1
|
Mahmoudian E, Jahani-Asl A. Establishing Brain Tumor Stem Cell Culture from Patient Brain Tumors and Imaging Analysis of Patient-Derived Xenografts. Methods Mol Biol 2024; 2736:177-192. [PMID: 37243860 DOI: 10.1007/7651_2023_482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Brain tumor stem cells (BTSCs) have been isolated from different types of brain tumors including glioblastoma. Although BTSCs share common characteristics with neural stem cells (NSCs), such as capacity to self-renew and undergo long-term proliferation, they have tumor-propagating capabilities. A small population of BTSC can give rise to secondary tumor when transplanted into severe immunodeficient (SCID) mice. The histological and cytological features, as well as genetic heterogeneity of the xenografted tumors in mice, closely resemble those of primary tumors in patients. Patient-derived xenografts (PDX), therefore, provide a clinically relevant model to study brain tumors. Here, we describe our protocol for establishing BTSC cultures following surgical excision of human brain tumors and the procedures to conduct PDX studies in SCID mice. We also provide our detailed step-by-step protocol on in vivo imaging system (IVIS) of the PDX tumors as a noninvasive method to trace the cells and tumor volume.
Collapse
Affiliation(s)
- Elham Mahmoudian
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Carrano A, Zarco N, Phillipps J, Lara-Velazquez M, Suarez-Meade P, Norton ES, Chaichana KL, Quiñones-Hinojosa A, Asmann YW, Guerrero-Cázares H. Human Cerebrospinal Fluid Modulates Pathways Promoting Glioblastoma Malignancy. Front Oncol 2021; 11:624145. [PMID: 33747938 PMCID: PMC7969659 DOI: 10.3389/fonc.2021.624145] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/05/2021] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary cancer of the central nervous system in adults. High grade gliomas are able to modify and respond to the brain microenvironment. When GBM tumors infiltrate the Subventricular zone (SVZ) they have a more aggressive clinical presentation than SVZ-distal tumors. We suggest that cerebrospinal fluid (CSF) contact contributes to enhance GBM malignant characteristics in these tumors. We evaluated the impact of human CSF on GBM, performing a transcriptome analysis on human primary GBM cells exposed to CSF to measure changes in gene expression profile and their clinical relevance on disease outcome. In addition we evaluated the proliferation and migration changes of CSF-exposed GBM cells in vitro and in vivo. CSF induced transcriptomic changes in pathways promoting cell malignancy, such as apoptosis, survival, cell motility, angiogenesis, inflammation, and glucose metabolism. A genetic signature extracted from the identified transcriptional changes in response to CSF proved to be predictive of GBM patient survival using the TCGA database. Furthermore, CSF induced an increase in viability, proliferation rate, and self-renewing capacity, as well as the migratory capabilities of GBM cells in vitro. In vivo, GBM cells co-injected with human CSF generated larger and more proliferative tumors compared to controls. Taken together, these results provide direct evidence that CSF is a key player in determining tumor growth and invasion through the activation of complex gene expression patterns characteristic of a malignant phenotype. These findings have diagnostic and therapeutic implications for GBM patients. The changes induced by CSF contact might play a role in the increased malignancy of SVZ-proximal GBM.
Collapse
Affiliation(s)
- Anna Carrano
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Natanael Zarco
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Jordan Phillipps
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Paola Suarez-Meade
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Emily S Norton
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biochemical Sciences, Mayo Clinic, Jacksonville, FL, United States.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Kaisorn L Chaichana
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Yan W Asmann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | | |
Collapse
|
3
|
Sustained NF-κB-STAT3 signaling promotes resistance to Smac mimetics in Glioma stem-like cells but creates a vulnerability to EZH2 inhibition. Cell Death Discov 2019; 5:72. [PMID: 30854231 PMCID: PMC6399311 DOI: 10.1038/s41420-019-0155-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is an incurable and highly aggressive brain tumor. Understanding therapeutic resistance and survival mechanisms driving this tumor type is key to finding effective therapies. Smac mimetics (SM) emerged as attractive cancer therapeutics particularly for tumor populations that are highly resistant to conventional apoptosis-inducing therapies. We evaluated the therapeutic efficacy of SM on Glioma stem-like cells (GSCs) and showed that this family of compounds stimulates an adaptive response triggered by TNFα. Increased expression of TNFα results in a prolonged and sustained activation of NF-κB and STAT3 signaling thus activating several tumor cell resistance mechanisms in GSCs. We show that STAT3 activation is contingent on EZH2 activation and uncover a synergistic lethality between SM and EZH2 inhibitors. Therapeutic inhibition of EZH2 impaired the viability of SM-treated GSCs. Our study outlines the molecular underpinnings of SM resistance in glioblastoma and provides mechanistic insight to overcome this resistance and increase therapeutic efficacy.
