors. In conclusion, not only Rac1 but also Rac2 and 3 are important for glioblastoma tumorigenesis and can serve as the potential therapeutic targets against glioblastoma and its stem-like cells.
Collapse
Affiliation(s)
- Yun-Ju Lai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ying-Ting Tseng
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Meng-Shih Wu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Shan Liu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hoi-Ian Lam
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jei-Hwa Yu
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, AL, USA
| | - Susan E. Nozell
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, AL, USA
| | - Etty N. Benveniste
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
18
|
Marsecano C, Bruno F, Michelini G, Perri M, Calvisi G, Di Cesare E, Splendiani A. Systemic metastases from central nervous system ependymoma: case report and review of the literature. Neuroradiol J 2017. [PMID: 28627990 DOI: 10.1177/1971400916689371] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ependymal tumours in adults are rare, accounting for less than 4% of primary tumours of the central nervous system, and exceptionally metastasise outside the nervous system. In this study, we present a case of anaplastic ependymoma, which developed metastases outside the nervous system less than a year after its clinical onset. A healthy 65-year-old woman suddenly presented with drowsiness of unknown origin, accompanied by ingravescent fatigue, inability to maintain the upright posture, headache, nausea and vomiting. Computed tomography study performed in the emergency department showed the presence of an inhomogeneously hypodense area, with temporoparietal extension and median line deviation. After surgical excision, performed after two days, the bioptic examination demonstrated an anaplastic ependymoma with multiple areas of necrosis. The patient was submitted to adjuvant radiation therapy. At magnetic resonance imaging follow-up, performed three months after surgery, neither local recurrences nor typical 'drop metastases' to the spinal area were observed. Three months later, magnetic resonance imaging control revealed bone metastases and recurrences in the left insula and at the ipsilateral cerebellar hemisphere. Total body computed tomography examination showed metastases in the liver, vertebrae and pelvic bones, and involvement of paratracheal lymph nodes.
Collapse
Affiliation(s)
- Claudia Marsecano
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| | - Federico Bruno
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| | - Giulia Michelini
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| | - Marco Perri
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| | - Giuseppe Calvisi
- 2 Department of Pathology, S. Salvatore Hospital L'Aquila, L'Aquila, Italy
| | - Ernesto Di Cesare
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| | - Alessandra Splendiani
- 1 Department of Biotechnological and Applied Clinical Science, Division of Radiology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
19
|
Spina R, Voss DM, Asnaghi L, Sloan A, Bar EE. Atracurium Besylate and other neuromuscular blocking agents promote astroglial differentiation and deplete glioblastoma stem cells. Oncotarget 2016; 7:459-72. [PMID: 26575950 PMCID: PMC4808011 DOI: 10.18632/oncotarget.6314] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 10/29/2015] [Indexed: 01/11/2023] Open
Abstract
Glioblastoma multiforme (GBM) are the most common primary malignant brain tumor in adults, with a median survival of about one year. This poor prognosis is attributed primarily to therapeutic resistance and tumor recurrence after surgical removal, with the root cause suggested to be found in glioblastoma stem cells (GSCs). Using glial fibrillary acidic protein (GFAP) as a reporter of astrocytic differentiation, we isolated multiple clones from three independent GSC lines which express GFAP in a remarkably stable fashion. We next show that elevated expression of GFAP is associated with reduced clonogenicity in vitro and tumorigenicity in vivo. Utilizing this in vitro cell-based differentiation reporter system we screened chemical libraries and identified the non-depolarizing neuromuscular blocker (NNMB), Atracurium Besylate, as a small molecule which effectively induces astroglial but not neuronal differentiation of GSCs. Functionally, Atracurium Besylate treatment significantly inhibited the clonogenic capacity of several independent patient-derived GSC neurosphere lines, a phenomenon which was largely irreversible. A second NNMB, Vecuronium, also induced GSC astrocytic differentiation while Dimethylphenylpiperazinium (DMPP), a nicotinic acetylcholine receptor (nAChR) agonist, significantly blocked Atracurium Besylate pro-differentiation activity. To investigate the clinical importance of nAChRs in gliomas, we examined clinical outcomes and found that glioma patients with tumors overexpressing CHRNA1 or CHRNA9 (encoding for the AChR-α1 or AChR-α9) exhibit significant shorter overall survival. Finally, we found that ex-vivo pre-treatment of GSCs, expressing CHRNA1 and CHRNA9, with Atracurium Besylate significantly increased the survival of mice xenotransplanted with these cells, therefore suggesting that tumor initiating subpopulations have been reduced.
Collapse
Affiliation(s)
- Raffaella Spina
- Department of Neurological Surgery, Case Western Reserve University School of Medicine and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Dillon M Voss
- Department of Neurological Surgery, Case Western Reserve University School of Medicine and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Laura Asnaghi
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Andrew Sloan
- Department of Neurological Surgery, Case Western Reserve University School of Medicine and Case Comprehensive Cancer Center, Cleveland, OH, USA.,Department of Neurological Surgery, University Hospital-Case Medical Center, Case Comprehensive Cancer Center, and Case Western Reserve University, Cleveland, OH, USA
| | - Eli E Bar
- Department of Neurological Surgery, Case Western Reserve University School of Medicine and Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
20
|
Oliveira-Nunes MC, Assad Kahn S, de Oliveira Barbeitas AL, E Spohr TCLDS, Dubois LGF, Ventura Matioszek GM, Querido W, Campanati L, de Brito Neto JM, Lima FRS, Moura-Neto V, Carneiro K. The availability of the embryonic TGF-β protein Nodal is dynamically regulated during glioblastoma multiforme tumorigenesis. Cancer Cell Int 2016; 16:46. [PMID: 27330409 PMCID: PMC4912793 DOI: 10.1186/s12935-016-0324-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/07/2016] [Indexed: 12/19/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary brain tumor presenting self-renewing cancer stem cells. The role of these cells on the development of the tumors has been proposed to recapitulate programs from embryogenesis. Recently, the embryonic transforming growth factor-β (TGF-β) protein Nodal has been shown to be reactivated upon tumor development; however, its availability in GBM cells has not been addressed so far. In this study, we investigated by an original approach the mechanisms that dynamically control both intra and extracellular Nodal availability during GBM tumorigenesis. Methods We characterized the dynamics of Nodal availability in both stem and more differentiated GBM cells through morphological analysis, immunofluorescence of Nodal protein and of early (EEA1 and Rab5) and late (Rab7 and Rab11) endocytic markers and Western Blot. Tukey’s test was used to analyze the prevalent correlation of Nodal with different endocytic markers inside specific differentiation states, and Sidak’s multiple comparisons test was used to compare the prevalence of Nodal/endocytic markers