1
|
Wang Y, Wang B, Cao W, Xu X. PTX3 activates POSTN and promotes the progression of glioblastoma via the MAPK/ERK signalling axis. Biochem Biophys Res Commun 2024; 703:149665. [PMID: 38359612 DOI: 10.1016/j.bbrc.2024.149665] [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: 12/24/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Intrinsic brain tumours such as glioblastoma (GBM) are believed to develop from neuroglial stem or progenitor cells. GBM accounts for approximately half of gliomas. GBM has a poor prognosis and a low 5-year survival rate. Pentraxin 3 (PTX3) is overexpressed in GBM, but the potential mechanism is unclear. METHODS Glioblastoma data from the TCGA and CGGA databases were used to analyse PTX3 expression. Subsequently, in vivo and in vitro experiments were conducted to verify the effect of PTX3 silencing in glioma cells on EMT like process and GSC maintenance. The JASPAR database was used to predict the downstream genes of PTX3. POSTN is a novel target gene of PTX3 in gliomas, and this finding was validated using a luciferase reporter gene assay. Western blotting and KEGG enrichment analysis were used to predict the downstream pathway of POSTN, and it was found that the MAPK/ERK pathway might be related to the function of POSTN. RESULTS GBM tissues have higher levels of PTX3 expression than normal brain tissues (NBTs). In functional tests, PTX3 promoted the EMT like process of GBM cells while maintaining the stem cell characteristics of GBM stem cells and enhancing their self-renewal. Moreover, we performed a dual luciferase reporter experiment to confirm that PTX3 binds to the POSTN promoter region. In addition, the expression of key proteins in the MAPK/ERK signalling pathway was increased after PTX3 overexpression. CONCLUSION POSTN is a direct target of PTX3 that promotes GBM growth via the MAPK/ERK signalling pathway.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
2
|
Ni W, Luo L, Zuo P, Li RP, Xu XB, Wen F, Hu D. lncRNA GHET1 down-regulation suppresses the cell activities of glioma. Cancer Biomark 2018; 23:9-22. [PMID: 30103301 DOI: 10.3233/cbm-171002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wei Ni
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Lin Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Ping Zuo
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Ren-Ping Li
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Xiao-Bing Xu
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Fan Wen
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| | - Dong Hu
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Hospital, Kunming 650118, Yunnan, China
- Department of Neurosurgery, Yunnan Cancer Center, Kunming 650118, Yunnan, China
| |
Collapse
|
3
|
Wang S, Hui Y, Li X, Jia Q. Silencing of lncRNA CCDC26 Restrains the Growth and Migration of Glioma Cells In Vitro and In Vivo via Targeting miR-203. Oncol Res 2017; 26:1143-1154. [PMID: 28600863 PMCID: PMC7844715 DOI: 10.3727/096504017x14965095236521] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Gliomas are the most common primary brain tumors with high mortality. The treatment for gliomas is largely limited due to its uncomprehending pathological mechanism. Here we aimed to investigate the effect of long noncoding RNA (lncRNA) coiled-coil domain-containing 26 (CCDC26) in glioma progression. In our study, the expression of CCDC26 was found upregulated in glioma tissues and cell lines compared with normal tissues and cell lines. Further exploration detected decreased cell proliferation and increased cell apoptosis in U-251 and M059J cells transfected with CCDC26-siRNA. In addition, the silencing of CCDC26 strongly reduced the wound closing rate and the number of invasive cells compared with the scramble group. Simultaneously, the expression of miR-203 was found suppressed in glioma tissues and cells lines. Suppressed level of miR-203 was then elevated in U-251 and M059J cells transfected with CCDC26-siRNA. The result of the luciferase activity assay also showed that the luciferase activity was strongly strengthened by adding the miR-203 inhibitor into the CCDC26 WT group. Moreover, CDCC26-siRNA counteracted the effect of the miR-203 inhibitor in facilitating cell viability and mobility in U-251 cells. The in vivo experiment also revealed that CCDC26-siRNA inhibited glioma growth and metastasis. Taken together, our research indicated a CCDC26/miR-203 pathway in regulating the growth and metastasis of gliomas, providing new viewpoints and promising targets for glioma therapy.
Collapse
Affiliation(s)
- Shilei Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Shandong, P.R. China
| | - Yuzuo Hui
- Department of Neurosurgery, Liaocheng People's Hospital, Shandong, P.R. China
| | - Xiaoming Li
- Department of Pharmacy, Liaocheng People's Hospital, Shandong, P.R. China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People's Hospital, Shandong, P.R. China
| |
Collapse
|
4
|
Barish ME, Herrmann K, Tang Y, Argalian Herculian S, Metz M, Aramburo S, Tirughana R, Gutova M, Annala A, Moats RA, Goldstein L, Rockne RC, Gutierrez J, Brown CE, Ghoda L, Aboody KS. Human Neural Stem Cell Biodistribution and Predicted Tumor Coverage by a Diffusible Therapeutic in a Mouse Glioma Model. Stem Cells Transl Med 2017; 6:1522-1532. [PMID: 28481046 PMCID: PMC5689763 DOI: 10.1002/sctm.16-0397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Engineered neural stem cells (NSCs) intrinsically migrating to brain tumors offer a promising mechanism for local therapeutic delivery. However, difficulties in quantitative assessments of NSC migration and in estimates of tumor coverage by diffusible therapeutics have impeded development and refinement of NSC-based therapies. To address this need, we developed techniques by which conventional serial-sectioned formalin-fixed paraffin-embedded (FFPE) brains can be analyzed in their entirety across multiple test animals. We considered a conventional human glioblastoma model: U251 glioma cells orthotopically engrafted in immunodeficient mice receiving intracerebral (i.c.) or intravenous (i.v.) administrations of NSCs expressing a diffusible enzyme to locally catalyze chemotherapeutic formation. NSC migration to tumor sites was dose-dependent, reaching 50%-60% of total administered NSCs for the i.c route and 1.5% for the i.v. route. Curiously, the most efficient NSC homing was seen with smaller NSC doses, implying existence of rate-limiting process active during administration and/or migration. Predicted tumor exposure to a diffusing therapeutic (assuming a 50 µm radius of action) could reach greater than 50% of the entire tumor volume for i.c. and 25% for i.v. administration. Within individual sections, coverage of tumor area could be as high as 100% for i.c. and 70% for i.v. routes. Greater estimated therapeutic coverage was observed for larger tumors and for larger tumor regions in individual sections. Overall, we have demonstrated a framework within which investigators may rationally evaluate NSC migration to, and integration into, brain tumors, and therefore enhance understanding of mechanisms that both promote and limit this therapeutic modality. Stem Cells Translational Medicine 2017;6:1522-1532.