Collapse
|
4
|
Fakiruddin KS, Ghazalli N, Lim MN, Zakaria Z, Abdullah S. Mesenchymal Stem Cell Expressing TRAIL as Targeted Therapy against Sensitised Tumour. Int J Mol Sci 2018; 19:ijms19082188. [PMID: 30060445 PMCID: PMC6121609 DOI: 10.3390/ijms19082188] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023] Open
Abstract
Tapping into the ability of engineered mesenchymal stem cells (MSCs) to mobilise into the tumour has expanded the scope of cancer treatment. Engineered MSCs expressing tumour necrosis factor (TNF)-related apoptosis inducing ligand (MSC-TRAIL) could serve as a platform for an efficient and targeted form of therapy. However, the presence of cancer stem cells (CSCs) that are resistant to TRAIL and apoptosis may represent a challenge for effective treatment. Nonetheless, with the discovery of small molecular inhibitors that could target CSCs and tumour signalling pathways, a higher efficacy of MSC-TRAIL mediated tumour inhibition can be achieved. This might pave the way for a more effective form of combined therapy, which leads to a better treatment outcome. In this review, we first discuss the tumour-homing capacity of MSCs, its effect in tumour tropism, the different approach behind genetically-engineered MSCs, and the efficacy and safety of each agent delivered by these MSCs. Then, we focus on how sensitisation of CSCs and tumours using small molecular inhibitors can increase the effect of these cells to either TRAIL or MSC-TRAIL mediated inhibition. In the conclusion, we address a few questions and safety concerns regarding the utilization of engineered MSCs for future treatment in patients.
Collapse
Affiliation(s)
- Kamal Shaik Fakiruddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Nadiah Ghazalli
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Moon Nian Lim
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
| | - Zubaidah Zakaria
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
| | - Syahril Abdullah
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
5
|
Serotonin transporter antagonists target tumor-initiating cells in a transgenic mouse model of breast cancer. Oncotarget 2018; 7:53137-53152. [PMID: 27447971 PMCID: PMC5288174 DOI: 10.18632/oncotarget.10614] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/09/2016] [Indexed: 12/18/2022] Open
Abstract
Accumulating data suggests that the initiation and progression of human breast tumors is fueled by a rare subpopulation of tumor cells, termed breast tumor-initiating cells (BTIC), which resist radiotherapy and chemotherapy. Consequently, therapies that abrogate BTIC activity are needed to achieve durable cures for breast cancer patients. To identify such therapies we used a sensitive assay to complete a high-throughput screen of small molecules, including approved drugs, with BTIC-rich mouse mammary tumor cell populations. We found that inhibitors of the serotonin reuptake transporter (SERT) and serotonin receptors, which include approved drugs used to treat mood disorders, were potent inhibitors of mouse BTIC activity as determined by functional sphere-forming assays and the initiation of tumor formation by transplant of drug-exposed tumor cells into syngeneic mice. Moreover, sertraline (Zoloft), a selective serotonin reuptake inhibitor (SSRI), synergized with docetaxel (Taxotere) to shrink mouse breast tumors in vivo. Hence drugs targeting the serotonergic system might be repurposed to treat breast cancer patients to afford more durable breast cancer remissions.
Collapse
|
6
|
Chandrika G, Natesh K, Ranade D, Chugh A, Shastry P. Mammalian target of rapamycin inhibitors, temsirolimus and torin 1, attenuate stemness-associated properties and expression of mesenchymal markers promoted by phorbol-myristate-acetate and oncostatin-M in glioblastoma cells. Tumour Biol 2017; 39:1010428317695921. [PMID: 28351321 DOI: 10.1177/1010428317695921] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway is crucial for tumor survival, proliferation, and progression, making it an attractive target for therapeutic intervention. In glioblastoma, activated mammalian target of rapamycin promotes invasive phenotype and correlates with poor patient survival. A wide range of mammalian target of rapamycin inhibitors are currently being evaluated for cytotoxicity and anti-proliferative activity in various tumor types but are not explored sufficiently for controlling tumor invasion and recurrence. We recently reported that mammalian target of rapamycin inhibitors-rapamycin, temsirolimus, torin 1, and PP242-suppressed invasion and migration promoted by tumor necrosis factor-alpha and phorbol-myristate-acetate in glioblastoma cells. As aggressive invasion and migration of tumors are associated with mesenchymal and stem-like cell properties, this study aimed to examine the effect of mammalian target of rapamycin inhibitors on these features in glioblastoma cells. We demonstrate that temsirolimus and torin 1 effectively reduced the constitutive as well as phorbol-myristate-acetate/oncostatin-M-induced expression of mesenchymal markers (fibronectin, vimentin, and YKL40) and neural stem cell markers (Sox2, Oct4, nestin, and mushashi1). The inhibitors significantly abrogated the neurosphere-forming capacity induced by phorbol-myristate-acetate and oncostatin-M. Furthermore, we demonstrate that the drugs dephosphorylated signal transducer and activator transcription factor 3, a major regulator of mesenchymal and neural stem cell markers implicating the role of signal transducer and activator transcription factor 3 in the inhibitory action of these drugs. The findings demonstrate the potential of mammalian target of rapamycin inhibitors as "stemness-inhibiting drugs" and a promising therapeutic approach to target glioma stem cells.