co-localization between two differentiation states of GBM cells. Paired t test was used to analyze the abundance of Nodal protein, in extra and intracellular media. Results The cytoplasmic distribution of Nodal was dynamically regulated and strongly correlated with the differentiation status of GBM cells. While Nodal-positive vesicle-like particles were symmetrically distributed in GBM stem cells (GBMsc), they presented asymmetric perinuclear localization in more differentiated GBM cells (mdGBM). Strikingly, when subjected to dedifferentiation, the distribution of Nodal in mdGBM shifted to a symmetric pattern. Moreover, the availability of both intracellular and secreted Nodal were downregulated upon GBMsc differentiation, with cells becoming elongated, negative for Nodal and positive for Nestin. Interestingly, the co-localization of Nodal with endosomal vesicles also depended on the differentiation status of the cells, with Nodal seen more packed in EEA1/Rab5 + vesicles in GBMsc and more in Rab7/11 + vesicles in mdGBM. Conclusions Our results show for the first time that Nodal availability relates to GBM cell differentiation status and that it is dynamically regulated by an endocytic pathway during GBM tumorigenesis, shedding new light on molecular pathways that might emerge as putative targets for Nodal signaling in GBM therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12935-016-0324-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Cecília Oliveira-Nunes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Suzana Assad Kahn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, 265 Campus Drive, Stanford, California 94305 USA ; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Ana Luiza de Oliveira Barbeitas
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Luiz Gustavo Feijó Dubois
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Grasiella Maria Ventura Matioszek
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, B1-29, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - William Querido
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-30, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Loraine Campanati
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - José Marques de Brito Neto
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Flavia Regina Souza Lima
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| | - Vivaldo Moura-Neto
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F1-20, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil ; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156, Rio de Janeiro, Rio de Janeiro 20231-092 Brazil
| | - Katia Carneiro
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, F2-01, Rio de Janeiro, Rio de Janeiro 21941-902 Brazil
| |
Collapse
|
21
|
Pérez-Ramírez M, Hernández-Jiménez AJ, Guerrero-Guerrero A, Benadón-Darszon E, Pérezpeña-Díazconti M, Siordia-Reyes AG, García-Méndez A, de León FCP, Salamanca-Gómez FA, García-Hernández N. Genomics and epigenetics: A study of ependymomas in pediatric patients. Clin Neurol Neurosurg 2016; 144:53-8. [PMID: 26971296 DOI: 10.1016/j.clineuro.2016.02.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/22/2016] [Accepted: 02/28/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We identify chromosomal alterations, the methylation pattern and gene expression changes in pediatric ependymomas. METHODS CGH microarray, methylation and gene expression were performed through the Agilent platform. The results were analyzed with the software MatLab, MapViewer, DAVID, GeneCards and Hippie. RESULTS Amplification was found in 14q32.33, 2p22.3 and 8p22, and deletion was found in 8p11.23-p11.22 and 1q21.3. We observed 42.387 CpG islands with changes in their methylation pattern, in which we found 272 genes involved in signaling pathways related to carcinogenesis. We found 481 genes with altered expression. The genes IMMT, JHDMD1D, ASAH1, ZWINT, IPO7, GNAO1 and CISD3 were found to be altered among the three levels. CONCLUSION The 2p22.3, 8p11.23-p11.22 and 14q32.33 regions were identified as the most important; the changes in the methylation pattern related to cell cycle and cancer genes occurred in MIB2, FGF18 and ITIH5. The IPO7, GNAO1 and ASAH1 genes may play a major role in ependymoma development.
Collapse
Affiliation(s)
- Monserrat Pérez-Ramírez
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría "Dr. Silvestre Frenk Freud", Centro Médico Nacional "Siglo XXI", IMSS, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc, 06720 México D. F., Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Avenida Ciudad Universitaria 3000, Coyoacán, 04360 México D.F., Mexico
| | - Alejo Justino Hernández-Jiménez
- Servicio de Neurocirugía Pediátrica, Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional "La Raza", IMSS, Calzada Vallejo y Jacarandas S/N, Col. La Raza, Del. Azcapotzalco, 02980 Mexico D.F, Mexico
| | - Armando Guerrero-Guerrero
- Servicio de Neurocirugía Pediátrica, Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional "La Raza", IMSS, Calzada Vallejo y Jacarandas S/N, Col. La Raza, Del. Azcapotzalco, 02980 Mexico D.F, Mexico
| | - Eduardo Benadón-Darszon
- Departamento de Pediatría Ambulatoria, Hospital Infantil de México "Federico Gómez", Dr. Márquez 162, Col. Doctores, Del. Cuauhtémoc, 06720 México D.F, Mexico
| | - Mario Pérezpeña-Díazconti
- Departamento de Patología, Hospital Infantil de México "Federico Gómez", Dr. Márquez 162, Col. Doctores, Del. Cuauhtémoc, 06720 México D.F., Mexico
| | - Alicia Georgina Siordia-Reyes
- Servicio de Patología, Hospital de Pediatría "Dr. Silvestre Frenk Freud", Centro Médico Nacional "Siglo XXI", IMSS, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc, 06720 México D.F., Mexico
| | - Antonio García-Méndez
- Servicio de Neurocirugía Pediátrica, Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional "La Raza", IMSS, Calzada Vallejo y Jacarandas S/N, Col. La Raza, Del. Azcapotzalco, 02980 Mexico D.F, Mexico
| | - Fernando Chico-Ponce de León
- Departamento de Neurocirugía, Hospital Infantil de México "Federico Gómez", Dr. Márquez 162, Col. Doctores, Del. Cuauhtémoc, 06720 México D.F., Mexico
| | - Fabio Abdel Salamanca-Gómez
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría "Dr. Silvestre Frenk Freud", Centro Médico Nacional "Siglo XXI", IMSS, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc, 06720 México D. F., Mexico
| | - Normand García-Hernández
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría "Dr. Silvestre Frenk Freud", Centro Médico Nacional "Siglo XXI", IMSS, Av. Cuauhtémoc 330, Col. Doctores, Del. Cuauhtémoc, 06720 México D. F., Mexico.
| |
Collapse
|
22
|
Li R, Li Y, Hu X, Lian H, Wang L, Fu H. Transcription factor 3 controls cell proliferation and migration in glioblastoma multiforme cell lines. Biochem Cell Biol 2016; 94:247-55. [PMID: 27105323 DOI: 10.1139/bcb-2015-0162] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Transcription factor 3 (TCF3) is a member of the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor family. Recent studies have demonstrated its potential carcinogenic properties. Here we show that TCF3 was upregulated in glioma tissues compared with normal brain tissues. This upregulation of the TCF3 gene probably has functional significance in brain-tumor progression. Our studies on glioblastoma multiforme (GBM) cell lines show that knock-down of TCF3 induced apoptosis and inhibited cell migration. Further analysis revealed that down-regulation of TCF3 gene expression inhibits Akt and Erk1/2 activation, suggesting that the carcinogenic properties of TCF3 in GBM are partially mediated by the phosphatidylinositol 3-kinase-Akt and MAPK-Erk signaling pathways. Considered together, the results of this study demonstrate that high levels of TCF3 in gliomas potentially promote glioma development through the Akt and Erk pathways.