Collapse
Affiliation(s)
- Michael E Barish
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Kelsey Herrmann
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Yang Tang
- Department of Radiology, University of Southern California, Los Angeles, California, USA
| | - Siranush Argalian Herculian
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Marianne Metz
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Soraya Aramburo
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Revathiswari Tirughana
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Margarita Gutova
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Alexander Annala
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Rex A Moats
- Department of Radiology, University of Southern California, Los Angeles, California, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Leanne Goldstein
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Russell C Rockne
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Jennifer Gutierrez
- Department of Information Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Christine E Brown
- Department of Hematology/HCT, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA.,Department of Immuno-Oncology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Lucy Ghoda
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| | - Karen S Aboody
- Department of Developmental & Stem Cell Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA.,Department of Division of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, California, USA
| |
Collapse
|
5
|
Ma J, Yu J, Liu J, Yang X, Lou M, Liu J, Feng F, Ji P, Wang L. MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma. Oncol Rep 2016; 37:1159-1167. [DOI: 10.3892/or.2016.5320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/01/2016] [Indexed: 11/06/2022] Open
|
6
|
Yue H, Zhu J, Xie S, Li F, Xu Q. MDC1-AS, an antisense long noncoding RNA, regulates cell proliferation of glioma. Biomed Pharmacother 2016; 81:203-209. [PMID: 27261595 DOI: 10.1016/j.biopha.2016.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Growing number of long noncoding RNAs (lncRNAs) are emerging as new modulators in cancer origination and progression. A lncRNA, mediator of DNA damage checkpoint protein 1antisense RNA (MDC1-AS), with unknown function, is the antisense transcript of tumor suppressor MDC1. METHOD In this study, we investigated the expression pattern and functional role of lncRNA MDC1-AS in glioma by using real time PCR and gain-/loss-of-function studies. RESULT The results showed that the expression levels of lncRNA MDC1-AS and MDC1 were significantly downregulated in glioma tissues compared with normal brain tissues, and in glioma cell lines U87MG, U251 and HEB. Overexpression of MDC1-AS resulted in significant inhibition of cell proliferation and cell cycle in U87MG and U251. We also found that MDC1-AS expression was positively correlated with MDC1 expression. In addition, the inhibitory role of MDC1-AS was remarkably diminished when MDC1 was knockdown. CONCLUSION Together, the results suggest that MDC1-AS is a novel tumor suppressor through up-regulation of its antisense tumor-suppressing gene MDC1 in glioma and leads us to propose that MDC1-AS may serve as a potential biomarker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Hongsheng Yue
- Department of Neurosurgery, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan 250013, PR China
| | - Jie Zhu
- Department of Neurosurgery, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan 250013, PR China
| | - Shugang Xie
- Department of Neurosurgery, Shanghe County People's Hospital, Ji'nan 251600, PR China
| | - Fangfang Li
- Nursing Department, Shandong Cancer Hospital and Institute, Ji'nan 250117, PR China
| | - Qun Xu
- Nursing Department, Ji'nan Vocational College of Nursing, Ji'nan 250102, PR China.
| |
Collapse
|
7
|
Hypoxia-cultured human adipose-derived mesenchymal stem cells are non-oncogenic and have enhanced viability, motility, and tropism to brain cancer. Cell Death Dis 2014; 5:e1567. [PMID: 25501828 PMCID: PMC4649837 DOI: 10.1038/cddis.2014.521] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/30/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023]
Abstract
Adult human adipose-derived mesenchymal stem cells (hAMSCs) are multipotent cells, which are abundant, easily collected, and bypass the ethical concerns that plague embryonic stem cells. Their utility and accessibility have led to the rapid development of clinical investigations to explore their autologous and allogeneic cellular-based regenerative potential, tissue preservation capabilities, anti-inflammatory properties, and anticancer properties, among others. hAMSCs are typically cultured under ambient conditions with 21% oxygen. However, physiologically, hAMSCs exist in an environment of much lower oxygen tension. Furthermore, hAMSCs cultured in standard conditions have shown limited proliferative and migratory capabilities, as well as limited viability. This study investigated the effects hypoxic culture conditions have on primary intraoperatively derived hAMSCs. hAMSCs cultured under hypoxia (hAMSCs-H) remained multipotent, capable of differentiation into osteogenic, chondrogenic, and adipogenic lineages. In addition, hAMSCs-H grew faster and exhibited less cell death. Furthermore, hAMSCs-H had greater motility than normoxia-cultured hAMSCs and exhibited greater homing ability to glioblastoma (GBM) derived from brain tumor-initiating cells from our patients in vitro and in vivo. Importantly, hAMSCs-H did not transform into tumor-associated fibroblasts in vitro and were not tumorigenic in vivo. Rather, hAMSCs-H promoted the differentiation of brain cancer cells in vitro and in vivo. These findings suggest an alternative culturing technique that can enhance the function of hAMSCs, which may be necessary for their use in the treatment of various pathologies including stroke, myocardial infarction, amyotrophic lateral sclerosis, and GBM.
Collapse
|
8
|
Azevedo H, Fujita A, Bando SY, Iamashita P, Moreira-Filho CA. Transcriptional network analysis reveals that AT1 and AT2 angiotensin II receptors are both involved in the regulation of genes essential for glioma progression. PLoS One 2014; 9:e110934. [PMID: 25365520 PMCID: PMC4217762 DOI: 10.1371/journal.pone.0110934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/26/2014] [Indexed: 01/25/2023] Open
Abstract
Gliomas are aggressive primary brain tumors with high infiltrative potential. The expression of Angiotensin II (Ang II) receptors has been associated with poor prognosis in human astrocytomas, the most common type of glioma. In this study, we investigated the role of Angiotensin II in glioma malignancy through transcriptional profiling and network analysis of cultured C6 rat glioma cells exposed to Ang II and to inhibitors of its membrane receptor subtypes. C6 cells were treated with Ang II and specific antagonists of AT1 and AT2 receptors. Total RNA was isolated after three and six hours of Ang II treatment and analyzed by oligonucleotide microarray technology. Gene expression data was evaluated through transcriptional network modeling to identify how differentially expressed (DE) genes are connected to each other. Moreover, other genes co-expressing with the DE genes were considered in these analyses in order to support the identification of enriched functions and pathways. A hub-based network analysis showed that the most connected nodes in Ang II-related networks exert functions associated with cell proliferation, migration and invasion, key aspects for glioma progression. The subsequent functional enrichment analysis of these central genes highlighted their participation in signaling pathways that are frequently deregulated in gliomas such as ErbB, MAPK and p53. Noteworthy, either AT1 or AT2 inhibitions were able to down-regulate different sets of hub genes involved in protumoral functions, suggesting that both Ang II receptors could be therapeutic targets for intervention in glioma. Taken together, our results point out multiple actions of Ang II in glioma pathogenesis and reveal the participation of both Ang II receptors in the regulation of genes relevant for glioma progression. This study is the first one to provide systems-level molecular data for better understanding the protumoral effects of Ang II in the proliferative and infiltrative behavior of gliomas.