Collapse
Affiliation(s)
- Goparaju Chandrika
- 1 National Centre for Cell Science (NCCS), Savitribai Phule Pune University, Pune, India
| | - Kumar Natesh
- 1 National Centre for Cell Science (NCCS), Savitribai Phule Pune University, Pune, India
| | - Deepak Ranade
- 2 Department of Neurosurgery, D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Ashish Chugh
- 3 Department of Neurosurgery, CIMET's Inamdar Multispecialty Hospital, Pune, India
| | - Padma Shastry
- 1 National Centre for Cell Science (NCCS), Savitribai Phule Pune University, Pune, India
| |
Collapse
|
7
|
Baharuddin P, Satar N, Fakiruddin KS, Zakaria N, Lim MN, Yusoff NM, Zakaria Z, Yahaya BH. Curcumin improves the efficacy of cisplatin by targeting cancer stem-like cells through p21 and cyclin D1-mediated tumour cell inhibition in non-small cell lung cancer cell lines. Oncol Rep 2015; 35:13-25. [PMID: 26531053 PMCID: PMC4699625 DOI: 10.3892/or.2015.4371] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/18/2015] [Indexed: 01/16/2023] Open
Abstract
Natural compounds such as curcumin have the ability to enhance the therapeutic effectiveness of common chemotherapy agents through cancer stem-like cell (CSC) sensitisation. In the present study, we showed that curcumin enhanced the sensitivity of the double-positive (CD166+/EpCAM+) CSC subpopulation in non-small cell lung cancer (NSCLC) cell lines (A549 and H2170) to cisplatin-induced apoptosis and inhibition of metastasis. Our results revealed that initial exposure of NSCLC cell lines to curcumin (10–40 µM) markedly reduced the percentage of viability to an average of ~51 and ~54% compared to treatment with low dose cisplatin (3 µM) with only 94 and 86% in both the A549 and H2170 cells. Moreover, sensitisation of NSCLC cell lines to curcumin through combined treatment enhanced the single effect induced by low dose cisplatin on the apoptosis of the double-positive CSC subpopulation by 18 and 20% in the A549 and H2170 cells, respectively. Furthermore, we found that curcumin enhanced the inhibitory effects of cisplatin on the highly migratory CD166+/EpCAM+ subpopulation, marked by a reduction in cell migration to 9 and 21% in the A549 and H2170 cells, respectively, indicating that curcumin may increase the sensitivity of CSCs to cisplatin-induced migratory inhibition. We also observed that the mRNA expression of cyclin D1 was downregulated, while a substantial increased in p21 expression was noted, followed by Apaf1 and caspase-9 activation in the double-positive (CD166+/EpCAM+) CSC subpopulation of A549 cells, suggested that the combined treatments induced cell cycle arrest, therefore triggering CSC growth inhibition via the intrinsic apoptotic pathway. In conclusion, we provided novel evidence of the previously unknown therapeutic effects of curcumin, either alone or in combination with cisplatin on the inhibition of the CD166+/EpCAM+ subpopulation of NSCLC cell lines. This finding demonstrated the potential therapeutic approach of using curcumin that may enhance the effects of cisplatin by targeting the CSC subpopulation in NSCLC.
Collapse
Affiliation(s)
- Puteri Baharuddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), Jalan Pahang, Kuala Lumpur 50588, Malaysia
| | - Nazilah Satar
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), University Sains Malaysia, Kepala Batas, Penang 13200, Malaysia
| | - Kamal Shaik Fakiruddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), Jalan Pahang, Kuala Lumpur 50588, Malaysia
| | - Norashikin Zakaria
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), University Sains Malaysia, Kepala Batas, Penang 13200, Malaysia
| | - Moon Nian Lim
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), Jalan Pahang, Kuala Lumpur 50588, Malaysia
| | - Narazah Mohd Yusoff
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), University Sains Malaysia, Kepala Batas, Penang 13200, Malaysia
| | - Zubaidah Zakaria
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), Jalan Pahang, Kuala Lumpur 50588, Malaysia
| | - Badrul Hisham Yahaya
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), University Sains Malaysia, Kepala Batas, Penang 13200, Malaysia
| |
Collapse
|
8
|
Davis B, Shen Y, Poon CC, Luchman HA, Stechishin OD, Pontifex CS, Wu W, Kelly JJ, Blough MD. Comparative genomic and genetic analysis of glioblastoma-derived brain tumor-initiating cells and their parent tumors. Neuro Oncol 2015; 18:350-60. [PMID: 26245525 DOI: 10.1093/neuonc/nov143] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a fatal cancer that has eluded major therapeutic advances. Failure to make progress may reflect the absence of a human GBM model that could be used to test compounds for anti-GBM activity. In this respect, the development of brain tumor-initiating cell (BTIC) cultures is a step forward because BTICs appear to capture the molecular diversity of GBM better than traditional glioma cell lines. Here, we perform a comparative genomic and genetic analysis of BTICs and their parent tumors as preliminary evaluation of the BTIC model. METHODS We assessed single nucleotide polymorphisms (SNPs), genome-wide copy number variations (CNVs), gene expression patterns, and molecular subtypes of 11 established BTIC lines and matched parent tumors. RESULTS Although CNV differences were noted, BTICs retained the major genomic alterations characteristic of GBM. SNP patterns were similar between BTICs and tumors. Importantly, recurring SNP or CNV alterations specific to BTICs were not seen. Comparative gene expression analysis and molecular subtyping revealed differences between BTICs and GBMs. These differences formed the basis of a 63-gene expression signature that distinguished cells from tumors; differentially expressed genes primarily involved metabolic processes. We also derived a set of 73 similarly expressed genes; these genes were not associated with specific biological functions. CONCLUSIONS Although not identical, established BTIC lines preserve the core molecular alterations seen in their parent tumors, as well as the genomic hallmarks of GBM, without acquiring recurring BTIC-specific changes.