Collapse
Affiliation(s)
- Ruiting Li
- a Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, Hubei, China
| | - Yinghui Li
- a Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, Hubei, China
| | - Xin Hu
- a Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, Hubei, China
| | - Haiwei Lian
- a Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, Hubei, China
| | - Lei Wang
- b Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Hui Fu
- a Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, Hubei, China
| |
Collapse
|
23
|
Ma J, Meng F, Li S, Liu L, Zhao L, Liu Y, Hu Y, Li Z, Yao Y, Xi Z, Teng H, Xue Y. Autophagy Induction by Endothelial-Monocyte Activating Polypeptide II Contributes to the Inhibition of Malignant Biological Behaviors by the Combination of EMAP II with Rapamycin in Human Glioblastoma. Front Mol Neurosci 2015; 8:74. [PMID: 26648842 PMCID: PMC4664732 DOI: 10.3389/fnmol.2015.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 11/20/2015] [Indexed: 12/19/2022] Open
Abstract
This study aims to investigate the effect of endothelial-monocyte activating polypeptide II (EMAP II) on human glioblastoma (GBM) cells and glioblastoma stem cells (GSCs) as well as its possible mechanisms. In this study, EMAP II inhibited the cell viability and decreased the mitochondrial membrane potential in human GBM cells and GSCs, and autophagy inhibitor 3-methyl adenine (3-MA) blocked these effects. Autophagic vacuoles were formed in these cells after EMAP II treatment and this phenomenon was blocked by 3-MA. In addition, the up-regulation of microtubule-associated protein-1 light chain-3 (LC3)-II and the down-regulation of autophagic degraded substrate p62/SQSTM1 caused by EMAP II were observed. Cells treated with EMAP-II inhibited the PI3K/Akt/mTOR signal pathway, and PI3K/Akt agonist insulin-like growth factor-1 (IGF-1) blocked the effect of EMAP II on the expression of LC3-II and p62/SQSTM1. Cells exposed to EMAP-II experienced mitophagy and ER stress. Furthermore, the inhibition of cell proliferation, migration and invasion of GBM cells and GSCs were more remarkable by the combination of EMAP II and rapamycin than either agent alone in vitro and in vivo. The current study demonstrated that the cytotoxicity of EMAP II in human GBM cells and GSCs was induced by autophagy, accompanied by the inhibition of PI3K/Akt/mTOR signal pathway, mitophagy and ER stress. The combination of EMAP II with rapamycin demonstrated the inhibitory effect on the malignant biological behaviors of human GBM cells and GSCs in vitro and in vivo.
Collapse
Affiliation(s)
- Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University Shenyang, China ; Institute of Pathology and Pathophysiology, China Medical University Shenyang, China
| | - Fanjie Meng
- Department of Neurobiology, College of Basic Medicine, China Medical University Shenyang, China ; Institute of Pathology and Pathophysiology, China Medical University Shenyang, China
| | - Shuai Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University Shenyang, China ; Institute of Pathology and Pathophysiology, China Medical University Shenyang, China
| | - Lini Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University Shenyang, China ; Institute of Pathology and Pathophysiology, China Medical University Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Yi Hu
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Yilong Yao
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Zhuo Xi
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Hao Teng
- Department of Neurosurgery, Shengjing Hospital of China Medical University Shenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University Shenyang, China ; Institute of Pathology and Pathophysiology, China Medical University Shenyang, China
| |
Collapse
|
24
|
WU ZHIWU, HAN YONG, LI YANYAN, LI XUETAO, SUN TING, CHEN GUILIN, HUANG YULUN, ZHOU YOUXIN, DU ZIWEI. miR-218-5p inhibits the stem cell properties and invasive ability of the A2B5+CD133− subgroup of human glioma stem cells. Oncol Rep 2015; 35:869-77. [DOI: 10.3892/or.2015.4418] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/23/2015] [Indexed: 11/06/2022] Open
|
25
|
Wang XF, Zhao ZF, Chen MH, Yuan QH, Li YL, Jiang CL. Epirubicin inhibits growth and alters the malignant phenotype of the U‑87 glioma cell line. Mol Med Rep 2015; 12:5917-23. [PMID: 26300546 DOI: 10.3892/mmr.2015.4220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 04/16/2015] [Indexed: 11/05/2022] Open
Abstract
Epirubicin, an anthracycline derivative, is one of the main line treatments for brain tumors. The aim of the present study was to verify that epirubicin alters the growth and morphological characteristics of U‑87 glioma cells. In the present study, the effects of epirubicin were tested using cellular and biochemical assays, which demonstrated its anti‑proliferative and cytotoxic effects, with an IC50 of 6.3 µM for the U‑87 cell line, while rat normal neuronal cells were resistant to epirubicin. Epirubicin also reduced the secretion of matrix metalloproteinase‑9 by 48 and 56% at concentrations of 2.5 and 5 µM, respectively. Exposure to epirubicin also diminished levels of vascular endothelial growth factor in U‑87 cells. Furthermore, a cell migration assay showed a significant decrease in cell migration from 28 to 59% following exposure to 1 µM epirubicin. The present study demonstrated the cytotoxic, anti‑proliferative and anti‑migrative potential of epirubicin against glioma cells in vitro.
Collapse
Affiliation(s)
- Xiao-Feng Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Zhe-Feng Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ming-Hui Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Qing-Hua Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yong-Li Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Chuan-Lu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
26
|
El Meskini R, Iacovelli AJ, Kulaga A, Gumprecht M, Martin PL, Baran M, Householder DB, Van Dyke T, Weaver Ohler Z. A preclinical orthotopic model for glioblastoma recapitulates key features of human tumors and demonstrates sensitivity to a combination of MEK and PI3K pathway inhibitors. Dis Model Mech 2015; 8:45-56. [PMID: 25431423 PMCID: PMC4283649 DOI: 10.1242/dmm.018168] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/18/2014] [Indexed: 12/25/2022] Open
Abstract
Current therapies for glioblastoma multiforme (GBM), the highest grade malignant brain tumor, are mostly ineffective, and better preclinical model systems are needed to increase the successful translation of drug discovery efforts into the clinic. Previous work describes a genetically engineered mouse (GEM) model that contains perturbations in the most frequently dysregulated networks in GBM (driven by RB, KRAS and/or PI3K signaling and PTEN) that induce development of Grade IV astrocytoma with properties of the human disease. Here, we developed and characterized an orthotopic mouse model derived from the GEM that retains the features of the GEM model in an immunocompetent background; however, this model is also tractable and efficient for preclinical evaluation of candidate therapeutic regimens. Orthotopic brain tumors are highly proliferative, invasive and vascular, and express histology markers characteristic of human GBM. Primary tumor cells were examined for sensitivity to chemotherapeutics and targeted drugs. PI3K and MAPK pathway inhibitors, when used as single agents, inhibited cell proliferation but did not result in significant apoptosis. However, in combination, these inhibitors resulted in a substantial increase in cell death. Moreover, these findings translated into the in vivo orthotopic model: PI3K or MAPK inhibitor treatment regimens resulted in incomplete pathway suppression and feedback loops, whereas dual treatment delayed tumor growth through increased apoptosis and decreased tumor cell proliferation. Analysis of downstream pathway components revealed a cooperative effect on target downregulation. These concordant results, together with the morphologic similarities to the human GBM disease characteristics of the model, validate it as a new platform for the evaluation of GBM treatment.