Collapse
Affiliation(s)
- Hátylas Azevedo
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - André Fujita
- Department of Computer Science, Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvia Yumi Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Priscila Iamashita
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Carlos Alberto Moreira-Filho
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
9
|
Khosh N, Brown CE, Aboody KS, Barish ME. Contact and encirclement of glioma cells in vitro is an intrinsic behavior of a clonal human neural stem cell line. PLoS One 2012; 7:e51859. [PMID: 23240066 PMCID: PMC3519902 DOI: 10.1371/journal.pone.0051859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 11/09/2012] [Indexed: 01/09/2023] Open
Abstract
Pathotropic neural stem and/or progenitor cells (NSCs) can potentially deliver therapeutic agents to otherwise inaccessible cancers. In glioma, NSCs are found in close contact with tumor cells, raising the possibility that specificity of NSC contact with glioma targets originates in the tumor cells themselves. Alternatively, target preferences may originate, at least in part, in the tumor microenvironment. To better understand mechanisms underlying NSC interactions with glioma cells, we examined NSC-target cell contacts in a highly simplified 3-dimensional peptide hydrogel (Puramatrix) in which cell behaviors can be studied in the relative absence of external cues. HB1.F3 is an immortalized clonal human NSC line extensively characterized in preclinical investigations. To study contact formation between HB1.F3 NSCs and glioma cells, we first examined co-cultures of eGFP-expressing HB1.F3 (HB1.F3.eGFP) NSCs and dsRed-expressing U251 glioma (U251.dsRed) cells. Using confocal microscopy, HB1.F3.eGFP cells were observed contacting or encircling U251.dsRed glioma cells, but never the reverse. Next, examining specificity of these contacts, no significant quantitative differences in either percentages of HB1.F3 NSCs contacting targets, or in the extent of target cell encirclement, were observed when HB1.F3.eGFP cells were presented with various potential target cells (human glioma and breast cancer cell lines, patient-derived brain tumor lines, non-tumor fibroblasts, primary mouse and human astroglial cells, and primary adult and newborn human dermal fibroblasts) except that interactions between HB1.F3 cells did not progress beyond establishing contacts. Finally cytoskeletal mechanisms employed by HB1.F3.eGFP cells varied with the substrate. When migrating in Puramatrix, HB1.F3 NSCs exhibited intermittent process extension followed by soma translocation, while during encirclement their movements were more amoeboid. We conclude that formation of contacts and subsequent encirclement of target cells by HB1.F3 NSCs is an intrinsic property of these NSCs, and that preferential contact formation with tumor cells in vivo must therefore be highly dependent on microenvironmental cues.
Collapse
Affiliation(s)
- Nousha Khosh
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Christine E. Brown
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Karen S. Aboody
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
- Division of Neurosurgery, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Michael E. Barish
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
10
|
Wang P, Ren Z, Sun P. Overexpression of the long non-coding RNA MEG3 impairs in vitro glioma cell proliferation. J Cell Biochem 2012; 113:1868-74. [PMID: 22234798 DOI: 10.1002/jcb.24055] [Citation(s) in RCA: 281] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gliomas are the most common type of primary brain tumor in the central nervous system of adults. Maternally Expressed Gene 3 (MEG3) is an imprinted gene located at 14q32 that encodes a non-coding RNA (ncRNA) associated with tumorigenesis. However, little is known about whether and how MEG3 regulates glioma development. In the present study we assayed the expression of MEG3 in glioma tissue samples by real-time polymerase chain reaction assay, and defined the biological functions and target genes by CCK-8 assay, flow cytometry, and RNA immunoprecipitation. We first demonstrated that MEG3 expression was markedly decreased in glioma tissues compared with adjacent normal tissues. Moreover, ectopic expression of MEG3 inhibited cell proliferation and promoted cell apoptosis in U251 and U87 MG human glioma cell lines. We further verified that MEG3 was associated with p53 and that this association was required for p53 activation. These data suggest an important role of MEG3 in the molecular etiology of glioma and implicate the potential application of MEG3 in glioma therapy.
Collapse
Affiliation(s)
- Pengjun Wang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | | | | |
Collapse
|
11
|
Geng J, Luo H, Pu Y, Zhou Z, Wu X, Xu W, Yang Z. Methylation mediated silencing of miR-23b expression and its role in glioma stem cells. Neurosci Lett 2012; 528:185-9. [PMID: 22982144 DOI: 10.1016/j.neulet.2012.08.055] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/13/2012] [Accepted: 08/26/2012] [Indexed: 01/05/2023]
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that negatively regulate gene expression at a post-transcriptional level. Some miRNAs harboring CGIs undergo methylation mediated silencing, a characteristic of many tumor suppressor genes. To identify such miRNAs in glioma stem cells (GSCs), we first showed that miR-23b is frequently methylated in GSCs but not in parallel U87 cells. Meanwhile, miR-23b expression was also markedly reduced in GSCs compared with matching U87 cells. Furthermore, treatment with 5-aza can increase miR-23b expression in GSCs. In addition, ectopic expression of miR-23b in GSCs induces cell cycle arrest and proliferation inhibition. Further analysis showed that miR-23b could enhance the sensitivity of U87 GSCs to TMZ. Our results suggest that miR-23b is epigenetically down-regulated and restoration of miR-23b can effectively suppress cell growth in GSCs.
Collapse
Affiliation(s)
- Jiong Geng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, PR China
| | | | | | | | | | | | | |
Collapse
|
12
|
Ehtesham M, Thompson RC. CXCR4-Expressing Glial Precursor Cells Demonstrate Enhanced Migratory Tropism for Glioma. ACTA ACUST UNITED AC 2012; 3:1086-1091. [PMID: 23293746 DOI: 10.4236/jct.2012.36142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Malignant glioma remains one of the most intractable of human cancers principally due to the highly infiltrative nature of these neoplasms. The use of neural precursor cells (NPC) has received considerable attention based on their ability to selectively migrate towards disseminated areas of tumor in vivo and their described ability to deliver tumor-directed therapies specifically to infiltrating tumor cells. Fundamental to optimizing the use of these cells for potential clinical translation is the development of an understanding regarding the biologic cues that govern their ability to migrate towards infiltrative glioma foci. To this end, in this paper we detail that NPC selected for double-expression of the glial-precursor marker A2B5 and the cell-surface chemokine receptor, CXCR4, demonstrate enhanced in vitro glioma-directed tropism. These findings demonstrate the relevance of these markers for the phenotypic segregation of an optimally tumor-tropic NPC sub-population as a means of enhancing NPC-based therapeutic strategies for the treatment of glioma.
