1
|
Nasrolahi A, Azizidoost S, Radoszkiewicz K, Najafi S, Ghaedrahmati F, Anbiyaee O, Khoshnam SE, Farzaneh M, Uddin S. Signaling pathways governing glioma cancer stem cells behavior. Cell Signal 2023; 101:110493. [PMID: 36228964 DOI: 10.1016/j.cellsig.2022.110493] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
Abstract
Glioma is the most common malignant brain tumor that develops in the glial tissue. Several studies have identified that glioma cancer stem cells (GCSCs) play important roles in tumor-initiating features in malignant gliomas. GCSCs are a small population in the brain that presents an essential role in the metastasis of glioma cells to other organs. These cells can self-renew and differentiate, which are thought to be involved in the pathogenesis of glioma. Therefore, targeting GCSCs might be a novel strategy for the treatment of glioma. Accumulating evidence revealed that several signaling pathways, including Notch, TGF-β, Wnt, STAT3, AKT, and EGFR mediated GCSC growth, proliferation, migration, and invasion. Besides, non-coding RNAs (ncRNAs), including miRNAs, circular RNAs, and long ncRNAs have been found to play pivotal roles in the regulation of GCSC pathogenesis and drug resistance. Therefore, targeting these pathways could open a new avenue for glioma management. In this review, we summarized critical signaling pathways involved in the stimulation or prevention of GCSCs tumorigenesis and invasiveness.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Poland
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Nemazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
2
|
Nathan J, Shameera R, Palanivel G. Studying molecular signaling in major angiogenic diseases. Mol Cell Biochem 2022; 477:2433-2450. [PMID: 35581517 DOI: 10.1007/s11010-022-04452-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/24/2022] [Indexed: 10/18/2022]
Abstract
The growth of blood vessels from already existing vasculature is angiogenesis and it is one of the fundamental processes in fetal development, tissue damage or repair, and the reproductive cycle. In a healthy person, angiogenesis is regulated by the balance between pro- and anti-angiogenic factors. However, when the balance is disturbed, it results in various diseases or disorders. The angiogenesis pathway is a sequential cascade and differs based on the stimuli. Therefore, targeting one of the factors involved in the process can help us find a therapeutic strategy to treat irregular angiogenesis. In the past three decades of cancer research, angiogenesis has been at its peak, where an anti-angiogenic agent inhibiting vascular endothelial growth factor acts as a promising substance to treat cancer. In addition, cancer can be assessed based on the expression of angiogenic factors and its response to therapies. Angiogenesis is important for all tissues, which might be normal or pathologically changed and occur through ages. In clinical therapeutics, target therapy focusing on discovery of novel anti-angiogenic agents like bevacizumab, cetuximab, sunitinib, imatinib, lenvatinib, thalidomide, everolimus etc., to block or inhibit the angiogenesis pathway is well explored in recent times. In this review, we will discuss about the molecular signaling pathways involved in major angiogenic diseases in detail.
Collapse
Affiliation(s)
- Jhansi Nathan
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India.
| | - Rabiathul Shameera
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| | - Gajalakshmi Palanivel
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, Tamil Nadu, 600044, India
| |
Collapse
|
3
|
Barati Bagherabad M, Afzaljavan F, ShahidSales S, Hassanian SM, Avan A. Targeted therapies in pancreatic cancer: Promises and failures. J Cell Biochem 2018; 120:2726-2741. [PMID: 28703890 DOI: 10.1002/jcb.26284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incidence rate nearly equal to its mortality rate. The poor prognosis of the disease can be explained by the absence of effective biomarkers for screening and early detection, together with the aggressive behavior and resistance to the currently available chemotherapy. The therapeutic failure can also be attributed to the inter-/intratumor genetic heterogeneity and the abundance of tumor stroma that occupies the majority of the tumor mass. Gemcitabine is used in the treatment of PDAC; however, the response rate is less than 12%. A recent phase III trial revealed that the combination of oxaliplatin, irinotecan, fluorouracil, and leucovorin could be an option for the treatment of metastatic PDAC patients with good performance status, although these approaches can result in high toxicity level. Further investigations are required to develop innovative anticancer agents that either improve gemcitabine activity, within novel combinatorial approaches or acts with a better efficacy than gemcitabine. The aim of the current review is to give an overview of preclinical and clinical studies targeting key dysregulated signaling pathways in PDAC.