Collapse
Affiliation(s)
- Rajaa El Meskini
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Anthony J Iacovelli
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Alan Kulaga
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Michelle Gumprecht
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Philip L Martin
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Maureen Baran
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Deborah B Householder
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Terry Van Dyke
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA. Mouse Cancer Genetics Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Zoë Weaver Ohler
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| |
Collapse
|
27
|
Balça-Silva J, Matias D, do Carmo A, Girão H, Moura-Neto V, Sarmento-Ribeiro AB, Lopes MC. Tamoxifen in combination with temozolomide induce a synergistic inhibition of PKC-pan in GBM cell lines. Biochim Biophys Acta Gen Subj 2014; 1850:722-32. [PMID: 25554223 DOI: 10.1016/j.bbagen.2014.12.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly proliferative, angiogenic grade IV astrocytoma that develops resistance to the alkylating agents used in chemotherapy, such as temozolomide (TMZ), which is considered the gold standard. The mean survival time for GBM patients is approximately 12 months, increasing to 14.6 months after TMZ treatment. The resistance of GBM to chemotherapy seems to be associated to genetic alterations and to the constitutive activation of several signaling pathways. Therefore, the combination of different drugs with different mechanisms of action may contribute to circumvent the chemoresistance of glioma cells. Here we describe the potential synergistic behavior of the therapeutic combination of tamoxifen (TMX), a known inhibitor of PKC, and TMZ in GBM. METHODS We used two GBM cell lines incubated in absence and presence of TMX and/or TMZ and measured cell viability, proliferation, apoptosis, cell cycle, migration ability, cytoskeletal organization and the phosphorylated amount of the p-PKC-pan. RESULTS The combination of low doses of TMX with increasing doses of TMZ shows an increased antiproliferative and apoptotic effect compared to the effect with TMX alone. CONCLUSIONS The combination of TMX and TMZ seems to potentiate the effect of each other. These alterations seem to be associated to a decrease in the phosphorylation status of PKC. GENERAL SIGNIFICANCE We emphasize that TMX is an inhibitor of the p-PKC-pan and that these combination is more effective in the reduction of proliferation and in the increase of apoptosis than each drug alone, which presents a new therapeutic strategy in GBM treatment.
Collapse
Affiliation(s)
- Joana Balça-Silva
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Diana Matias
- Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rio de Janeiro, Brazil.
| | - Anália do Carmo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Henrique Girão
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center of Ophthalmology and Vision Sciences, Institute of Biomedical Imaging and Life Sciences (IBILI), Portugal.
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rio de Janeiro, Brazil.
| | - Ana Bela Sarmento-Ribeiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal; Hematology Department, Centro Hospitalar Universitário de Coimbra (CHUC), Portugal.
| | - Maria Celeste Lopes
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
28
|
Involvement of the neural stem cell compartment by pediatric and adult gliomas: a retrospective review of 377 cases. J Neurooncol 2014; 122:105-10. [DOI: 10.1007/s11060-014-1682-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 12/02/2014] [Indexed: 12/22/2022]
|
29
|
Yao Y, Ma J, Xue Y, Wang P, Li Z, Li Z, Hu Y, Shang X, Liu Y. MiR-449a exerts tumor-suppressive functions in human glioblastoma by targeting Myc-associated zinc-finger protein. Mol Oncol 2014; 9:640-56. [PMID: 25487955 DOI: 10.1016/j.molonc.2014.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is one of the most common and aggressive primary brain tumors in adults. Deregulated expression of microRNAs (miRNAs) has been associated with GBM progression through alterations in either oncogenic or tumor suppressor targets. Here, we elucidated the function and the possible molecular mechanisms of miR-449a in human GBM cell lines and tumor specimens-derived glioblastoma stem cells (GSCs). Quantitative real-time PCR demonstrated that miR-449a was down-regulated in human GBM cell lines and GSCs. Functionally, miR-449a acted as a tumor suppressor by reducing cell proliferation, migration and invasion as well as inducing apoptosis in human GBM cell lines and GSCs. Myc-associated zinc-finger protein (MAZ) was identified as a direct target of miR-449a, mediating these tumor-suppressive effects, demonstrated by Western blot assay and luciferase assays. Moreover, over-expression of miR-449a inhibited the expression of Podoplanin (PDPN) by down-regulating MAZ which could positively control the promoter activities via binding to the promoter of PDPN, demonstrated by luciferase assays and chromatin immunoprecipitation assays. Further, the PI3K/AKT pathway was blocked when MAZ was down-regulated by miR-449a. This process was coincided with the up-regulation of apoptotic proteins and the down-regulation of anti-apoptotic proteins, MMP2 and MMP9. Furthermore, nude mice carrying over-expressed miR-449a combined with knockdown MAZ tumors produced the smallest tumors and the highest survival. These results elucidated a novel molecular mechanism of GBM progression, and may thus suggest a promising application for GBM treatment.
Collapse
Affiliation(s)
- Yilong Yao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Zhiqing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yi Hu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiuli Shang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
30
|
Marfia G, Campanella R, Navone SE, Di Vito C, Riccitelli E, Hadi LA, Bornati A, de Rezende G, Giussani P, Tringali C, Viani P, Rampini P, Alessandri G, Parati E, Riboni L. Autocrine/paracrine sphingosine-1-phosphate fuels proliferative and stemness qualities of glioblastoma stem cells. Glia 2014; 62:1968-81. [PMID: 25042636 DOI: 10.1002/glia.22718] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 11/08/2022]
Abstract
Accumulating reports suggest that human glioblastoma contains glioma stem-like cells (GSCs) which act as key determinants driving tumor growth, angiogenesis, and contributing to therapeutic resistance. The proliferative signals involved in GSC proliferation and progression remain unclear. Using GSC lines derived from human glioblastoma specimens with different proliferative index and stemness marker expression, we assessed the hypothesis that sphingosine-1-phosphate (S1P) affects the proliferative and stemness properties of GSCs. The results of metabolic studies demonstrated that GSCs rapidly consume newly synthesized ceramide, and export S1P in the extracellular environment, both processes being enhanced in the cells exhibiting high proliferative index and stemness markers. Extracellular S1P levels reached nM concentrations in response to increased extracellular sphingosine. In addition, the presence of EGF and bFGF potentiated the constitutive capacity of GSCs to rapidly secrete newly synthesized S1P, suggesting that cooperation between S1P and these growth factors is of central importance in the maintenance and proliferation of GSCs. We also report for the first time that S1P is able to act as a proliferative and pro-stemness autocrine factor for GSCs, promoting both their cell cycle progression and stemness phenotypic profile. These results suggest for the first time that the GSC population is critically modulated by microenvironmental S1P, this bioactive lipid acting as an autocrine signal to maintain a pro-stemness environment and favoring GSC proliferation, survival and stem properties.
Collapse
Affiliation(s)
- Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico Milan, University of Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chen G, Kong J, Tucker-Burden C, Anand M, Rong Y, Rahman F, Moreno CS, Van Meir EG, Hadjipanayis CG, Brat DJ. Human Brat ortholog TRIM3 is a tumor suppressor that regulates asymmetric cell division in glioblastoma. Cancer Res 2014; 74:4536-48. [PMID: 24947043 DOI: 10.1158/0008-5472.can-13-3703] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer stem cells, capable of self-renewal and multipotent differentiation, influence tumor behavior through a complex balance of symmetric and asymmetric cell divisions. Mechanisms regulating the dynamics of stem cells and their progeny in human cancer are poorly understood. In Drosophila, mutation of brain tumor (brat) leads to loss of normal asymmetric cell division by developing neural cells and results in a massively enlarged brain composed of neuroblasts with neoplastic properties. Brat promotes asymmetric cell division and directs neural differentiation at least partially through its suppression on Myc. We identified TRIM3 (11p15.5) as a human ortholog of Drosophila brat and demonstrate its regulation of asymmetric cell division and stem cell properties of glioblastoma (GBM), a highly malignant human brain tumor. TRIM3 gene expression is markedly reduced in human GBM samples, neurosphere cultures, and cell lines and its reconstitution impairs growth properties in vitro and in vivo. TRIM3 expression attenuates stem-like qualities of primary GBM cultures, including neurosphere formation and the expression of stem cell markers CD133, Nestin, and Nanog. In GBM stem cells, TRIM3 expression leads to a greater percentage dividing asymmetrically rather than symmetrically. As with Brat in Drosophila, TRIM3 suppresses c-Myc expression and activity in human glioma cell lines. We also demonstrate a strong regulation of Musashi-Notch signaling by TRIM3 in GBM neurospheres and neural stem cells that may better explain its effect on stem cell dynamics. We conclude that TRIM3 acts as a tumor suppressor in GBM by restoring asymmetric cell division.