Collapse
Affiliation(s)
- Brad Davis
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Yaoqing Shen
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Candice C Poon
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - H Artee Luchman
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Owen D Stechishin
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Carly S Pontifex
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Wei Wu
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - John J Kelly
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Michael D Blough
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | | |
Collapse
|
9
|
Liu H, Cadaneanu RM, Lai K, Zhang B, Huo L, An DS, Li X, Lewis MS, Garraway IP. Differential gene expression profiling of functionally and developmentally distinct human prostate epithelial populations. Prostate 2015; 75:764-76. [PMID: 25663004 PMCID: PMC4409819 DOI: 10.1002/pros.22959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 12/05/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND Human fetal prostate buds appear in the 10th gestational week as solid cords, which branch and form lumens in response to androgen 1. Previous in vivo analysis of prostate epithelia isolated from benign prostatectomy specimens indicated that Epcam⁺ CD44⁻ CD49f(Hi) basal cells possess efficient tubule initiation capability relative to other subpopulations 2. Stromal interactions and branching morphogenesis displayed by adult tubule-initiating cells (TIC) are reminiscent of fetal prostate development. In the current study, we evaluated in vivo tubule initiation by human fetal prostate cells and determined expression profiles of fetal and adult epithelial subpopulations in an effort to identify pathways used by TIC. METHODS Immunostaining and FACS analysis based on Epcam, CD44, and CD49f expression demonstrated the majority (99.9%) of fetal prostate epithelial cells (FC) were Epcam⁺ CD44⁻ with variable levels of CD49f expression. Fetal populations isolated via cell sorting were implanted into immunocompromised mice. Total RNA isolation from Epcam⁺ CD44⁻ CD49f(Hi) FC, adult Epcam⁺ CD44⁻ CD49f(Hi) TIC, Epcam⁺ CD44⁺ CD49f(Hi) basal cells (BC), and Epcam⁺ CD44⁻ CD49f(Lo) luminal cells (LC) was performed, followed by microarray analysis of 19 samples using the Affymetrix Gene Chip Human U133 Plus 2.0 Array. Data was analyzed using Partek Genomics Suite Version 6.4. Genes selected showed >2-fold difference in expression and P < 5.00E-2. Results were validated with RT-PCR. RESULTS Grafts retrieved from Epcam⁺ CD44⁻ fetal cell implants displayed tubule formation with differentiation into basal and luminal compartments, while only stromal outgrowths were recovered from Epcam- fetal cell implants. Hierarchical clustering revealed four distinct groups determined by antigenic profile (TIC, BC, LC) and developmental stage (FC). TIC and BC displayed basal gene expression profiles, while LC expressed secretory genes. FC had a unique profile with the most similarities to adult TIC. Functional, network, and canonical pathway identification using Ingenuity Pathway Analysis Version 7.6 compiled genes with the highest differential expression (TIC relative to BC or LC). Many of these genes were found to be significantly associated with prostate tumorigenesis. CONCLUSIONS Our results demonstrate clustering gene expression profiles of FC and adult TIC. Pathways associated with TIC are known to be deregulated in cancer, suggesting a cell-of-origin role for TIC versus re-emergence of pathways common to these cells in tumorigenesis.
Collapse
Affiliation(s)
- Haibo Liu
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
| | - Radu M Cadaneanu
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
| | - Kevin Lai
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
| | - Baohui Zhang
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
| | - Lihong Huo
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
| | - Dong Sun An
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
- UCLA School of NursingLos Angeles, California
- Broad Stem Cell Center, UCLALos Angeles, California
| | - Xinmin Li
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
- Department of Pathology and Laboratory Medicine, David
Geffen School of Medicine at UCLALos Angeles, California
| | - Michael S Lewis
- West Los Angeles VA Hospital, Greater Los Angeles
Veterans Affairs Healthcare SystemLos Angeles, California
| | - Isla P Garraway
- Department of Urology, David Geffen School of Medicine at
UCLALos Angeles, California
- Jonsson Comprehensive Cancer Center, UCLALos Angeles, California
- Broad Stem Cell Center, UCLALos Angeles, California
- West Los Angeles VA Hospital, Greater Los Angeles
Veterans Affairs Healthcare SystemLos Angeles, California
- *Correspondence to: Isla P. Garraway, Department of Urology, David Geffen
School of Medicine at University of California, Los Angeles, CA, USA. E-mail:
| |
Collapse
|
10
|
Sundar SJ, Hsieh JK, Manjila S, Lathia JD, Sloan A. The role of cancer stem cells in glioblastoma. Neurosurg Focus 2014; 37:E6. [DOI: 10.3171/2014.9.focus14494] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recurrence in glioblastoma is nearly universal, and its prognosis remains dismal despite significant advances in treatment over the past decade. Glioblastoma demonstrates considerable intratumoral phenotypic and molecular heterogeneity and contains a population of cancer stem cells that contributes to tumor propagation, maintenance, and treatment resistance. Cancer stem cells are functionally defined by their ability to self-renew and to differentiate, and they constitute the diverse hierarchy of cells composing a tumor. When xenografted into an appropriate host, they are capable of tumorigenesis. Given the critical role of cancer stem cells in the pathogenesis of glioblastoma, research into their molecular and phenotypic characteristics is a therapeutic priority. In this review, the authors discuss the evolution of the cancer stem cell model of tumorigenesis and describe the specific role of cancer stem cells in the pathogenesis of glioblastoma and their molecular and microenvironmental characteristics. They also discuss recent clinical investigations into targeted therapies against cancer stem cells in the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Jason K. Hsieh