Collapse
Affiliation(s)
- Matineh Barati Bagherabad
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine group, Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Liu HW, Su YK, Bamodu OA, Hueng DY, Lee WH, Huang CC, Deng L, Hsiao M, Chien MH, Yeh CT, Lin CM. The Disruption of the β-Catenin/TCF-1/STAT3 Signaling Axis by 4-Acetylantroquinonol B Inhibits the Tumorigenesis and Cancer Stem-Cell-Like Properties of Glioblastoma Cells, In Vitro and In Vivo. Cancers (Basel) 2018; 10:E491. [PMID: 30563094 PMCID: PMC6315804 DOI: 10.3390/cancers10120491] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM), a malignant form of glioma, is characterized by resistance to therapy and poor prognosis. Accumulating evidence shows that the initiation, propagation, and recurrence of GBM is attributable to the presence of GBM stem cells (GBM-CSCs). EXPERIMENTAL APPROACH Herein, we investigated the effect of 4-Acetylantroquinonol B (4-AAQB), a bioactive isolate of Antrodia cinnamomea, on GBM cell viability, oncogenic, and CSCs-like activities. RESULTS We observed that aberrant expression of catenin is characteristic of GBM, compared to other glioma types (p = 0.0001, log-rank test = 475.2), and correlates with poor prognosis of GBM patients. Lower grade glioma and glioblastoma patients (n = 1152) with low catenin expression had 25% and 21.5% better overall survival than those with high catenin expression at the 5 and 10-year time-points, respectively (p = 3.57e-11, log-rank test = 43.8). Immunohistochemistry demonstrated that compared with adjacent non-tumor brain tissue, primary and recurrent GBM exhibited enhanced catenin expression (~10-fold, p < 0.001). Western blot analysis showed that 4-AAQB significantly downregulated β-catenin and dysregulated the catenin/LEF1/Stat3 signaling axis in U87MG and DBTRG-05MG cells, dose-dependently. 4-AAQB⁻induced downregulation of catenin positively correlated with reduced Sox2 and Oct4 nuclear expression in the cells. Furthermore, 4-AAQB markedly reduced the viability of U87MG and DBTRG-05MG cells with 48 h IC50 of 9.2 M and 12.5 M, respectively, effectively inhibited the nuclear catenin, limited the migration and invasion of GBM cells, with concurrent downregulation of catenin, vimentin, and slug; similarly, colony and tumorsphere formation was significantly attenuated with reduced expression of c-Myc and KLF4 proteins. CONCLUSIONS Summarily, we show for the first time that 4-AAQB suppresses the tumor-promoting catenin/LEF1/Stat3 signaling, and inhibited CSCs-induced oncogenic activities in GBM in vitro, with in vivo validation; thus projecting 4-AAQB as a potent therapeutic agent for anti-GBM target therapy.
Collapse
Affiliation(s)
- Heng-Wei Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Kai Su
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
| | - Oluwaseun Adebayo Bamodu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
| | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan, ROC.
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, Taipei 23561, Taiwan.
| | - Chun-Chih Huang
- Department of Applied Chemistry, Chaoyang University of Technology, Taichung 41147, Taiwan.
| | - Li Deng
- Beijing Bioprocess Key Laboratory, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
- Amoy-BUCT Industrial Bio-technovation Institute, Amoy 361022, China.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
| | - Chi-Tai Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
| | - Chien-Min Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan.
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
5
|
Li B, McCrudden CM, Yuen HF, Xi X, Lyu P, Chan KW, Zhang SD, Kwok HF. CD133 in brain tumor: the prognostic factor. Oncotarget 2017; 8:11144-11159. [PMID: 28055976 PMCID: PMC5355253 DOI: 10.18632/oncotarget.14406] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 12/26/2016] [Indexed: 12/31/2022] Open
Abstract
CD133 has been shown to be an important stem cell factor that promotes glioma progression. However, the mechanism for CD133-mediated glioma progression has yet to be fully elucidated. In this study, we found that CD133 mRNA expression was a prognostic marker in three independent glioma patient cohorts, corroborating a putative role for CD133 in glioma progression. Importantly, we found that CD133 expression in glioma was highly correlated with the expression of HOX gene stem cell factors (HOXA5, HOXA7, HOXA10, HOXC4 and HOXC6). The expression of these HOX genes individually was significantly associated with survival. Interestingly, the prognostic significance of CD133 was dependent on the expression level of HOX genes, and vice versa. CD133 (p = 0.021) and HOXA7 (p = 0.001) were independent prognostic markers when the three glioma patient cohorts were combined (n = 231). Our results suggest that HOX genes may play a more important role in progression of glioma when CD133 expression is low. Furthermore, we showed that low-level expression of LIM2 in CD133-high glioma was associated with poorer survival, suggesting that LIM2 could be a therapeutic target for glioma expressing a high level of CD133. Connectivity mapping identified vinblastine and vincristine as agents that could reverse the CD133/HOX genes/LIM2-signature, and we confirmed this by in vitro analysis in glioma cell lines, demonstrating that CD133 and HOX genes were co-expressed and could be downregulated by vincristine. In conclusion, our data show that CD133 and HOX genes are important prognostic markers in glioma and shed light on possible treatment strategies for glioma expressing a high level of CD133.