Collapse
Affiliation(s)
- Gang Chen
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Jun Kong
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia
| | - Carol Tucker-Burden
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Monika Anand
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Yuan Rong
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Fahmia Rahman
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia. Department of Biomedical Informatics, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Erwin G Van Meir
- Department of Neurosurgery, Emory University, Atlanta, Georgia. Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Constantinos G Hadjipanayis
- Department of Neurosurgery, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Daniel J Brat
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia. Department of Biomedical Informatics, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia.
| |
Collapse
|
32
|
Tumor suppressive miRNA-34a suppresses cell proliferation and tumor growth of glioma stem cells by targeting Akt and Wnt signaling pathways. FEBS Open Bio 2014; 4:485-95. [PMID: 24944883 PMCID: PMC4060015 DOI: 10.1016/j.fob.2014.05.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/07/2014] [Accepted: 05/10/2014] [Indexed: 12/03/2022] Open
Abstract
miR-34a was decreased in both glioma and glioma stem cell-lines as compared to normal brain tissues. Glioma stem cell-lines HNGC-2 and NSG-K16 possess the mesenchymal glioblastoma phenotype. miR-34a over-expression in these cell lines decreased their proliferative and migratory potential, and induced apoptosis. Rictor, a part of the mTORC2 complex, is a novel target for miR-34a in glioma stem cells. The tumor suppressive function of miR-34a is mediated via Rictor and affects the AKT/mTOR pathway and Wnt signaling.
MiRNA-34a is considered as a potential prognostic marker for glioma, as studies suggest that its expression negatively correlates with patient survival in grade III and IV glial tumors. Here, we show that expression of miR-34a was decreased in a graded manner in glioma and glioma stem cell-lines as compared to normal brain tissues. Ectopic expression of miR-34a in glioma stem cell-lines HNGC-2 and NSG-K16 decreased the proliferative and migratory potential of these cells, induced cell cycle arrest and caused apoptosis. Notably, the miR-34a glioma cells formed significantly smaller xenografts in immuno-deficient mice as compared with control glioma stem cell-lines. Here, using a bioinformatics approach and various biological assays, we identify Rictor, as a novel target for miR-34a in glioma stem cells. Rictor, a defining component of mTORC2 complex, is involved in cell survival signaling. mTORC2 lays downstream of Akt, and thus is a direct activator of Akt. Our earlier studies have elaborated on role of Rictor in glioma invasion (Das et al., 2011). Here, we demonstrate that miR34a over-expression in glioma stem cells profoundly decreased levels of p-AKT (Ser473), increased GSK-3β levels and targeted for degradation β-catenin, an important mediator of Wnt signaling pathway. This led to diminished levels of the Wnt effectors cyclin D1 and c-myc. Collectively, we show that the tumor suppressive function of miR-34a in glioblastoma is mediated via Rictor, which through its effects on AKT/mTOR pathway and Wnt signaling causes pronounced effects on glioma malignancy.
Collapse
Key Words
- Beta-catenin
- CNS, central nervous system
- EGF, epidermal growth factor
- EMT, epithelial–mesenchymal transition
- EV, empty vector
- GBM, glioblastoma multiforme
- GIC, glioma initiating cell
- GSC, glioma stem cell
- GSK-3β, glycogen synthase kinase 3β
- Glioblastoma
- Heterogeneity
- Mesenchymal
- NOD/SCID, nonobese diabetic/severe combined immunodeficiency
- PARP, poly ADP-ribose polymerases
- PDGFRA, platelet-derived growth factor receptor-α
- Rictor
- TCGA, the cancer genome atlas database
- bFGF, basic fibroblast growth factor
- qRT-PCR, quantitative real time PCR
Collapse
|
33
|
The role of targeted therapies in the management of progressive glioblastoma. J Neurooncol 2014; 118:557-99. [DOI: 10.1007/s11060-013-1339-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/28/2013] [Indexed: 12/28/2022]
|
34
|
Meco D, Servidei T, Lamorte G, Binda E, Arena V, Riccardi R. Ependymoma stem cells are highly sensitive to temozolomide in vitro and in orthotopic models. Neuro Oncol 2014; 16:1067-77. [PMID: 24526307 DOI: 10.1093/neuonc/nou008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Ependymoma management remains challenging because of the inherent chemoresistance of this tumor. To determine whether ependymoma stem cells (SCs) might contribute to therapy resistance, we investigated the sensitivity of ependymoma SCs to temozolomide and etoposide. METHODS The efficacies of the two DNA damaging agents were explored in two ependymoma SC lines in vitro and in vivo models. RESULTS Ependymoma SC lines were highly sensitive to temozolomide and etoposide in vitro, but only temozolomide impaired tumor-initiation properties. Consistently, temozolomide but not etoposide showed significant antitumoral activity on ependymoma SC-driven subcutaneous and orthotopic xenografts by reducing the mitotic fraction. In vitro temozolomide at the EC50 (10 µM) induced accumulation of cells in the G2/M phase that was unexpectedly accompanied by downregulation of p27 and p21 without modulation of full-length p53 (FLp53). Differentiation-committed ependymoma SCs acquired resistance to temozolomide. Inhibition of proliferation was partly due to apoptosis, that occurred earlier in differentiated cells as compared to neurospheres. The activation of apoptosis correlated with an increase in p53β/γ isoforms without modulation of FLp53 under both serum-free and differentiation-promoting media. Incubation of cells in both conditions with temozolomide resulted in increased glioneuronal differentiation exhibiting elevated glial fibrillary acidic protein, galactosylceramidase, and βIII-tubulin expression compared to untreated controls. O(6)-methylguanine DNA methyltransferase (MGMT) transcript levels were very low in SCs, and were increased by treatment and, epigenetically, by differentiation through MGMT promoter unmethylation. CONCLUSION Ependymoma growth might be impaired by temozolomide through preferential depletion of a less differentiated, more tumorigenic, MGMT-negative cell population with stem-like properties.
Collapse
Affiliation(s)
- Daniela Meco
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| | - Tiziana Servidei
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| | - Giuseppe Lamorte
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| | - Elena Binda
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| | - Vincenzo Arena
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| | - Riccardo Riccardi
- Department of Pediatric Oncology, Catholic University, Rome, Italy (D.M., T.S., R.R.); Istituto CSS - Mendel Laboratory, Rome, Italy (G.L.); Department of Biotechnology and Biosciences, Building U3, University of Milan Bicocca, Milan, Italy (E.B.); Institute of Pathology, Catholic University, Rome, Italy (V.A.)
| |
Collapse
|
35
|
Katsushima K, Kondo Y. Non-coding RNAs as epigenetic regulator of glioma stem-like cell differentiation. Front Genet 2014; 5:14. [PMID: 24550934 PMCID: PMC3910277 DOI: 10.3389/fgene.2014.00014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/15/2014] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas show heterogeneous histological features. These distinct phenotypic states are thought to be associated with the presence of glioma stem cells (GSCs), which are highly tumorigenic and self-renewing sub-population of tumor cells that have different functional characteristics. Differentiation of GSCs may be regulated by multi-tiered epigenetic mechanisms that orchestrate the expression of thousands of genes. One such regulatory mechanism involves functional non-coding RNAs (ncRNAs), such as microRNAs (miRNAs); a large number of ncRNAs have been identified and shown to regulate the expression of genes associated with cell differentiation programs. Given the roles of miRNAs in cell differentiation, it is possible they are involved in the regulation of gene expression networks in GSCs that are important for the maintenance of the pluripotent state and for directing differentiation. Here, we review recent findings on ncRNAs associated with GSC differentiation and discuss how these ncRNAs contribute to the establishment of tissue heterogeneity during glioblastoma tumor formation.