- 1Case Western Reserve University School of Medicine
- 2Cleveland Clinic Lerner College of Medicine
| | - Sunil Manjila
- 3Department of Neurological Surgery, University Hospitals Case Medical Center
| | - Justin D. Lathia
- 2Cleveland Clinic Lerner College of Medicine
- 4Department of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic; and
- 5Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew Sloan
- 1Case Western Reserve University School of Medicine
- 3Department of Neurological Surgery, University Hospitals Case Medical Center
- 5Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
11
|
Karim BO, Rhee KJ, Liu G, Yun K, Brant SR. Prom1 function in development, intestinal inflammation, and intestinal tumorigenesis. Front Oncol 2014; 4:323. [PMID: 25452936 PMCID: PMC4231842 DOI: 10.3389/fonc.2014.00323] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/28/2014] [Indexed: 12/17/2022] Open
Abstract
Prom1/CD133 has been identified in colorectal, hepatocellular, and pancreatic cancer as a cancer stem cell marker and has been used as such to predict colon cancer recurrence in humans. Its potential molecular function as well as its role as a marker of intestinal regeneration is still not fully known. We evaluated the role of Prom1 in intestinal regeneration in inflammatory bowel disease (IBD), determined the function of Prom1, and characterized the effect of a lack of Prom1 on intestinal tumor formation in animal models. Our results suggest that Apc mutations lead to an increase in Prom1 expressing cells in the intestinal crypt stem cell compartment and in early intestinal adenomas. Also, Prom1 knockout mice are more susceptible to intestinal tumor formation. We conclude that Prom1 likely plays a role in regulating intestinal homeostasis and that these results clearly illustrate the role of Prom1 in intestinal regeneration. We further conclude that Prom1 may provide a novel therapeutic target for patients with gastrointestinal conditions such as IBD, short bowel syndrome, and colorectal cancer.
Collapse
Affiliation(s)
- Baktiar O Karim
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University , Baltimore, MD , USA
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, Yonsei University , Gangwon-do, Wonju , South Korea
| | - Guosheng Liu
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University , Baltimore, MD , USA
| | - Kyuson Yun
- The Jackson Laboratory , Bar Harbor, ME , USA
| | - Steven R Brant
- Department of Medicine, The Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
12
|
Abstract
INTRODUCTION Caspase-9 is the apoptotic initiator protease of the intrinsic or mitochondrial apoptotic pathway, which is activated at multi-protein activation platforms. Its activation is believed to involve homo-dimerization of the monomeric zymogens. It binds to the apoptosome to retain substantial catalytic activity. Variety of apoptotic stimuli can regulate caspase-9. However, the mechanism of action of various regulators of caspase-9 has not been summarized and compared yet. In this article, we elucidate the regulators of caspase-9 including microRNAs, natural compounds that are related to caspase-9 and ongoing clinical trials with caspase-9 to better understand the caspase-9 in suppressing cancer. AREAS COVERED In this study, the basic mechanism of apoptosis pathways, regulators of caspase-9 and the development of drugs to regulate caspase-9 are reviewed. Also, ongoing clinical trials for caspase-9 are discussed. EXPERT OPINION Apoptosis has crucial role in cancer, brain disease, aging and heart disease to name a few. Since caspase-9 is an initiator caspase of apoptosis, it is an important therapeutic target of various diseases related to apoptosis. Therefore, a deep understanding on the roles as well as regulators of caspase-9 is required to find more effective ways to conquer apoptosis-related diseases especially cancer.
Collapse
Affiliation(s)
- Bonglee Kim
- Kyunghee University, College of Korean Medicine, Cancer Preventive Material Development Research Center , 1 Hoegi-dong, Dongdaemun-ku, Seoul 131-701 , South Korea
| | | | | |
Collapse
|
13
|
Growth arrest and forced differentiation of human primary glioblastoma multiforme by a novel small molecule. Sci Rep 2014; 4:5546. [PMID: 24989033 PMCID: PMC4080225 DOI: 10.1038/srep05546] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/30/2014] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme is the most common malignant brain tumor in adults, with an average survival of less than one year due to its resistance to therapy. Recent studies reported that GBM initiates from CD133-expressing cancer stem cells (CSC). However, the efficacy of CSC targeting is limited. A newly developed approach in cancer treatment is the forced differentiation of cancer cells. Here, we show that the treatment of the novel small molecule, CG500354, into CD133-expressing human primary GBM cells induces growth arrest by cell cycle regulators, p53, p21, p27 and phase-specific cyclins, and neural differentiation, as confirmed by neural progenitor/precursor markers, nestin, GFAP and Tuj1. When GBM-derived cells caused the tumors in NOD/SCID mice, CG500354 induced GBM-derived cells differentiation into Tuj1 and GFAP expressing cells. We next demonstrated that CG500354 plays a tumor-suppressive role via cAMP/CREB signaling pathway. CG500354 increases not only the extracellular cAMP level but also the protein level of PKA and CREB. Additionally, both mimetic substances, Forskolin and Rolipram, revealed comparable results with CG500354. Our findings indicate that induction of growth arrest and neural differentiation via cAMP/CREB signaling pathway by CG500354 treatment suggests the novel targeting of PDE4D in the development of new drugs for brain tumor therapy.