Collapse
Affiliation(s)
- Moneeb Ehtesham
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, USA
| | | |
Collapse
|
13
|
Growth Inhibitory Effect of Palatine Tonsil-derived Mesenchymal Stem Cells on Head and Neck Squamous Cell Carcinoma Cells. Clin Exp Otorhinolaryngol 2012; 5:86-93. [PMID: 22737289 PMCID: PMC3380118 DOI: 10.3342/ceo.2012.5.2.86] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Revised: 12/25/2011] [Accepted: 01/19/2012] [Indexed: 02/06/2023] Open
Abstract
Objectives Mesenchymal stem cells (MSCs) play an important role in the development and growth of tumor cells. However, the effect of human MSCs on the growth of human tumors is not well understood. The purpose of this study is to confirm the growth effect of palatine tonsil-derived MSCs (TD-MSCs) on head and neck squamous cell carcinoma (HNSCC) cell lines and to elucidate the mechanism of their action. Methods TD-MSCs were isolated from patient with chronic tonsillitis and tonsillar hypertrophy. Two human HNSCC cell lines (PNUH-12 and SNU-899) were studied and cocultured with isolated palatine tonsil-derived MSC. The growth inhibitory effect of MSCs on HNSCC cell lines was tested through methylthiazolyldiphenyl-tetrazolium (MTT) assay. The apoptosis induction effect of MSCs on cell lines was assessed with flow cytometry and reverse transcriptase (RT)-PCR. Results Palatine tonsil-derived MSCs exhibited a growth inhibitory effect on both cell lines. Cell cycle analysis showed an accumulation of tumor cells predominantly in G0/G1 phase with an increase in concentration of TD-MSCs, which was confirmed by increased mRNA expression of cell cycle negative regulator p21. Apoptosis of tumor cells increased significantly as concentration of cocultured TD-MSCs increased. Additionally, mRNA expression of caspase 3 was upregulated with increased concentration of TD-MSCs. Conclusion TD-MSCs have a potential growth inhibitory effect on HNSCC cell lines in vitro by inducing apoptotic cell death and G1 phase arrest of cell lines.
Collapse
|
14
|
Ahmed AU, Tyler MA, Thaci B, Alexiades NG, Han Y, Ulasov IV, Lesniak MS. A comparative study of neural and mesenchymal stem cell-based carriers for oncolytic adenovirus in a model of malignant glioma. Mol Pharm 2011; 8:1559-72. [PMID: 21718006 DOI: 10.1021/mp200161f] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme is a primary malignancy of the central nervous system that is universally fatal due to its disseminated nature. Recent investigations have focused on the unique tumor-tropic properties of stem cells as a novel platform for targeted delivery of anticancer agents to the brain. Neural stem cells (NSCs) and mesenchymal stem cells (MSCs) both have the potential to function as cell carriers for targeted delivery of a glioma restricted oncolytic virus to disseminated tumor due to their reported tumor tropism. In this study, we evaluated NSCs and MSCs as cellular delivery vehicles for an oncolytic adenovirus in the context of human glioma. We report the first preclinical comparison of the two cell lines and show that, while both stem cell lines are able to support therapeutic adenoviral replication intracellularly, the amount of virus released from NSCs was a log higher than the MSC (p < 0.001). Moreover, only virus loaded NSCs that were administered intracranially in an orthotopic glioma model significantly prolonged the survival of tumor bearing animals (median survival for NSCs 68.5 days vs 44 days for MSCs, p < 0.002). Loading oncolytic adenovirus into NSCs and MSCs also led to expression of both pro- and anti-inflammatory genes and decreased vector-mediated neuroinflammation. Our results indicate that, despite possessing a comparable migratory capacity, NSCs display superior therapeutic efficacy in the context of intracranial tumors. Taken together, these findings argue in favor of NSCs as an effective cell carrier for antiglioma oncolytic virotherapy.
Collapse
Affiliation(s)
- Atique U Ahmed
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Chien LY, Hsiao JK, Hsu SC, Yao M, Lu CW, Liu HM, Chen YC, Yang CS, Huang DM. In vivo magnetic resonance imaging of cell tropism, trafficking mechanism, and therapeutic impact of human mesenchymal stem cells in a murine glioma model. Biomaterials 2011; 32:3275-84. [PMID: 21295344 DOI: 10.1016/j.biomaterials.2011.01.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 01/13/2011] [Indexed: 02/07/2023]
Abstract
Stem cells have offered much promise as delivery vehicles for brain tumor therapy, with the development of modalities to track the tumor tropism of stem cells receiving intense focus. Cellular magnetic resonance imaging (MRI) allows serial high-resolution in vivo detection of transplanted stem cells' tropism toward gliomas in the mouse brain once these cells are internally labeled with iron oxide particles, but has been impeded by low labeling efficiencies. In this study, we describe the use of ferucarbotran and protamine (Fer-Pro) complexes for labeling human mesenchymal stem cells (hMSCs) for MRI tracking of glioma tropism in vivo. We found that Fer-Pro was not toxic and was highly efficient for labeling in vitro. Cell labeling with Fer-Pro promoted the migration of hMSCs toward glioma U87MG cells in vitro, which was mediated by stromal-derived factor-1/CXCR4 (SDF-1/CXCR4) signaling. Fer-Pro-labeled hMSCs could migrate specifically toward gliomas in vivo, which was observed with a clinical 1.5-T MRI system. The efficient labeling of Fer-Pro also allowed a tropic mechanism mediated by SDF-1/CXCR4 signaling to be detected by MRI in vivo. Additionally, the potential intrinsic inhibitory effect of hMSCs on glioma progression was estimated simultaneously. This is the first report to have used a clinical MRI modality to simultaneously study the migration, the therapeutic impact on tumors, and above all the trafficking mechanism of bone marrow-derived mesenchymal stem cells from human in a murine glioma xenograft model. The use of Fer-Pro for stem cell labeling may have potential clinical applications in stem cell guided therapy.