Collapse
Affiliation(s)
- Bin Li
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Cian M McCrudden
- School of Pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Hiu Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Xinping Xi
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Kwok Wah Chan
- Department of Pathology, University of Hong Kong, Hong Kong
| | - Shu Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, Londonderry, United Kingdom
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| |
Collapse
|
6
|
Yang X, Xiao Z, Du X, Huang L, Du G. Silencing of the long non-coding RNA NEAT1 suppresses glioma stem-like properties through modulation of the miR-107/CDK6 pathway. Oncol Rep 2016; 37:555-562. [DOI: 10.3892/or.2016.5266] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/29/2016] [Indexed: 11/06/2022] Open
|
7
|
Acanda de la Rocha AM, López-Bertoni H, Guruceaga E, González-Huarriz M, Martínez-Vélez N, Xipell E, Fueyo J, Gomez-Manzano C, Alonso MM. Analysis of SOX2-Regulated Transcriptome in Glioma Stem Cells. PLoS One 2016; 11:e0163155. [PMID: 27669421 PMCID: PMC5036841 DOI: 10.1371/journal.pone.0163155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/02/2016] [Indexed: 12/22/2022] Open
Abstract
Introduction Glioblastoma is the most malignant brain tumor in adults and is associated with poor survival despite multimodal treatments. Glioma stem-like cells (GSCs) are cells functionally defined by their self-renewal potential and the ability to reconstitute the original tumor upon orthotopic implantation. They have been postulated to be the culprit of glioma chemo- and radio-resistance ultimately leading to relapse. Understanding the molecular circuits governing the GSC compartment is essential. SOX2, a critical transcription regulator of embryonic and neural stem cell function, is deregulated in GSCs however; the precise molecular pathways regulated by this gene in GSCs remain poorly understood. Results We performed a genome-wide analysis of SOX2-regulated transcripts in GSCs, using a microarray. We identified a total of 2048 differentially expressed coding transcripts and 261 non-coding transcripts. Cell adhesion and cell-cell signaling are among the most enriched terms using Gene Ontology (GO) classification. The pathways altered after SOX2 down-modulation includes multiple cellular processes such as amino-acid metabolism and intercellular signaling cascades. We also defined and classified the set of non-coding transcripts differentially expressed regulated by SOX2 in GSCs, and validated two of them. Conclusions We present a comprehensive analysis of the transcriptome controlled by SOX2 in GSCs, gaining insights in the understanding of the potential roles of SOX2 in glioblastoma.
Collapse
Affiliation(s)
- Arlet M. Acanda de la Rocha
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Hernando López-Bertoni
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Elizabeth Guruceaga
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Bioinformatics Unit, Center for Applied Medical Research, Pamplona, Spain
| | - Marisol González-Huarriz
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Naiara Martínez-Vélez
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Enric Xipell
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Juan Fueyo
- Brain Tumor Center, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Candelaria Gomez-Manzano
- Brain Tumor Center, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Marta M. Alonso
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
- Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
- * E-mail:
| |
Collapse
|
8
|
Su X, Liu X, Ni L, Shi W, Zhu H, Shi J, Chen J, Gu Z, Gao Y, Lan Q, Huang Q. GFAP expression is regulated by Pax3 in brain glioma stem cells. Oncol Rep 2016; 36:1277-84. [PMID: 27432276 DOI: 10.3892/or.2016.4917] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/06/2016] [Indexed: 11/05/2022] Open
Abstract
Glioblastomas are understood to evolve from brain glioma stem cells (BGSCs), and yet the biology underlying this model of tumorigenesis is largely unknown. Paired box 3 protein (Pax3) is a member of the paired box (Pax) family of transcription factors that is normally expressed during embryonic development, but has recently been implicated in tumorigenesis. The present study demonstrated that Pax3 is differentially expressed in U87MG human glioma cell, BGSC and normal 1800 human astrocyte lines. Herein, we identified that the glial fibrillary acidic protein (GFAP), a major intermediate filament protein of mature astrocytes, is directly downregulated during the differentiation of BGSCs via the binding of Pax3 to the promoter region of GFAP. Moreover, siRNA silencing of Pax3 arrested BGSC differentiation, while overexpression of Pax3 promoted the differentiation in BGSCs. Furthermore, we studied the cell proliferation, invasion, apoptosis, differentiation and expression of Pax3 and GFAP in Pax3 siRNA-knockdown and Pax3-overexpressing BGSC models by CCK-8, Transwell migration, flow cytometry and western blot assays. The results indicate that Pax3 regulates GFAP expression, and that Pax3 may contribute to the evolution of BGSCs towards malignancy.
Collapse
Affiliation(s)
- Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaojiang Liu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lanchun Ni
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hui Zhu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jinlong Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jian Chen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhikai Gu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yilu Gao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qing Lan
- Department of Neurosurgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Qingfeng Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
9
|
Song Y, Mu L, Han X, Liu X, Fu S. siRNA targeting stathmin inhibits invasion and enhances chemotherapy sensitivity of stem cells derived from glioma cell lines. Acta Biochim Biophys Sin (Shanghai) 2014; 46:1034-40. [PMID: 25348735 DOI: 10.1093/abbs/gmu099] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glioma is one of the most highly angiogenic tumors, and glioma stem cells (GSCs) are responsible for resistance to chemotherapy and radiotherapy, as well as recurrence after operation. Stathmin is substantial for mitosis and plays an important role in proliferation and migration of glioma-derived endothelial cells. However, the relationship between stathmin and GSCs is incompletely understood. Here we isolated GSCs from glioma cell lines U87MG and U251, and then used siRNA targeting stathmin for silencing. We showed that silencing of stathmin suppressed the proliferation, increased the apoptosis rate, and arrested the cell cycle at G2/M phase in GSCs. Silencing of stathmin in GSCs also resulted in inhibited the migration/invasion as well as the capability of vasculogenic mimicry. The susceptibility of GSCs to temozolomide was also enhanced by stathmin silencing. Our findings suggest stathmin as a potential target in GSCs for glioma treatment.