Collapse
|
14
|
Floyd DH, Zhang Y, Dey BK, Kefas B, Breit H, Marks K, Dutta A, Herold-Mende C, Synowitz M, Glass R, Abounader R, Purow BW. Novel anti-apoptotic microRNAs 582-5p and 363 promote human glioblastoma stem cell survival via direct inhibition of caspase 3, caspase 9, and Bim. PLoS One 2014; 9:e96239. [PMID: 24805821 PMCID: PMC4013090 DOI: 10.1371/journal.pone.0096239] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 04/04/2014] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common and lethal primary brain tumor. Tumor initiation and recurrence are likely caused by a sub-population of glioblastoma stem cells, which may derive from mutated neural stem and precursor cells. Since CD133 is a stem cell marker for both normal brain and glioblastoma, and to better understand glioblastoma formation and recurrence, we looked for dys-regulated microRNAs in human CD133+ glioblastoma stem cells as opposed to CD133+ neural stem cells isolated from normal human brain. Using FACS sorting of low-passage cell samples followed by microRNA microarray analysis, we found 43 microRNAs that were dys-regulated in common in three separate CD133+ human glioblastomas compared to CD133+ normal neural stem cells. Among these were several microRNAs not previously associated with cancer. We then verified the microRNAs dys-regulated in glioblastoma using quantitative real time PCR and Taqman analysis of the original samples, as well as human GBM stem cell and established cell lines and many human specimens. We show that two candidate oncogenic microRNAs, miR-363 and miR-582-5p, can positively influence glioblastoma survival, as shown by forced expression of the microRNAs and their inhibitors followed by cell number assay, Caspase 3/7 assay, Annexin V apoptosis/fluorescence activated cell sorting, siRNA rescue of microRNA inhibitor treatment, as well as 3′UTR mutagenesis to show luciferase reporter rescue of the most successful targets. miR-582-5p and miR-363 are shown to directly target Caspase 3, Caspase 9, and Bim.
Collapse
Affiliation(s)
- Desiree Hunt Floyd
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Ying Zhang
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Bijan K. Dey
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Benjamin Kefas
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Hannah Breit
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Kaitlyn Marks
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Anindya Dutta
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Christel Herold-Mende
- Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Michael Synowitz
- Charité University Clinics, Clinic for Neurosurgery, Berlin, Germany
| | - Rainer Glass
- Neurosurgical Research, University Clinics Munich, München, Germany
| | - Roger Abounader
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
- * E-mail:
| | - Benjamin W. Purow
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, Virginia, United States of America
| |
Collapse
|
15
|
Bayin NS, Modrek AS, Placantonakis DG. Glioblastoma stem cells: Molecular characteristics and therapeutic implications. World J Stem Cells 2014; 6:230-238. [PMID: 24772249 PMCID: PMC3999780 DOI: 10.4252/wjsc.v6.i2.230] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 01/25/2014] [Accepted: 04/11/2014] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma Multiforme (GBM) is a grade IV astrocytoma, with a median survival of 14.6 mo. Within GBM, stem-like cells, namely glioblastoma stem cells (GSCs), have the ability to self-renew, differentiate into distinct lineages within the tumor and initiate tumor xenografts in immunocompromised animal models. More importantly, GSCs utilize cell-autonomous and tumor microenvironment-mediated mechanisms to overcome current therapeutic approaches. They are, therefore, very important therapeutic targets. Although the functional criteria defining GSCs are well defined, their molecular characteristics, the mechanisms whereby they establish the cellular hierarchy within tumors, and their contribution to tumor heterogeneity are not well understood. This review is aimed at summarizing current findings about GSCs and their therapeutic importance from a molecular and cellular point of view. A better characterization of GSCs is crucial for designing effective GSC-targeted therapies.
Collapse
|
16
|
Emlet DR, Gupta P, Holgado-Madruga M, Del Vecchio CA, Mitra SS, Han SY, Li G, Jensen KC, Vogel H, Xu LW, Skirboll SS, Wong AJ. Targeting a glioblastoma cancer stem-cell population defined by EGF receptor variant III. Cancer Res 2014; 74:1238-49. [PMID: 24366881 PMCID: PMC5661963 DOI: 10.1158/0008-5472.can-13-1407] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The relationship between mutated proteins and the cancer stem-cell population is unclear. Glioblastoma tumors frequently express EGFRvIII, an EGF receptor (EGFR) variant that arises via gene rearrangement and amplification. However, expression of EGFRvIII is restricted despite the prevalence of the alteration. Here, we show that EGFRvIII is highly coexpressed with CD133 and that EGFRvIII(+)/CD133(+) defines the population of cancer stem cells (CSC) with the highest degree of self-renewal and tumor-initiating ability. EGFRvIII(+) cells are associated with other stem/progenitor markers, whereas markers of differentiation are found in EGFRvIII(-) cells. EGFRvIII expression is lost in standard cell culture, but its expression is maintained in tumor sphere culture, and cultured cells also retain the EGFRvIII(+)/CD133(+) coexpression, self-renewal, and tumor initiating abilities. Elimination of the EGFRvIII(+)/CD133(+) population using a bispecific antibody reduced tumorigenicity of implanted tumor cells better than any reagent directed against a single epitope. This work demonstrates that a mutated oncogene can have CSC-specific expression and be used to specifically target this population.