Collapse
Affiliation(s)
- Li-Ying Chien
- Center for Nanomedicine Research, National Health Research Institutes, Miaoli 350, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Malakootian M, Mowla SJ, Saberi H, Asadi MH, Atlasi Y, Shafaroudi AM. Differential expression of nucleostemin, a stem cell marker, and its variants in different types of brain tumors. Mol Carcinog 2010; 49:818-25. [PMID: 20572164 DOI: 10.1002/mc.20658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Nucleostemin (NS) is implicated in the control of stem and cancer cell proliferation. In the present study, we have examined the expression of NS and its spliced variants in various brain tumors. Total RNA was extracted from 59 brain tumor samples, and the expression of different NS spliced variants was measured by semi-quantitative RT-PCR. The subcellular distribution of NS protein in brain tumors was further examined by immunohistochemistry. Furthermore, to decipher the potential involvement of NS in brain tumorogenesis, its expression was knocked-down by means of RNA interference (RNAi) in two malignant glioma (U-87MG and A172), one astrocytoma (1321N1) and one medulloblastoma (DAOY) cell lines. The alterations in cell-cycle progression of the treated cells were then analyzed by flow cytometry. Our data revealed that NS and its variants are widely expressed in different types of brain tumors. Among the NS spliced variants, variant "1" and variant "3" were detected in the majority of tumor samples, whereas variant "2" was only detectable in few samples. Moreover, the intensity of the expression was correlated with the grade of the tumors (P < 0.05). Accordingly, the expression was much higher in glial tumors compared to that of meningiomas. As expected, a nucleolar/nucleoplasmic localization of NS protein was observed in the examined tumor samples. RNAi results revealed a significant reduction of NS expression along with a moderate blockade of the cell cycle in G(2)/M and S phases of NS-siRNA treated cells. All in all, our data suggest a potential role for NS in tumorogenesis of brain cancers.
Collapse
|
17
|
Achanta P, Sedora Roman NI, Quiñones-Hinojosa A. Gliomagenesis and the use of neural stem cells in brain tumor treatment. Anticancer Agents Med Chem 2010; 10:121-30. [PMID: 20184546 DOI: 10.2174/187152010790909290] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 12/29/2009] [Indexed: 01/08/2023]
Abstract
The role of neural stem cells (NSCs) in both the physiological and pathological processes in the brain has been refined through recent studies within the neuro-oncological field. Alterations in NSC regulatory mechanisms may be fundamental for the development and progression of malignant gliomas. A subpopulation of cells within the tumor known as brain tumor stem cells (BTSCs) have been shown to share key properties with NSCs. The BTSC hypothesis has significantly contributed to a potential understanding as to why brain tumors hold such dismal prognosis. On the other hand, the normal NSCs possess the capacity to migrate extensively towards the tumor bulk as well as to lingering neoplastic regions of the brain. The tropism of NSCs towards brain tumors may provide an additional tool for the treatment of brain cancer. The creation of potential therapies through the use of NSCs has been studied and includes the delivery of gene products to specific locations of the central nervous system selectively targeting malignant brain tumor cells and maximizing the efficiency of their delivery. Here, the proposed mechanisms of how brain tumors emerge, the molecular pathways interrupted in NSC pathogenesis and the most recent preclinical results in the use of NSCs for glioma treatment are reviewed.
Collapse
Affiliation(s)
- Pragathi Achanta
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD 21231, USA
| | | | | |
Collapse
|
18
|
Kranzler J, Tyler MA, Sonabend AM, Ulasov IV, Lesniak MS. Stem cells as delivery vehicles for oncolytic adenoviral virotherapy. Curr Gene Ther 2010; 9:389-95. [PMID: 19860653 DOI: 10.2174/156652309789753347] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme is the most common primary intracranial tumor in humans. Despite continued advances in cancer therapy, the outcome for patients diagnosed with this disease remains bleak. Novel treatments involving the use of conditionally replicating adenoviruses (CRAds) to target malignant brain tumors have undergone extensive research and proven to be a promising mode of glioblastoma therapy. CRAds are genetically manipulated to replicate within tumor cells, exhibiting a high degree of infectivity, cytotoxicity, and transgene expression. While the use of various CRAds has been deemed safe for intracranial injection in preclinical trials, a significant therapeutic effect has yet to be seen in patients. This shortcoming stems from the distribution limitations involved with local delivery of virolytic agents. To enhance this modality of treatment, stem cells have been explored as cellular vehicles in virotherapy applications, given that they possess an intrinsic tropism for malignant brain tumors. Stem cell loaded CRAd delivery offers a more specific and effective method of targeting disseminated tumor cells and forms the basis for this review.
Collapse
Affiliation(s)
- Justin Kranzler
- The Brain Tumor Center, The University of Chicago, Pritzker School of Medicine, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
19
|
Gondi CS, Veeravalli KK, Gorantla B, Dinh DH, Fassett D, Klopfenstein JD, Gujrati M, Rao JS. Human umbilical cord blood stem cells show PDGF-D-dependent glioma cell tropism in vitro and in vivo. Neuro Oncol 2010; 12:453-65. [PMID: 20406896 DOI: 10.1093/neuonc/nop049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite advances in clinical therapies and technologies, the prognosis for patients with malignant glioma is poor. Neural stem cells (NSCs) have a chemotactic tropism toward glioma cells. The use of NSCs as carriers of therapeutic agents for gliomas is currently being explored. Here, we demonstrate that cells isolated from the umbilical cord blood show mesenchymal characteristics and can differentiate to adipocytes, osteocytes, and neural cells and show tropism toward cancer cells. We also show that these stem cells derived from the human umbilical cord blood (hUCB) induce apoptosis-like cell death in the glioma cell line SNB19 via Fas-mediated caspase-8 activation. From our glioma tropism studies, we have observed that hUCB cells show tropism toward glioma cells in vitro, in vivo, and ex vivo. We determined that this migration is partially dependent on the expression levels of platelet-derived growth factor (PDGF)-D from glioma cells and have observed that local concentration gradient of PDGF-D is sufficient to cause migration of hUCB cells toward the gradient as seen from our brain slice cultures. In our animal experiment studies, we observed that intracranially implanted SNB19 green fluorescent protein cells induced tropism of the hUCB cells toward themselves. In addition, the ability of these hUCBs to inhibit established intracranial tumors was also observed. We also determined that the migration of stem cells toward glioma cells was partially dependent on PDGF secreted by glioma cells and that the presence of PDGF-receptor (PDGFR) on hUCB is required for migration. Our results demonstrate that hUCB are capable of inducing apoptosis in human glioma cells and also show that glioma tropism and hUCB tropism toward glioma cells are partially dependent on the PDGF/PGGFR system.