Collapse
Affiliation(s)
- Yuwen Song
- Department of Neurosurgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Luyan Mu
- Department of Neurosurgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xuezhe Han
- Neurosurgery and Vascular Biology Program, Children's Hospital Boston/Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoqian Liu
- Department of Neurosurgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Songbin Fu
- Department of Genetics, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
10
|
Yusubalieva GM, Baklaushev VP, Gurina OI, Zorkina YA, Gubskii IL, Kobyakov GL, Golanov AV, Goryainov SA, Gorlachev GE, Konovalov AN, Potapov AA, Chekhonin VP. Treatment of Poorly Differentiated Glioma Using a Combination of Monoclonal Antibodies to Extracellular Connexin-43 Fragment, Temozolomide, and Radiotherapy. Bull Exp Biol Med 2014; 157:510-5. [DOI: 10.1007/s10517-014-2603-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Indexed: 12/15/2022]
|
11
|
Paul I, Bhattacharya S, Chatterjee A, Ghosh MK. Current Understanding on EGFR and Wnt/β-Catenin Signaling in Glioma and Their Possible Crosstalk. Genes Cancer 2014; 4:427-46. [PMID: 24386505 DOI: 10.1177/1947601913503341] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/31/2013] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiformes (GBMs) are extensively heterogeneous at both cellular and molecular levels. Current therapeutic strategies include targeting of key signaling molecules using pharmacological inhibitors in combination with genotoxic agents such as temozolomide. In spite of all efforts, the prognosis of glioma patients remains dismal. Therefore, a proper understanding of individual molecular pathways responsible for the progression of GBM is necessary. The epidermal growth factor receptor (EGFR) pathway is probably the most significant signaling pathway clinically implicated in glioma. Not surprisingly, anti-EGFR therapies mostly prevail for therapeutic purposes. The Wnt/β-catenin pathway is well implicated in multiple tumors; however, its role in glioma has only recently started to emerge. We give a concise account of the current understanding of the role of both these pathways in glioma. Last, taking evidences from a limited literature, we outline a number of points where these pathways intersect each other and put forward the possibility of combinatorially targeting them for treatment of glioma.
Collapse
Affiliation(s)
- Indranil Paul
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Seemana Bhattacharya
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Anirban Chatterjee
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mrinal K Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
12
|
Zhou K, Song Y, Zhou W, Zhang C, Shu H, Yang H, Wang B. mGlu3 receptor blockade inhibits proliferation and promotes astrocytic phenotype in glioma stem cells. Cell Biol Int 2014; 38:426-34. [PMID: 24482010 DOI: 10.1002/cbin.10207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 10/10/2013] [Indexed: 12/22/2022]
Abstract
We have characterised, using both in vivo and in vitro methods, the effects of the metabotropic glutamate receptor subtype 3 (mGlu3) antagonist (LY341495) and agonist (LY379268) on the proliferation and differentiation of glioma stem cells (GSC). For in vitro studies, a CCK-8 assay was used to determine the cell proliferation, flow cytometry was performed to determine cell cycle phases, and immunohistochemistry and laser confocal microscopy were employed to detect CD133 expression. For in vivo studies, GSCs were injected into nude mice treated with either LY379268 or LY341495 and the growth of the tumours was measured after 3 weeks. When compared with controls, the proliferation rates and proportion of cells in S phase within the LY341495 treated group decreased in a time-dependent manner. In the presence of differentiation medium containing LY341495, GSC differentiation was diverted into an astrocyte rather than neuronal phenotype. The growth rate and volume of tumours injected into nude mice was reduced in LY341495 treated mice compared with controls. Thus pharmacological blockade of mGlu3 receptor signalling pathway significantly inhibits the growth and proliferation of GSCs both in vitro and in vivo while promoting differentiation to astrocytes. These results further implicate mGlu3 in the biology of glioma and as a target for continued research.