Collapse
Affiliation(s)
- David R. Emlet
- Brain Tumor Research Laboratories, Program in Cancer Biology, Stanford University Medical Center, Stanford, CA 94305
| | - Puja Gupta
- Brain Tumor Research Laboratories, Program in Cancer Biology, Stanford University Medical Center, Stanford, CA 94305
| | - Marina Holgado-Madruga
- Department of Physiology and Pharmacology, IBSAL, School of Medicine, University of Salamanca, C/Alfonso X El Sabio s/n. 37007 Salamanca, Spain
| | | | - Siddhartha S. Mitra
- Institute of Stem Cell Biology and Regenerative Medicine, 265 Campus Drive, Stanford, CA 94305
| | - Shuang-Yin Han
- Department of Gastroenterology, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, People’s Republic of China
| | - Gordon Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 943005
| | - Kristin C. Jensen
- Department of Pathology, Stanford University Medical Center, Stanford, CA, 94305
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94305
| | - Hannes Vogel
- Department of Pathology, Stanford University Medical Center, Stanford, CA, 94305
| | - Linda Wei Xu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 943005
| | - Stephen S. Skirboll
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 943005
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94305
| | - Albert J. Wong
- Brain Tumor Research Laboratories, Program in Cancer Biology, Stanford University Medical Center, Stanford, CA 94305
| |
Collapse
|
17
|
Drappatz J, Norden AD, Wen PY. Therapeutic strategies for inhibiting invasion in glioblastoma. Expert Rev Neurother 2014; 9:519-34. [DOI: 10.1586/ern.09.10] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
18
|
Hitting Them Where They Live: Targeting the Glioblastoma Perivascular Stem Cell Niche. CURRENT PATHOBIOLOGY REPORTS 2013; 1:101-110. [PMID: 23766946 DOI: 10.1007/s40139-013-0012-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Glioblastoma growth potential and resistance to therapy is currently largely attributed to a subset of tumor cells with stem-like properties. If correct, this means that cure will not be possible without eradication of the stem cell fraction and abrogation of those mechanisms through which stem cell activity is induced and maintained. Glioblastoma stem cell functions appear to be non-cell autonomous and the consequence of tumor cell residence within specialized domains such as the perivascular stem cell niche. In this review we consider the multiple cellular constituents of the perivascular niche, the molecular mechanisms that support niche structure and function and the implications of the perivascular localization of stem cells for anti-angiogenic approaches to cure.
Collapse
|
19
|
Marsh JC, Goldfarb J, Shafman TD, Diaz AZ. Current Status of Immunotherapy and Gene Therapy for High-Grade Gliomas. Cancer Control 2013; 20:43-8. [DOI: 10.1177/107327481302000107] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- James C. Marsh
- 21st Century Oncology Translational Research Consortium, Peoria, Arizona
| | | | - Timothy D. Shafman
- 21st Century Oncology Translational Research Consortium, Providence, Rhode Island
| | - Aidnag Z. Diaz
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
20
|
Corbeil D, Karbanová J, Fargeas CA, Jászai J. Prominin-1 (CD133): Molecular and Cellular Features Across Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 777:3-24. [DOI: 10.1007/978-1-4614-5894-4_1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Li SC, Vu LT, Ho HW, Yin HZ, Keschrumrus V, Lu Q, Wang J, Zhang H, Ma Z, Stover A, Weiss JH, Schwartz PH, Loudon WG. Cancer stem cells from a rare form of glioblastoma multiforme involving the neurogenic ventricular wall. Cancer Cell Int 2012; 12:41. [PMID: 22995409 PMCID: PMC3546918 DOI: 10.1186/1475-2867-12-41] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 08/10/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The cancer stem cell (CSC) hypothesis posits that deregulated neural stem cells (NSCs) form the basis of brain tumors such as glioblastoma multiforme (GBM). GBM, however, usually forms in the cerebral white matter while normal NSCs reside in subventricular and hippocampal regions. We attempted to characterize CSCs from a rare form of glioblastoma multiforme involving the neurogenic ventricular wall. METHODS We described isolating CSCs from a GBM involving the lateral ventricles and characterized these cells with in vitro molecular biomarker profiling, cellular behavior, ex vivo and in vivo techniques. RESULTS The patient's MRI revealed a heterogeneous mass with associated edema, involving the left subventricular zone. Histological examination of the tumor established it as being a high-grade glial neoplasm, characterized by polygonal and fusiform cells with marked nuclear atypia, amphophilic cytoplasm, prominent nucleoli, frequent mitotic figures, irregular zones of necrosis and vascular hyperplasia. Recurrence of the tumor occurred shortly after the surgical resection. CD133-positive cells, isolated from the tumor, expressed stem cell markers including nestin, CD133, Ki67, Sox2, EFNB1, EFNB2, EFNB3, Cav-1, Musashi, Nucleostemin, Notch 2, Notch 4, and Pax6. Biomarkers expressed in differentiated cells included Cathepsin L, Cathepsin B, Mucin18, Mucin24, c-Myc, NSE, and TIMP1. Expression of unique cancer-related transcripts in these CD133-positive cells, such as caveolin-1 and -2, do not appear to have been previously reported in the literature. Ex vivo organotypic brain slice co-culture showed that the CD133+ cells behaved like tumor cells. The CD133-positive cells also induced tumor formation when they were stereotactically transplanted into the brains of the immune-deficient NOD/SCID mice. CONCLUSIONS This brain tumor involving the neurogenic lateral ventricular wall was comprised of tumor-forming, CD133-positive cancer stem cells, which are likely the driving force for the rapid recurrence of the tumor in the patient.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County (CHOC) Research Institute, 455 South Main Street, Orange, CA 92868, USA