Collapse
Affiliation(s)
- Christopher S Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61605, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Thu MS, Najbauer J, Kendall SE, Harutyunyan I, Sangalang N, Gutova M, Metz MZ, Garcia E, Frank RT, Kim SU, Moats RA, Aboody KS. Iron labeling and pre-clinical MRI visualization of therapeutic human neural stem cells in a murine glioma model. PLoS One 2009; 4:e7218. [PMID: 19787043 PMCID: PMC2746284 DOI: 10.1371/journal.pone.0007218] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Accepted: 08/05/2009] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment strategies for the highly invasive brain tumor, glioblastoma multiforme, require that cells which have invaded into the surrounding brain be specifically targeted. The inherent tumor-tropism of neural stem cells (NSCs) to primary and invasive tumor foci can be exploited to deliver therapeutics to invasive brain tumor cells in humans. Use of the strategy of converting prodrug to drug via therapeutic transgenes delivered by immortalized therapeutic NSC lines have shown efficacy in animal models. Thus therapeutic NSCs are being proposed for use in human brain tumor clinical trials. In the context of NSC-based therapies, MRI can be used both to non-invasively follow dynamic spatio-temporal patterns of the NSC tumor targeting allowing for the optimization of treatment strategies and to assess efficacy of the therapy. Iron-labeling of cells allows their presence to be visualized and tracked by MRI. Thus we aimed to iron-label therapeutic NSCs without affecting their cellular physiology using a method likely to gain United States Federal Drug Administration (FDA) approval. METHODOLOGY For human use, the characteristics of therapeutic Neural Stem Cells must be clearly defined with any pertubation to the cell including iron labeling requiring reanalysis of cellular physiology. Here, we studied the effect of iron-loading of the therapeutic NSCs, with ferumoxide-protamine sulfate complex (FE-Pro) on viability, proliferation, migratory properties and transgene expression, when compared to non-labeled cells. FE-Pro labeled NSCs were imaged by MRI at tumor sites, after intracranial administration into the hemisphere contralateral to the tumor, in an orthotopic human glioma xenograft mouse model. CONCLUSION FE-Pro labeled NSCs retain their proliferative status, tumor tropism, and maintain stem cell character, while allowing in vivo cellular MRI tracking at 7 Tesla, to monitor their real-time migration and distribution at brain tumor sites. Of significance, this work directly supports the use of FE-Pro-labeled NSCs for real-time tracking in the clinical trial under development: "A Pilot Feasibility Study of Oral 5-Fluorocytosine and Genetically modified Neural Stem Cells Expressing Escherichia coli Cytosine Deaminase for Treatment of Recurrent High-Grade Gliomas".
Collapse
Affiliation(s)
- Mya S. Thu
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (MYT); (KSA)
| | - Joseph Najbauer
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Stephen E. Kendall
- Division of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Ira Harutyunyan
- Radiology MS 81, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nicole Sangalang
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Margarita Gutova
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Marianne Z. Metz
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Elizabeth Garcia
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Richard T. Frank
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Regenerative Medicine, Gachon University Gil Hospital, Inchon, Korea
| | - Rex A. Moats
- Radiology MS 81, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Karen S. Aboody
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- Division of Neuroscience, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (MYT); (KSA)
| |
Collapse
|
21
|
Lee DH, Ahn Y, Kim SU, Wang KC, Cho BK, Phi JH, Park IH, Black PM, Carroll RS, Lee J, Kim SK. Targeting rat brainstem glioma using human neural stem cells and human mesenchymal stem cells. Clin Cancer Res 2009; 15:4925-34. [PMID: 19638465 DOI: 10.1158/1078-0432.ccr-08-3076] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE Brainstem gliomas are usually inoperable and have a dismal prognosis. Based on the robust tropisms of neural stem cells (NSC) and mesenchymal stem cells (MSC) to brain tumors, we compared the tumor-tropic migratory capacities of these stem cells and evaluated the therapeutic potential of genetically engineered human NSCs encoding cytosine deaminase (CD) and IFNbeta against brainstem gliomas. EXPERIMENTAL DESIGN The directed migratory capacities of NSCs and MSCs to brainstem glioma (F98) were evaluated both in vitro and in vivo. The human NSCs (HB1.F3) and various human MSCs, such as bone marrow-derived MSCs (HM3.B10), adipose tissue-derived MSCs, and umbilical cord blood-derived MSCs, were tested. Human fibroblast cells (HFF-1) were used as the negative control. As a proof of concept, the bioactivity of HB1.F3-CD-IFNbeta was analyzed with a cell viability assay, and animals with brainstem gliomas were injected with HB1.F3-CD-IFNbeta cells followed by systemic 5-fluorocytosine treatment. RESULTS In an in vitro modified Transwell migration assay and in vivo stem cell injection into established brainstem gliomas in rats, all the stem cells showed a significant migratory capacity compared with that of the control (P < 0.01). Histologic analysis showed a 59% reduction in tumor volume in the HB1.F3-CD-IFNbeta-treated group (P < 0.05). Apoptotic cells were increased 2.33-fold in animals treated with HB1.F3-CD-IFNbeta compared with the respective control groups (P < 0.01). CONCLUSION The brainstem glioma-tropic migratory capacities of MSCs from various sources were similar to those of NSCs. Genetically engineered NSCs show therapeutic efficacy against brainstem gliomas.
Collapse
Affiliation(s)
- Do-Hun Lee
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Combination of adenoviral virotherapy and temozolomide chemotherapy eradicates malignant glioma through autophagic and apoptotic cell death in vivo. Br J Cancer 2009; 100:1154-64. [PMID: 19277041 PMCID: PMC2664399 DOI: 10.1038/sj.bjc.6604969] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Conditionally replicative adenoviruses (CRAds) represent a novel treatment strategy for malignant glioma. Recent studies suggest that the cytopathic effect elicited by these vectors is mediated through autophagy, a form of programmed cell death. Likewise, temozolomide (TMZ), a chemotherapeutic agent used for the treatment of malignant gliomas, also triggers autophagic cell death. In this study, we examined the potential to combine the two treatments in the setting of experimental glioma. In vitro, pretreatment with TMZ followed by CRAd-Surivin-pk7 enhanced cytotoxicity against a panel of glioma cell lines. Western blot analysis showed increased expression of BAX and p53, decreased expression of BCL2 and elevated level of APG5. Treatment with TMZ followed by CRAd-Survivin-pk7 (CRAd-S-pk7) led to a significant over-expression of autophagy markers, acidic vesicular organelles and light-chain 3 (LC3). These results were further evaluated in vivo, in which 90% of the mice with intracranial tumours were long-term survivors (>100 days) after treatment with TMZ and CRAd-S-pk7 (P<0.01). Analysis of tumours ex vivo showed expression of both LC3 and cleaved Caspase-3, proving that both autophagy and apoptosis are responsible for cell death in vivo. These results suggest that combination of chemovirotherapy offers a powerful tool against malignant glioma and should be further explored in the clinical setting.