Collapse
Affiliation(s)
- Kun Zhou
- Department of Emergency, Xinqiao Hospital, Chongqing, 400037, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Liu Z, Zhao X, Wang Y, Mao H, Huang Y, Kogiso M, Qi L, Baxter PA, Man TK, Adesina A, Su JM, Picard D, Ching Ho K, Huang A, Perlaky L, Lau CC, Chintagumpala M, Li XN. A patient tumor-derived orthotopic xenograft mouse model replicating the group 3 supratentorial primitive neuroectodermal tumor in children. Neuro Oncol 2014; 16:787-99. [PMID: 24470556 DOI: 10.1093/neuonc/not244] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Supratentorial primitive neuroectodermal tumor (sPNET) is a malignant brain tumor with poor prognosis. New model systems that replicate sPNET's molecular subtype(s) and maintain cancer stem cell (CSC) pool are needed. METHODS A fresh surgical specimen of a pediatric sPNET was directly injected into the right cerebrum of Rag2/SCID mice. The xenograft tumors were serially sub-transplanted in mouse brains, characterized histopathologically, and subclassified into molecular subtype through qRT-PCR and immunohistochemical analysis. CSCs were identified through flow cytometric profiling of putative CSC markers (CD133, CD15, CD24, CD44, and CD117), functional examination of neurosphere forming efficiency in vitro, and tumor formation capacity in vivo. To establish a neurosphere line, neurospheres were propagated in serum-free medium. RESULTS Formation of intracerebral xenograft tumors was confirmed in 4 of the 5 mice injected with the patient tumor. These xenograft tumors were sub-transplanted in vivo 5 times. They replicated the histopathological features of the original patient tumor and expressed the molecular markers (TWIST1 and FOXJ1) of group 3 sPNET. CD133(+) and CD15(+) cells were found to have strong neurosphere-forming efficiency in vitro and potent tumor-forming capacity (with as few as 100 cells) in vivo. A neurosphere line BXD-2664PNET-NS was established that preserved stem cell features and expressed group 3 markers. CONCLUSION We have established a group 3 sPNET xenograft mouse model (IC-2664PNET) with matching neurosphere line (BXD-2664PNET-NS) and identified CD133(+) and CD15(+) cells as the major CSC subpopulations. This novel model system should facilitate biological studies and preclinical drug screenings for childhood sPNET.
Collapse
Affiliation(s)
- Zhigang Liu
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Xiumei Zhao
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Yue Wang
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Hua Mao
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Yulun Huang
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Mari Kogiso
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Lin Qi
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Patricia A Baxter
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Tsz-Kwong Man
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Adekunle Adesina
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Jack M Su
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Daniel Picard
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - King Ching Ho
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Annie Huang
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Laszlo Perlaky
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Ching C Lau
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Murali Chintagumpala
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| | - Xiao-Nan Li
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (Z.L., X.N.L); Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Houston, Texas (Z.L., X.Z., Y.W., H.M., M.K., L.Q., X.N.L.); Texas Children's Cancer Center, Houston, Texas (P.A.B., T.K.M., J.M.S., L.P., C.C.L., M.C.); Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas (A.A.); Division of Hematology-Oncology, Arthur and Sonia Labatt Brain Tumor Research Center, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada (D.P., K.C. H., A.H.)
| |
Collapse
|
14
|
Gu W, Shou J, Gu S, Sun B, Che X. Identifying hedgehog signaling specific microRNAs in glioblastomas. Int J Med Sci 2014; 11:488-93. [PMID: 24688313 PMCID: PMC3970102 DOI: 10.7150/ijms.6764] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 03/11/2014] [Indexed: 12/19/2022] Open
Abstract
Aberrant activation of hedgehog (Hh) signaling pathway plays an important role in the development and proliferation of glioblastoma (GBM) cells. However, its mechanism remains unknown. MicroRNAs (miRNAs) are short non-coding RNA molecules which are involved in the post-transcriptional regulation of genes, and enrolled in signaling transduction network in tumors. This study was designed to investigate the role of miRNAs targeting the Hh signaling pathway in GBMs. According to the expression level of Gli1 mRNA measured by real time PCR, GBM samples were assigned to Gli1 high or low expression group. MiRNA microarray was applied to screen the dysregulated miRNA. As a result, 17 miRNAs were differentially expressed between Gli1 high expression and low expression groups (p < 0.005). Thirteen miRNAs including miR-125b-1 were downregulated, while only 4 miRNAs including miR-144 were upregulated in Gli1 high expression group. In summary, our study presents a subset of miRNAs which target the Hh signaling pathway in GBMs, and throws some light on the aberrant activation mechanism.
Collapse
Affiliation(s)
- Wentao Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jiajun Shou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shixin Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Bin Sun
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaoming Che
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
15
|
Sarkar S, Döring A, Zemp FJ, Silva C, Lun X, Wang X, Kelly J, Hader W, Hamilton M, Mercier P, Dunn JF, Kinniburgh D, van Rooijen N, Robbins S, Forsyth P, Cairncross G, Weiss S, Yong VW. Therapeutic activation of macrophages and microglia to suppress brain tumor-initiating cells. Nat Neurosci 2013; 17:46-55. [PMID: 24316889 DOI: 10.1038/nn.3597] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/06/2013] [Indexed: 12/15/2022]
Abstract
Brain tumor initiating cells (BTICs) contribute to the genesis and recurrence of gliomas. We examined whether the microglia and macrophages that are abundant in gliomas alter BTIC growth. We found that microglia derived from non-glioma human subjects markedly mitigated the sphere-forming capacity of glioma patient-derived BTICs in culture by inducing the expression of genes that control cell cycle arrest and differentiation. This sphere-reducing effect was mimicked by macrophages, but not by neurons or astrocytes. Using a drug screen, we validated amphotericin B (AmpB) as an activator of monocytoid cells and found that AmpB enhanced the microglial reduction of BTIC spheres. In mice harboring intracranial mouse or patient-derived BTICs, daily systemic treatment with non-toxic doses of AmpB substantially prolonged life. Notably, microglia and monocytes cultured from glioma patients were inefficient at reducing the sphere-forming capacity of autologous BTICs, but this was rectified by AmpB. These results provide new insights into the treatment of gliomas.