Collapse
Affiliation(s)
- Keisuke Katsushima
- Division of Epigenomics, Aichi Cancer Center Research Institute, Nagoya Japan
| | - Yutaka Kondo
- Division of Epigenomics, Aichi Cancer Center Research Institute, Nagoya Japan
| |
Collapse
|
36
|
Sassi FDA, Caesar L, Jaeger M, Nör C, Abujamra AL, Schwartsmann G, de Farias CB, Brunetto AL, Lopez PLDC, Roesler R. Inhibitory activities of trichostatin a in U87 glioblastoma cells and tumorsphere-derived cells. J Mol Neurosci 2014; 54:27-40. [PMID: 24464841 DOI: 10.1007/s12031-014-0241-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/14/2014] [Indexed: 12/11/2022]
Abstract
Epigenetic alterations have been increasingly implicated in glioblastoma (GBM) pathogenesis, and epigenetic modulators including histone deacetylase inhibitors (HDACis) have been investigated as candidate therapies. GBMs are proposed to contain a subpopulation of glioblastoma stem cells (GSCs) that sustain tumor progression and therapeutic resistance and can form tumorspheres in culture. Here, we investigate the effects of the HDACi trichostatin A (TSA) in U87 GBM cultures and tumorsphere-derived cells. Using approaches that include a novel method to measure tumorsphere sizes and the area covered by spheres in GBM cultures, as well as a nuclear morphometric analysis, we show that TSA reduced proliferation and colony sizes, led to G2/M arrest, induced alterations in nuclear morphology consistent with cell senescence, and increased the protein content of GFAP, but did not affect migration, in cultured human U87 GBM cells. In cells expanded in tumorsphere assays, TSA reduced sphere formation and induced neuron-like morphological changes. The expression of stemness markers in these cells was detected by reverse transcriptase polymerase chain reaction. These findings indicate that HDACis can inhibit proliferation, survival, and tumorsphere formation, and promote differentiation of U87 GBM cells, providing further evidence for the development of HDACis as potential therapeutics against GBM.
Collapse
Affiliation(s)
- Felipe de Almeida Sassi
- Cancer Research Laboratory, University Hospital Research Center (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhou S, Wang F, Zhang Y, Johnson MR, Qian S, Wu M, Wu E. Salinomycin Suppresses PDGFRβ, MYC, and Notch Signaling in Human Medulloblastoma. AUSTIN JOURNAL OF PHARMACOLOGY AND THERAPEUTICS 2014; 2:1020. [PMID: 25478603 PMCID: PMC4251667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Medulloblastoma (MB) is the most common childhood brain tumor. Despite improved therapy and management, approximately 30% of patients die of the disease. To search for a more effective therapeutic strategy, the effects of salinomycin were tested on cell proliferation, cell death, and cell cycle progression in human MB cell lines. The results demonstrated that salinomycin inhibits cell proliferation, induces cell death , and disrupts cell cycle progression in MB cells. Salinomycin was also tested on the expression levels of key genes involved in proliferation and survival signaling and revealed that salinomycin down-regulates the expression of PDGFRβ, MYC, p21 and Bcl-2 as well as up-regulates the expression of cyclin A. In addition, the results reveal that salinomycin suppresses the expression of Hes1 and Hes5 in MB cells. Our data shed light on the potential of using salinomycin as a novel therapeutic agent for patients with MB.
Collapse
Affiliation(s)
- Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, USA
| | - Fengfei Wang
- Department of Pharmaceutical Sciences, North Dakota State University, USA
| | - Ying Zhang
- Department of Pharmaceutical Sciences, North Dakota State University, USA
| | - Max R Johnson
- Retina Consultants Ltd and University of North Dakota, USA
| | - Steven Qian
- Department of Pharmaceutical Sciences, North Dakota State University, USA
| | - Min Wu
- Department of Basic Sciences, University of North Dakota, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, USA
| |
Collapse
|
38
|
Alexander BM, Lee EQ, Reardon DA, Wen PY. Current and future directions for Phase II trials in high-grade glioma. Expert Rev Neurother 2013; 13:369-87. [PMID: 23545053 DOI: 10.1586/ern.12.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite surgery, radiation and chemotherapy, the prognosis for high-grade glioma (HGG) is poor. Our understanding of the molecular pathways involved in gliomagenesis and progression has increased in recent years, leading to the development of novel agents that specifically target these pathways. Results from most single-agent trials have been modest at best, however. Despite the initial success of antiangiogenesis agents in HGG, the clinical benefit is short-lived and most patients eventually progress. Several novel agents, multi-targeted agents and combination therapies are now in clinical trials for HGG and several more strategies are being pursued.
Collapse
Affiliation(s)
- Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
39
|
Chen L, Chen XR, Chen FF, Liu Y, Li P, Zhang R, Yan K, Yi YJ, Xu ZM, Jiang XD. MicroRNA-107 inhibits U87 glioma stem cells growth and invasion. Cell Mol Neurobiol 2013; 33:651-7. [PMID: 23572380 DOI: 10.1007/s10571-013-9927-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/12/2013] [Indexed: 10/27/2022]
Abstract
Glioma stem cells (GSCs) are thought to be critical for resistance to radiotherapy and chemotherapy and for tumor recurrence after surgery in glioma patients. Identification of new therapeutic strategies that can target GSCs may thus be critical for improving patient survival. MicroRNAs (miRNAs) are small non-coding RNAs that function as tumor suppressors or oncogenes. In this study, we confirmed that miR-107 was down-regulated in GSCs. To investigate the role of miR-107 in tumorigenesis of GSCs, a lentiviral vector over-expressing miR-107 in U87GSCs was constructed. We found that over-expression of miR-107 suppressed proliferation and down-regulated Notch2 protein and stem cell marker (CD133 and Nestin) expression in U87GSCs. Furthermore, enhanced miR-107 expression significantly inhibited U87GSC invasion and reduced matrix metalloproteinase-12 expression. miR-107 also suppressed U87GSCs xenograft growth in vivo. These findings suggest that miR-107 is involved in U87GSCs growth and invasion and may provide a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Lei Chen
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dominguez CL, Floyd DH, Xiao A, Mullins GR, Kefas BA, Xin W, Yacur MN, Abounader R, Lee JK, Wilson GM, Harris TE, Purow BW. Diacylglycerol kinase α is a critical signaling node and novel therapeutic target in glioblastoma and other cancers. Cancer Discov 2013; 3:782-97. [PMID: 23558954 DOI: 10.1158/2159-8290.cd-12-0215] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although diacylglycerol kinase α (DGKα) has been linked to several signaling pathways related to cancer cell biology, it has been neglected as a target for cancer therapy. The attenuation of DGKα activity via DGKα-targeting siRNA and small-molecule inhibitors R59022 and R59949 induced caspase-mediated apoptosis in glioblastoma cells and in other cancers, but lacked toxicity in noncancerous cells. We determined that mTOR and hypoxia-inducible factor-1α (HIF-1α) are key targets of DGKα inhibition, in addition to its regulation of other oncogenes. DGKα regulates mTOR transcription via a unique pathway involving cyclic AMP. Finally, we showed the efficacy of DGKα inhibition with short hairpin RNA or a small-molecule agent in glioblastoma and melanoma xenograft treatment models, with growth delay and decreased vascularity. This study establishes DGKα as a central signaling hub and a promising therapeutic target in the treatment of cancer.