Collapse
|
23
|
Neural stem cells target intracranial glioma to deliver an oncolytic adenovirus in vivo. Gene Ther 2008; 16:262-78. [PMID: 19078993 DOI: 10.1038/gt.2008.165] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adenoviral oncolytic virotherapy represents an attractive treatment modality for central nervous system (CNS) neoplasms. However, successful application of virotherapy in clinical trials has been hampered by inadequate distribution of oncolytic vectors. Neural stem cells (NSCs) have been shown as suitable vehicles for gene delivery because they track tumor foci. In this study, we evaluated the capability of NSCs to deliver a conditionally replicating adenovirus (CRAd) to glioma. We examined NSC specificity with respect to viral transduction, migration and capacity to deliver a CRAd to tumor cells. Fluorescence-activated cell sorter (FACS) analysis of NSC shows that these cells express a variety of surface receptors that make them amenable to entry by recombinant adenoviruses. Luciferase assays with replication-deficient vectors possessing a variety of transductional modifications targeted to these receptors confirm these results. Real-time PCR analysis of the replication profiles of different CRAds in NSCs and a representative glioma cell line, U87MG, identified the CRAd-Survivin (S)-pk7 virus as optimal vector for further delivery studies. Using in vitro and in vivo migration studies, we show that NSCs infected with CRAd-S-pk7 virus migrate and preferentially deliver CRAd to U87MG glioma. These results suggest that NSCs mediate an enhanced intratumoral distribution of an oncolytic vector in malignant glioma when compared with virus injection alone.
Collapse
|
24
|
Colleoni F, Torrente Y. The new challenge of stem cell: brain tumour therapy. Cancer Lett 2008; 272:1-11. [PMID: 18621474 DOI: 10.1016/j.canlet.2008.05.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 01/07/2008] [Accepted: 05/08/2008] [Indexed: 11/15/2022]
Abstract
The surprising similarity of much brain tumour behavior to the intrinsic properties of the neural stem/progenitor cell has triggered a recent interest in both arming stem cells to track and help eradicate tumours and in viewing stem cell biology as somehow integral to the emergence and/or production of the neoplasm itself. Moreover, based on the unique capacity of neural stem cells (NSCs) to migrate throughout the brain and to target invading tumour cells, the transplantation of NSCs offers a new potential therapeutic approach as a cell-based delivery system for gene therapy in brain tumours. On the one hand, both stem cells and cancer cells are thought to be capable of unlimited proliferation. While on the other, many tumours and cancer cell lines express stem cell markers, suggesting either that cancer cells resemble stem cells or that cancers contain stem-like cells. In this review we highlight the close relationship between normal neural stem cells and brain tumour stem cells and also suggest the possible clinical implications that these similarities could offer.
Collapse
Affiliation(s)
- F Colleoni
- Stem Cell Laboratory, Department of Neurological Science, University of Milan, Fondazione IRCCS Ospedale Maggiore Policlinico, Centro Dino Ferrari, Italy
| | | |
Collapse
|
25
|
A safety and efficacy study of local delivery of interleukin-12 transgene by PPC polymer in a model of experimental glioma. Anticancer Drugs 2008; 19:133-42. [PMID: 18176109 DOI: 10.1097/cad.0b013e3282f24017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Interleukin-12 (IL-12) triggers an antitumoral immune response and an antiangiogenic effect against cancer. In this study, we tested a novel polymeric vehicle for IL-12 gene therapy along with adjuvant local biodegradable carmustine (BCNU) chemotherapy for the treatment of malignant glioma. Highly concentrated DNA/PPC (polyethylenimine covalently modified with methoxypolyethyleneglycol and cholesterol) complexes were used to deliver a murine plasmid encoding IL-12 (pmIL-12). For toxicity assessment, mice received intracranial injections with different volumes of pmIL-12/PPC. For efficacy, mice with intracranial GL261 glioma were treated with local delivery of pmIL-12/PPC and/or BCNU-containing polymers. Intracranial injections of 5-10 microl of pmIL-12/PPC were well tolerated and led to IL-12 expression in the brains of treated animals. Treatment with pmIL-12/PPC led to a significant increase in survival compared with untreated mice (median survival 57 days; 25% long-term survival >95 vs. 45 days for control; P<0.05). Treatment with BCNU led to a significant increase in survival compared with untreated mice, with 75% of treated mice having a long-term survival >95 days, (P<0.05). Most importantly, the combination of BCNU and pmIL-12/PPC led to a survival of 100% of the mice for 95 days after treatment (P<0.0001). This novel strategy is safe and effective for the treatment of malignant glioma. The synergy resultant from the combination of locally administered pmIL-12/PPC and BCNU suggests a role for this approach in the treatment of malignant brain tumors.
Collapse
|
26
|
Aboody KS, Najbauer J, Danks MK. Stem and progenitor cell-mediated tumor selective gene therapy. Gene Ther 2008; 15:739-52. [PMID: 18369324 DOI: 10.1038/gt.2008.41] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The poor prognosis for patients with aggressive or metastatic tumors and the toxic side effects of currently available treatments necessitate the development of more effective tumor-selective therapies. Stem/progenitor cells display inherent tumor-tropic properties that can be exploited for targeted delivery of anticancer genes to invasive and metastatic tumors. Therapeutic genes that have been inserted into stem cells and delivered to tumors with high selectivity include prodrug-activating enzymes (cytosine deaminase, carboxylesterase, thymidine kinase), interleukins (IL-2, IL-4, IL-12, IL-23), interferon-beta, apoptosis-promoting genes (tumor necrosis factor-related apoptosis-inducing ligand) and metalloproteinases (PEX). We and others have demonstrated that neural and mesenchymal stem cells can deliver therapeutic genes to elicit a significant antitumor response in animal models of intracranial glioma, medulloblastoma, melanoma brain metastasis, disseminated neuroblastoma and breast cancer lung metastasis. Most studies reported reduction in tumor volume (up to 90%) and increased survival of tumor-bearing animals. Complete cures have also been achieved (90% disease-free survival for >1 year of mice bearing disseminated neuroblastoma tumors). As we learn more about the biology of stem cells and the molecular mechanisms that mediate their tumor-tropism and we identify efficacious gene products for specific tumor types, the clinical utility of cell-based delivery strategies becomes increasingly evident.