Collapse
Affiliation(s)
- Susobhan Sarkar
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Axinia Döring
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. [3]
| | - Franz J Zemp
- 1] The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada. [2]
| | - Claudia Silva
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Xueqing Lun
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Xiuling Wang
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - John Kelly
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Walter Hader
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mark Hamilton
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Philippe Mercier
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F Dunn
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dave Kinniburgh
- Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Stephen Robbins
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Peter Forsyth
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Gregory Cairncross
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Wu Q, Chang Y, Zhang L, Zhang Y, Tian T, Feng G, Zhou S, Zheng Q, Han F, Huang F. SRPK1 Dissimilarly Impacts on the Growth, Metastasis, Chemosensitivity and Angiogenesis of Glioma in Normoxic and Hypoxic Conditions. J Cancer 2013; 4:727-35. [PMID: 24312143 PMCID: PMC3842442 DOI: 10.7150/jca.7576] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/19/2013] [Indexed: 12/12/2022] Open
Abstract
Glioma is among the ten most common causes of cancer-related death and has no effective treatment for it, so we are trying to find a new target for anticancer treatment. This study investigates the different expression of SRPK1 as a novel protein in glioma, which can influence tumor cells biological characteristics in normoxic and hypoxic environment. The expression levels of SRPK1 protein in glioma cell lines transfected with siSRPK1 or not were examined using immunofluorescence, RT-PCR and Western blot analysis, respectively. The impact of SRPK1 on the biological characteristics of U251 cells was further studied using methylthiazol tetrazolium assays, flow cytometry, and Transwell invasion chamber assays. The results showed that knockdown of SRPK1 inhibited tumor cells growth, invasion and migration in normoxic condition, but portion of the effect could be reversed in hypoxia. SRPK1 expression was induced in glioma cells by DDP treated, but not TMZ, in both normoxia and hypoxia conditions. We propose SRPK1 as a new molecular player contributing to the early treatment of glioma.
Collapse
Affiliation(s)
- Qianqian Wu
- Institute of Human Anatomy and Histology and Embryology, Otology & Neuroscience Center, Binzhou Medical University, 346 Guanhai Road, Laishan, Shandong Province, 264003, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yan GN, Lv YF, Yang L, Yao XH, Cui YH, Guo DEY. Glioma stem cells enhance endothelial cell migration and proliferation via the Hedgehog pathway. Oncol Lett 2013; 6:1524-1530. [PMID: 24179553 PMCID: PMC3813800 DOI: 10.3892/ol.2013.1569] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 08/27/2013] [Indexed: 12/18/2022] Open
Abstract
The aim of the present study was to determine the possible mechanism underlying the enhanced migration and proliferation of endothelial cells caused by glioma stem cells (GSCs). Tumor spheres enriched in GSCs derived from the mouse GL261 glioma cell line, and the brain microvessel endothelial cell line, b.END3, were used in this study. A Transwell co-culture system, RNAi experiments, quantitative polymerase chain reaction, western blotting and enzyme-linked immunosorbent, cell counting kit-8 (CCK-8) proliferation, Transwell migration and wound-healing assays were used in this study to determine the migration and proliferation ability, as well as the Hedgehog (HH) pathway-related gene expression in the b.END3 cells. Based on the results, it was demonstrated that the migration and proliferation of the endothelial cells were enhanced following co-culture with GSCs. The gene expression of the HH pathway-related genes, Sonic Hedgehog (Shh) and Hedgehog-interacting protein (Hhip) was altered in the endothelial cells when co-cultured with GSCs. Overexpression of glioma-associated oncogene homolog 1 indicated activation of the HH pathway. Following knockdown of smoothened (Smo) in the endothelial cells, the migration and proliferation abilities of the cells were inhibited. GSCs have little effect on enhancing these behaviors in endothelial cells following Smo-knockdown. Further investigation revealed that Shh levels in the supernatant of the co-culture system were elevated, indicating the importance of secreted Shh from the endothelial cells. In conclusion, GSCs enhanced the migration and proliferation of the endothelial cells in vitro, which was likely associated with the activation of the HH pathway in the endothelial cells, caused by the increased secretion of Shh.