Collapse
Affiliation(s)
- Charli L Dominguez
- Division of Neuro-Oncology, Department of Neurology, College of Nursing and Health Professions, University of Southern Indiana, Evansville, Indiana, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nör C, Sassi FA, de Farias CB, Schwartsmann G, Abujamra AL, Lenz G, Brunetto AL, Roesler R. The histone deacetylase inhibitor sodium butyrate promotes cell death and differentiation and reduces neurosphere formation in human medulloblastoma cells. Mol Neurobiol 2013; 48:533-43. [PMID: 23516101 DOI: 10.1007/s12035-013-8441-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/08/2013] [Indexed: 01/07/2023]
Abstract
Increasing evidence suggests that alterations in epigenetic mechanisms regulating chromatin state play a role in the pathogenesis of medulloblastoma (MB), the most common malignant brain tumor of childhood. Histone deacetylase (HDAC) inhibitors, which increase chromatin relaxation, have been shown to display anticancer activities. Here we show that the HDAC inhibitor sodium butyrate (NaB) markedly increases cell death and reduces colony formation in human MB cell lines. In addition, NaB increased the mRNA expression of Gria2, a neuronal differentiation marker, in D283 and DAOY cells and reduced the number of neurospheres in D283 cell cultures. Finally, NaB reduced the viability of D283 cells when combined with etoposide. These data show that NaB displays pronounced inhibitory effects on the survival of human MB cells and suggest that NaB might potentiate the effects of etoposide. In addition, our study suggests that HDAC inhibition might promote the neuronal differentiation of MB cells and provides the first evidence that an HDAC inhibitor might suppress the expansion or survival of MB cancer stem cells.
Collapse
Affiliation(s)
- Carolina Nör
- Cancer Research Laboratory, University Hospital Research Center (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
42
|
VEGF promotes proliferation of human glioblastoma multiforme stem-like cells through VEGF receptor 2. ScientificWorldJournal 2013; 2013:417413. [PMID: 23533349 PMCID: PMC3603324 DOI: 10.1155/2013/417413] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 02/03/2013] [Indexed: 12/24/2022] Open
Abstract
Cancer stem-like cells, which have been described as tumor-initiating cells or tumor-propagating cells, play a crucial role in our fundamental understanding of glioblastoma multiforme (GBM) and its recurrence. GBM is a lethal cancer, characterized by florid vascularization and aberrantly elevated vascular endothelial growth factor (VEGF). VEGF promotes tumorigenesis and angiogenesis of human GBM stem-like cells (GBSCs). However, whether and how VEGF contributes to GBSCs proliferation remain largely uncertain. In this study, human GBSCs were isolated from surgical specimens of glioblastoma and cultured in medium favored for stem cell growth. Neural Colony-Forming Cell Assay and ATP assay were performed to measure GBSC proliferation under normoxia (20% O2) and hypoxia (1% O2). Our observations demonstrate that exogenous VEGF stimulates GBSC proliferation in a dose-dependent manner via VEGF Receptor 2 (VEGFR2); while VEGF Receptor 1 (VEGFR1) has a negative feedback effect on VEGFR2 when cells were exposed to higher concentration of VEGF. These results suggest that suppressing VEGFR2-dependent GBSC proliferation is a potentially therapeutic strategy in GBM.
Collapse
|
43
|
Akt and c-Myc induce stem-cell markers in mature primary p53⁻/⁻ astrocytes and render these cells gliomagenic in the brain of immunocompetent mice. PLoS One 2013; 8:e56691. [PMID: 23424671 PMCID: PMC3570527 DOI: 10.1371/journal.pone.0056691] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/14/2013] [Indexed: 11/25/2022] Open
Abstract
Astrocytomas and their most malignant variant glioblastoma multiforme (GBM) represent the vast majority of primary brain tumors. Despite the current progress in neurosurgery, radiation therapy and chemotherapy, most astrocytomas remain fatal disorders. Although brain tumor biology is a matter of intense research, the cell-of-origin and the complete astrocytoma-inducing signaling pathway remain unknown. To further identify the mechanisms leading to gliomagenesis, we transduced primary astrocytes on a p53−/− background with c-Myc, constitutively active myr-Akt or both, myr-Akt and c-Myc. Transduced astrocytes showed oncogene-specific alterations of morphology, proliferation and differentiation. Following prolonged periods of cultivation, oncogene-transduced astrocytes expressed several stem-cell markers. Furthermore, astrocytes coexpressing c-Myc and Akt were tumorigenic when implanted into the brain of immunocompetent C57BL/6 mice. Our results reveal that the loss of p53 combined with oncogene overexpression in mature astrocytes simulates pivotal features of glioma pathogenesis, providing a good model for assessing the development of secondary glioblastomas.
Collapse
|
44
|
Abstract
Brain tumors are the leading cause of cancer death in children, with ependymoma being the third most common and posing a significant clinical burden. Its mechanism of pathogenesis, reliable prognostic indicators, and effective treatments other than surgical resection have all remained elusive. Until recently, ependymoma research was hindered by the small number of tumors available for study, low resolution of cytogenetic techniques, and lack of cell lines and animal models. Ependymoma heterogeneity, which manifests as variations in tumor location, patient age, histological grade, and clinical behavior, together with the observation of a balanced genomic profile in up to 50% of cases, presents additional challenges in understanding the development and progression of this disease. Despite these difficulties, we have made significant headway in the past decade in identifying the genetic alterations and pathways involved in ependymoma tumorigenesis through collaborative efforts and the application of microarray-based genetic (copy number) and transcriptome profiling platforms. Genetic characterization of ependymoma unraveled distinct mRNA-defined subclasses and led to the identification of radial glial cells as its cell type of origin. This review summarizes our current knowledge in the molecular genetics of ependymoma and proposes future research directions necessary to further advance this field.
Collapse
Affiliation(s)
- Yuan Yao
- Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | |
Collapse
|
45
|
Nagasawa DT, Trang A, Choy W, Spasic M, Yew A, Zarinkhou G, Garcia HM, Yang I. Genetic expression profiles of adult and pediatric ependymomas: molecular pathways, prognostic indicators, and therapeutic targets. Clin Neurol Neurosurg 2013; 115:388-99. [PMID: 23374238 DOI: 10.1016/j.clineuro.2012.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 11/06/2012] [Accepted: 12/02/2012] [Indexed: 12/21/2022]
Abstract
Ependymomas are tumors that can present within either the intracranial or spinal regions. While 90% of all pediatric ependymomas are intracranial, spinal cord ependymomas are more commonly found in patients 20-40 years old. Treatment for spinal lesions has achieved local control rates up to 100% following gross total resection, while pediatric intracranial tumors have 40-60% mortality. Given the inability to effectively treat ependymomas with current standard practices, researchers have focused their efforts on evaluating chromosomal alterations, genetic expression profiles, epigenetic events, and molecular pathways. While these studies have provided critical insight into the potential mechanisms underlying ependymoma pathogenesis, understanding of the intricate interplay between the various pathways involved in tumor initiation, development, and progression will require deeper investigation. However, several potential prognostic markers and therapeutic targets have been identified, providing key areas of focus for future research. The utilization of unique genetic expression profiles based upon patient age, tumor location, tumor grade, and subtype has revealed a multitude of findings warranting further study. Inspection of various molecular pathways associated with ependymomas may establish the foundation for developing novel therapies capable of achieving significant clinical improvements with individualized regimens specifically designed for personalized treatment strategies.