Collapse
Affiliation(s)
- K S Aboody
- Division of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| | | | | |
Collapse
|
27
|
Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, Dimeco F, Olivi A, Eberhart CG. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 2007; 25:2524-33. [PMID: 17628016 PMCID: PMC2610257 DOI: 10.1634/stemcells.2007-0166] [Citation(s) in RCA: 446] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brain tumors can arise following deregulation of signaling pathways normally activated during brain development and may derive from neural stem cells. Given the requirement for Hedgehog in non-neoplastic stem cells, we investigated whether Hedgehog blockade could target the stem-like population in glioblastoma multiforme (GBM). We found that Gli1, a key Hedgehog pathway target, was highly expressed in 5 of 19 primary GBM and in 4 of 7 GBM cell lines. Shh ligand was expressed in some primary tumors, and in GBM-derived neurospheres, suggesting a potential mechanism for pathway activation. Hedgehog pathway blockade by cyclopamine caused a 40%-60% reduction in growth of adherent glioma lines highly expressing Gli1 but not in those lacking evidence of pathway activity. When GBM-derived neurospheres were treated with cyclopamine and then dissociated and seeded in media lacking the inhibitor, no new neurospheres formed, suggesting that the clonogenic cancer stem cells had been depleted. Consistent with this hypothesis, the stem-like fraction in gliomas marked by both aldehyde dehydrogenase activity and Hoechst dye excretion (side population) was significantly reduced or eliminated by cyclopamine. In contrast, we found that radiation treatment of our GBM neurospheres increased the percentage of these stem-like cells, suggesting that this standard therapy preferentially targets better-differentiated neoplastic cells. Most importantly, viable GBM cells injected intracranially following Hedgehog blockade were no longer able to form tumors in athymic mice, indicating that a cancer stem cell population critical for ongoing growth had been removed. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Eli E. Bar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aneeka Chaudhry
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alex Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xing Fan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karisa Schreck
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sara Piccirillo
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | - Angelo L. Vescovi
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | | | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G. Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Ozolek JA, Jane EP, Krowsoski L, Sammak PJ. Human Embryonic Stem Cells (HSF-6) Show Greater Proliferation and Apoptoses When Grown on Glioblastoma Cells Than Mouse Embryonic Fibroblasts at Day 19 in Culture: Comparison of Proliferation, Survival, and Neural Differentiation on Two Different Feeder Cell Types. Stem Cells Dev 2007; 16:403-12. [PMID: 17610370 DOI: 10.1089/scd.2006.0109] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recent years have seen a surge of scientific research examining the interdependence of one germ layer in the development of the other, both in vivo and in vitro. For example, the endoderm is believed to play a crucial role in the formation of mesoderm and subsequent maturation of cells belonging to the mesodermal lineage. Our understanding of this complex relationship is continuously growing with reinterpretation of earlier concepts and apprehension of newer hypotheses into the biology of embryonic development. Here we discuss some of the events governing the cooperative control of endoderm over mesoderm, and propose a perspective based on the existing literature and our own experience.
Collapse
Affiliation(s)
- John A Ozolek
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | |
Collapse
|
29
|
Lin D, Najbauer J, Salvaterra PM, Mamelak AN, Barish ME, Garcia E, Metz MZ, Kendall SE, Bowers M, Kateb B, Kim SU, Johnson M, Aboody KS. Novel method for visualizing and modeling the spatial distribution of neural stem cells within intracranial glioma. Neuroimage 2007; 37 Suppl 1:S18-26. [PMID: 17560798 DOI: 10.1016/j.neuroimage.2007.03.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2007] [Revised: 03/20/2007] [Accepted: 03/26/2007] [Indexed: 12/18/2022] Open
Abstract
Neural stem cells (NSCs) hold great promise for glioma therapy due to their inherent tumor-tropic properties, enabling them to deliver therapeutic agents directly to invasive tumor sites. In the present study, we visualized and quantitatively analyzed the spatial distribution of tumor-tropic NSCs in a mouse model of orthotopic glioma in order to predict the therapeutic efficacy of a representative NSC-based glioma therapy. U251.eGFP human glioma was established in the brain of athymic mice, followed by stereotactic injection of CM-DiI-labeled human NSCs posterior-lateral to the tumor site. Confocal microscopy, three-dimensional modeling and mathematical algorithms were used to visualize and characterize the spatial distribution of NSCs throughout the tumor. The pattern of NSC distribution showed a gradient with higher densities toward the centroid of the tumor mass. We estimate that NSC-mediated therapy would eradicate 70-90% of the primary tumor mass and the majority of invasive tumor foci. Our method may serve as a model for optimizing the efficacy of NSC-based glioma therapy.
Collapse
Affiliation(s)
- David Lin
- Division of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang C, Wängler B, Morgenstern B, Zentgraf H, Eisenhut M, Untenecker H, Krüger R, Huss R, Seliger C, Semmler W, Kiessling F. Silica- and alkoxysilane-coated ultrasmall superparamagnetic iron oxide particles: a promising tool to label cells for magnetic resonance imaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2007; 23:1427-34. [PMID: 17241069 DOI: 10.1021/la061879k] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
In this study silica- and alkoxysilane-coated ultrasmall superparamagnetic iron oxide (USPIO) particles were synthesized, and their ability to label immortalized progenitor cells for magnetic resonance imaging (MRI) was compared. USPIO particles were synthesized by coprecipitation of ferric and ferrous salts. Subsequently, the particles were coated with silica, (3-aminopropyl)trimethoxysilane (APTMS), and [N-(2-aminoethyl)-3-aminopropyl]trimethoxysilane (AEAPTMS). The size of the USPIO particles was about 10 nm without a significant increase in diameter after coating. The highest T2 relaxivity was achieved for silica-coated USPIO particles, 339.80 +/- 0.22 s-1 mM-1, as compared with APTMS- and AEAPTMS-coated ones, reaching 134.40 +/- 0.01 and 84.79 +/- 0.02 s-1 mM-1, respectively. No toxic effects on the cells could be detected by trypan blue, TUNEL, and MTS assays. Uptake of USPIO particles was evaluated by Prussian blue staining, transmission electron microscopy, T2-MR relaxometry, and mass spectrometry. It was found that cell uptake of the different USPIO particles increased for longer incubation times and higher doses. Maximum cellular iron concentrations of 42.1 +/- 4.0 pg/cell (silica-coated USPIO particles), 37.1 +/- 3.5 pg/cell (APTMS-coated USPIO particles), and 32.7 +/- 4.0 pg/cell (AEAPTMS-coated USPIO particles) were achieved after incubation of the cells with USPIO particles at a dose of 3 micromol/mL for 6 h. The decrease of the T2 relaxation time of the cell pellets was most pronounced for cells incubated with silica-coated USPIO particles followed by APTMS- and AEAPTMS-coated particles, respectively. In gelatin gels even small clusters of labeled cells were detected by 1.5 T MRI, and significant changes in the T2 relaxation times of the gels were determined for 10000 labeled cells/mL for all particles. In summary, as compared with APTMS- and AEAPTMS-coated particles, silica-coated USPIO particles provide the highest T2 relaxivity and most effectively reduce the T2 relaxation time of immortalized progenitor cells after internalization. This suggests silica-coated USPIO particles are most suited for cell labeling approaches in MRI.
Collapse
Affiliation(s)
- Chunfu Zhang
- Junior Group Molecular Imaging and Departments of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Clark PA, Treisman DM, Ebben J, Kuo JS. Developmental signaling pathways in brain tumor-derived stem-like cells. Dev Dyn 2007; 236:3297-308. [DOI: 10.1002/dvdy.21381] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|