Collapse
Affiliation(s)
- Guang-Ning Yan
- Department of Pathology, Southwest Hospital, Third Military Medical University of PLA, Shapingba District, Chongqing 400038, P.R. China
| | | | | | | | | | | |
Collapse
|
18
|
Sze CI, Su WP, Chiang MF, Lu CY, Chen YA, Chang NS. Assessing current therapeutic approaches to decode potential resistance mechanisms in glioblastomas. Front Oncol 2013; 3:59. [PMID: 23516171 PMCID: PMC3601334 DOI: 10.3389/fonc.2013.00059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 03/04/2013] [Indexed: 12/12/2022] Open
Abstract
Unique astrocytic cell infiltrating growth and glial tumor growth in the confined skull make human glioblastoma (GBM) one of the most difficult cancers to treat in modern medicine. Prognosis for patients is very poor, as they die more or less within 12 months. Patients either die of the cancer itself, or secondary complications such as cerebral edema, herniations, or hemorrhages. GBMs rarely metastasize to other organs. However, GBM recurrence associated with resistance to therapeutic drugs is common. Patients die shortly after relapse. GBM is indeed an outstanding cancer model to search for potential mechanisms for drug resistance. Here, we reviewed the current cancer biology of gliomas and their pathophysiological events that contribute to the development of therapeutic resistance. We have addressed the potential roles of cancer stem cells, epigenetic modifications, and epithelial mesenchymal transition (EMT) in the development of resistance to inhibitor drugs in GBMs. The potential role of TIAF1 (TGF-β-induced antiapoptotic factor) overexpression and generation of intratumor amyloid fibrils for conferring drug resistance in GBMs is discussed.
Collapse
Affiliation(s)
- Chun-I Sze
- Department of Anatomy and Cell Biology, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
19
|
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]
|
20
|
Abstract
Glioblastomas are heterogeneous neoplasms that are driven by complex signalling pathways, and are among the most aggressive and challenging cancers to treat. Despite standard treatment with resection, radiation and chemotherapy, the prognosis of patients with glioblastomas remains poor. An increasing understanding of the molecular pathogenesis of glioblastomas has stimulated the development of novel therapies, including the use of molecular-targeted agents. Identification and validation of diagnostic, prognostic and predictive biomarkers has led to the advancement of clinical trial design, and identification of glioblastoma subgroups with a more-favourable prognosis and response to therapy. In this Review, we discuss common molecular alterations relevant to the biology of glioblastomas, targeted, antiangiogenic and immunotherapies that have impacted on the treatment of this disease, and the challenges and pitfalls associated with these therapies. In addition, we emphasize current biomarkers relevant to the management of patients with glioblastoma.
Collapse
|
21
|
Yusubalieva GM, Baklaushev VP, Gurina OI, Gulyaev MV, Pirogov YA, Chekhonin VP. Antitumor Effects of Monoclonal Antibodies to Connexin 43 Extracellular Fragment in Induced Low-Differentiated Glioma. Bull Exp Biol Med 2012; 153:163-9. [DOI: 10.1007/s10517-012-1667-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
22
|
Cho J, Pastorino S, Zeng Q, Xu X, Johnson W, Vandenberg S, Verhaak R, Cherniack A, Watanabe H, Dutt A, Kwon J, Chao YS, Onofrio RC, Chiang D, Yuza Y, Kesari S, Meyerson M. Glioblastoma-derived epidermal growth factor receptor carboxyl-terminal deletion mutants are transforming and are sensitive to EGFR-directed therapies. Cancer Res 2011; 71:7587-96. [PMID: 22001862 PMCID: PMC3242822 DOI: 10.1158/0008-5472.can-11-0821] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Genomic alterations of the epidermal growth factor receptor (EGFR) gene play a crucial role in pathogenesis of glioblastoma multiforme (GBM). By systematic analysis of GBM genomic data, we have identified and characterized a novel exon 27 deletion mutation occurring within the EGFR carboxyl-terminus domain (CTD), in addition to identifying additional examples of previously reported deletion mutations in this region. We show that the GBM-derived EGFR CTD deletion mutants are able to induce cellular transformation in vitro and in vivo in the absence of ligand and receptor autophosphorylation. Treatment with the EGFR-targeted monoclonal antibody, cetuximab, or the small molecule EGFR inhibitor, erlotinib, effectively impaired tumorigenicity of oncogenic EGFR CTD deletion mutants. Cetuximab in particular prolonged the survival of intracranially xenografted mice with oncogenic EGFR CTD deletion mutants, compared with untreated control mice. Therefore, we propose that erlotinib and, especially, cetuximab treatment may be a promising therapeutic strategy in GBM patients harboring EGFR CTD deletion mutants.