Collapse
Affiliation(s)
- Daniel T Nagasawa
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kaur N, Chettiar S, Rathod S, Rath P, Muzumdar D, Shaikh ML, Shiras A. Wnt3a mediated activation of Wnt/β-catenin signaling promotes tumor progression in glioblastoma. Mol Cell Neurosci 2013; 54:44-57. [PMID: 23337036 DOI: 10.1016/j.mcn.2013.01.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 12/21/2012] [Accepted: 01/11/2013] [Indexed: 12/28/2022] Open
Abstract
Presence of a distinct population of cells that drives tumor progression supports the hierarchical model of tumor development in Glioblastoma (GBM) and substantiates the cancer stem cell hypothesis. Amongst the various developmental signaling pathways that are aberrantly activated, we here show that activated Wnt/β-catenin signaling pathway plays a critical role in malignant transformation and tumor progression in gliomas. We demonstrate that Wnt ligands - Wnt1 and Wnt3a are expressed in a graded manner in these tumors as well as over-expressed in glioma stem cell-lines. A selective inhibition of Wnt signaling pathway by selective knock-down of its ligands Wnt1 and Wnt3a in glioma-derived stem-like cells led to decreased cell proliferation, cell migration and chemo-resistance. Furthermore, Wnt silencing in glioma cells reduced the capacity to form intra-cranial tumors in vivo. Taken together, our study indicates Wnt/β-catenin signaling pathway as an essential driver of glioma tumorigenesis, recognizing role of Wnt3a as an oncogene and thereby offering novel therapeutic strategies for management of these tumors.
Collapse
Affiliation(s)
- Navjot Kaur
- National Centre for Cell Science (NCCS), NCCS Complex, University of Pune Campus, Ganeshkhind, Pune 411007, Maharashtra, India.
| | | | | | | | | | | | | |
Collapse
|
47
|
Oncogenic effects of miR-10b in glioblastoma stem cells. J Neurooncol 2013; 112:153-63. [PMID: 23307328 DOI: 10.1007/s11060-013-1047-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/01/2013] [Indexed: 12/13/2022]
Abstract
MicroRNAs and cancer stem cells have emerged as critical players in glioblastoma, one of the deadliest human cancers. In this study, we investigated the expression and function of microRNA-10b in glioblastoma cells and stem cells. An analysis of The Cancer Genome Atlas data revealed a correlation between high miR-10b levels and poor prognosis in glioblastoma patients. We measured the levels of miR-10b and found that it is upregulated in human glioblastoma tissues, glioblastoma cell and stem cell lines as compared to normal human tissues or astrocytes. Inhibition of miR-10b with a specific antagomir inhibited the proliferation of glioblastoma established and stem cell lines. Inhibition of miR-10b strongly reduced cell invasion and migration in glioblastoma cell and stem cell lines while overexpression of miR-10b induced cell migration and invasion. We also investigated several predicted targets of miR-10b but could not verify any of them experimentally. Additionally, miR-10b inhibition significantly decreased the in vivo growth of stem cell-derived orthotopic GBM xenografts. Altogether, our findings confirm the oncogenic effects of miR-10b in GBM cells and show for the first time a role of this microRNA in GBM stem cells. Targeting miR-10b might therefore inhibit glioblastoma stem cells, which are thought to be at the origin of glioblastoma and to contribute its recurrence and resistance to therapy.
Collapse
|
48
|
Glioblastoma, a Brief Review of History, Molecular Genetics, Animal Models and Novel Therapeutic Strategies. Arch Immunol Ther Exp (Warsz) 2012; 61:25-41. [DOI: 10.1007/s00005-012-0203-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 11/22/2012] [Indexed: 01/06/2023]
|
49
|
Type-3 metabotropic glutamate receptors regulate chemoresistance in glioma stem cells, and their levels are inversely related to survival in patients with malignant gliomas. Cell Death Differ 2012; 20:396-407. [PMID: 23175182 DOI: 10.1038/cdd.2012.150] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Drug treatment of malignant gliomas is limited by the intrinsic resistance of glioma stem cells (GSCs) to chemotherapy. GSCs isolated from human glioblastoma multiforme (GBM) expressed metabotropic glutamate receptors (mGlu3 receptors). The DNA-alkylating agent, temozolomide, killed GSCs only if mGlu3 receptors were knocked down or pharmacologically inhibited. In contrast, mGlu3 receptor blockade did not affect the action of paclitaxel, etoposide, cis-platinum, and irinotecan. mGlu3 receptor blockade enabled temozolomide toxicity by inhibiting a phosphatidylinositol-3-kinase/nuclear factor-κB pathway that supports the expression of O(6)-methylguanine-DNA methyltransferase (MGMT), an enzyme that confers resistance against DNA-alkylating agents. In mice implanted with GSCs into the brain, temozolomide combined with mGlu3 receptor blockade substantially reduced tumor growth. Finally, 87 patients with GBM undergoing surgery followed by adjuvant chemotherapy with temozolomide survived for longer time if tumor cells expressed low levels of mGlu3 receptors. In addition, the methylation state of the MGMT gene promoter in tumor extracts influenced survival only in those patients with low expression of mGlu3 receptors in the tumor. These data encourage the use of mGlu3 receptor antagonists as add-on drugs in the treatment of GBM, and suggest that the transcript of mGlu3 receptors should be measured in tumor specimens for a correct prediction of patients' survival in response to temozolomide treatment.
Collapse
|
50
|
Rosenfeld MR, Pruitt AA. Management of malignant gliomas and primary CNS lymphoma: standard of care and future directions. Continuum (Minneap Minn) 2012; 18:406-15. [PMID: 22810135 DOI: 10.1212/01.con.0000413666.88539.0b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article review the current standard of care of astrocytic gliomas and primary CNS lymphoma and discusses promising new therapeutic targets. RECENT FINDINGS Standard treatment modalities for primary malignant brain tumors include resection, radiation, local or systemic chemotherapy, and, most recently, antiangiogenic agents. However, these tumors often have a rapid course, and patients usually die within a few years of diagnosis. Improved surgical techniques and radiation and chemotherapy can prolong survival while maintaining quality of life, but these therapies remain inadequate. SUMMARY The care of patients with malignant brain tumors is challenging. A better understanding of the pathogenesis of primary malignant brain tumors and the elucidation of aberrant molecular pathways are leading to novel treatment strategies and the ability to identify patients who may benefit from specific treatments.
Collapse
Affiliation(s)
- Myrna R Rosenfeld
- Hospital Clinic/Institute of Biomedical Investigations, University of Barcelona, Spain.
| | | |
Collapse
|