Collapse
Affiliation(s)
- Jeonghee Cho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Genomic Analysis Center, Samsung Cancer Reseacrh Institute, Samsung Medical Center, Seoul, 135-710, Republic of Korea
| | - Sandra Pastorino
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | - Qing Zeng
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Xiaoyin Xu
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - William Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | | | - Roel Verhaak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Andrew Cherniack
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Hideo Watanabe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Amit Dutt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Jihyun Kwon
- Genomic Analysis Center, Samsung Cancer Reseacrh Institute, Samsung Medical Center, Seoul, 135-710, Republic of Korea
| | - Ying S. Chao
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | | | - Derek Chiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Yuki Yuza
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| | - Santosh Kesari
- Department of Neurosciences, Moores Cancer Center, UC San Diego, La Jolla, CA, 92093, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of M.I.T. and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
23
|
Kesari S. Understanding Glioblastoma Tumor Biology: The Potential to Improve Current Diagnosis and Treatments. Semin Oncol 2011; 38 Suppl 4:S2-10. [DOI: 10.1053/j.seminoncol.2011.09.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Corle C, Makale M, Kesari S. Cell phones and glioma risk: a review of the evidence. J Neurooncol 2011; 106:1-13. [PMID: 21853424 DOI: 10.1007/s11060-011-0663-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 07/04/2011] [Indexed: 01/12/2023]
Affiliation(s)
- Courtney Corle
- Department of Neurosciences, UC San Diego, Moores UCSD Cancer Center, 3855 Health Sciences Drive, MC 0819, La Jolla, CA 92093-0819, USA
| | | | | |
Collapse
|
25
|
Chekhonin VP, Baklaushev VP, Yusubalieva GM, Belorusova AE, Gulyaev MV, Tsitrin EB, Grinenko NF, Gurina OI, Pirogov YA. Targeted delivery of liposomal nanocontainers to the peritumoral zone of glioma by means of monoclonal antibodies against GFAP and the extracellular loop of Cx43. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:63-70. [PMID: 21703991 DOI: 10.1016/j.nano.2011.05.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 05/13/2011] [Accepted: 05/18/2011] [Indexed: 01/29/2023]
Abstract
UNLABELLED The selectivity of PEGylated immunoliposomes based on monoclonal antibodies against GFAP and the E2 extracellular loop of connexin 43 (MAbE2Cx43) with respect to the focus of a glioma was estimated in experiments on animals with intracranial C6 glioma. Stealth immunoliposomes were labeled with 2 alternative labels, a fluorescent (Dil C18) and a paramagnetic (Gd-DTPA) one. Fluorescent-labeled liposomal nanocontainers were detected at the periphery of the glioma, where the target antigens were overexpressed, 48 hours after injection. Dynamic T1 MRI of rats injected with paramagnetic immunoliposomes carrying MAbE2Cx43 showed distinct accumulation of the paramagnetic contrast agent at the periphery of the glioma, which began 6 hours after administration. These data suggest that immunoliposomal nanocontainers based on antibodies against GFAP and the E2 extracellular fragment of connexin 43 are suitable for targeted delivery of diagnostic and therapeutic drugs to the peritumoral invasion zone of high-grade gliomas. FROM THE CLINICAL EDITOR PEGylated immunoliposomes based on monoclonal antibodies against GFAP and the E2 extracellular loop of connexin 43 were investigated in animals with intracranial C6 glioma. These immunoliposomal nanocontainers were found suitable for targeted delivery of diagnostic and therapeutic drugs to the peritumoral invasion zone of high-grade gliomas.
Collapse
Affiliation(s)
- Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health and Social Development of the Russian Federation, Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Baklaushev VP, Yusubalieva GM, Tsitrin EB, Gurina OI, Grinenko NP, Victorov IV, Chekhonin VP. Visualization of Connexin 43-positive cells of glioma and the periglioma zone by means of intravenously injected monoclonal antibodies. Drug Deliv 2011; 18:331-7. [DOI: 10.3109/10717544.2010.549527] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
27
|
Novel Perspectives on p53 Function in Neural Stem Cells and Brain Tumors. JOURNAL OF ONCOLOGY 2010; 2011:852970. [PMID: 21209724 PMCID: PMC3010739 DOI: 10.1155/2011/852970] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/18/2010] [Accepted: 10/29/2010] [Indexed: 12/30/2022]
Abstract
Malignant glioma is the most common brain tumor in adults and is associated with a very poor prognosis. Mutations in the p53 tumor suppressor gene are frequently detected in gliomas. p53 is well-known for its ability to induce cell cycle arrest, apoptosis, senescence, or differentiation following cellular stress. That the guardian of the genome also controls stem cell self-renewal and suppresses pluripotency adds a novel level of complexity to p53. Exactly how p53 works in order to prevent malignant transformation of cells in the central nervous system remains unclear, and despite being one of the most studied proteins, there is a need to acquire further knowledge about p53 in neural stem cells. Importantly, the characterization of glioma cells with stem-like properties, also known as brain tumor stem cells, has opened up for the development of novel targeted therapies. Here, we give an overview of what is currently known about p53 in brain tumors and neural stem cells. Specifically, we review the literature regarding transformation of adult neural stem cells and, we discuss how the loss of p53 and deregulation of growth factor signaling pathways, such as increased PDGF signaling, lead to brain tumor development. Reactivation of p53 in brain tumor stem cell populations in combination with current treatments for glioma should be further explored and may become a viable future therapeutic approach.
Collapse
|