- Department of Neurology, University of California Irvine, Orange, CA 92862, USA
- Department of Biological Science, California State University, Fullerton, CA, 92834, USA
| | - Long T Vu
- Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County (CHOC) Research Institute, 455 South Main Street, Orange, CA 92868, USA
- Department of Biological Science, California State University, Fullerton, CA, 92834, USA
| | - Hector W Ho
- Department of Neurological Surgery, Saint Joseph Hospital, Orange, CA, 92868, USA
- Department of Neurological Surgery, University of California Irvine, Orange, CA, 92862, USA
| | - Hong Zhen Yin
- Department of Neurology, University of California Irvine, Orange, CA 92862, USA
| | - Vic Keschrumrus
- Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County (CHOC) Research Institute, 455 South Main Street, Orange, CA 92868, USA
| | - Qiang Lu
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Jun Wang
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Heying Zhang
- Department of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Zhiwei Ma
- Department of Pathology and Laboratory Medicine, Good Samaritan Hospital Medical Center, 1000 Montauk Highway, West Islip, NY, 11795, USA
| | - Alexander Stover
- National Human Neural Stem Cell Resource, Center for Neuroscience and Stem Cell Research, CHOC Children's Hospital Research Institute, 455 South Main Street, Orange, CA, 92868, USA
| | - John H Weiss
- Department of Neurology, University of California Irvine, Orange, CA 92862, USA
| | - Philip H Schwartz
- National Human Neural Stem Cell Resource, Center for Neuroscience and Stem Cell Research, CHOC Children's Hospital Research Institute, 455 South Main Street, Orange, CA, 92868, USA
- Developmental Biology Center, University of California Irvine, Irvine, CA, 92612, USA
| | - William G Loudon
- Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County (CHOC) Research Institute, 455 South Main Street, Orange, CA 92868, USA
- Department of Neurological Surgery, Saint Joseph Hospital, Orange, CA, 92868, USA
- Department of Neurological Surgery, University of California Irvine, Orange, CA, 92862, USA
| |
Collapse
|
22
|
Cortes-Dericks L, Galetta D, Spaggiari L, Schmid RA, Karoubi G. High expression of octamer-binding transcription factor 4A, prominin-1 and aldehyde dehydrogenase strongly indicates involvement in the initiation of lung adenocarcinoma resulting in shorter disease-free intervals. Eur J Cardiothorac Surg 2012; 41:e173-81. [PMID: 22529186 DOI: 10.1093/ejcts/ezs170] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The increasing relevance of the cancer stem cell (CSC) hypothesis and the impact of CSC-associated markers in the carcinogenesis of solid tumours may provide potential prognostic implications in lung cancer. We propose that a collective genetic analysis of established CSC-related markers will generate data to better define the role of putative CSCs in lung adenocarcinoma (LAC). METHODS Sixty-four paired tumour and non-tumour biopsies from LAC patients were included in this study. Using the quantitative reverse transcriptase-polymerase chain reaction, we assessed the expression profiles of established CSC-related biomarkers: octamer-binding transcription factor 4 (OCT4A), CD133, aldehyde dehydrogenase (ALDH), BMI-1, ATP-binding cassette subfamily G, member 2 (ABCG2), SRY (sex-determining region Y)-box 2 (SOX2) and uPAR, and evaluated their relation to clinicopathological parameters and disease prognosis. RESULTS All of the above-mentioned CSC-related markers were detectable in both tumour and corresponding normal tissues. Importantly, expression levels of OCT4A, CD133, BMI-1, SOX2 and uPAR were significantly higher (OCT4A, P = 0.0003; CD133, P = 0.002; BMI-1, P = 0.04; SOX2, P = 0.0003; uPAR, P = 0.03) in the tumour compared with those in the non-tumour tissues. By contrast, the quantities of ACBG2 and ALDH were markedly reduced (ACBG2, P = 0.0006; ALDH, P = 0.007) in the tumour relative to those in the normal biopsies. Using multivariate analysis, elevated ALDH and CD133 revealed significant associations in tumour stage (ALDH, P = 0.03; CD133, P = 0.007) and differentiation (ALDH, P = 0.03; CD133, P = 0.018). We observed that ALDH and OCT4A were associated with nodal status (ALDH, P = 0.05; OCT4A, P = 0.03) having lower mRNA levels in tumours with lymph node metastasis, N+, compared with that in N0. High OCT4A levels were significantly correlated with tumour size of <3 cm, decrease in tumours >3 cm (P = 0.03). Kaplan-Meier correlation analyses, showed that OCT4A and CD133 were correlated to short disease-free intervals (OCT4A, P = 0.047; CD133, P = 0.033) over a period of 29 months. CONCLUSIONS Our study reveals that CSC-associated markers: OCT4A, CD133 and ALDH are involved in the initial phase of carcinogenesis of LAC, and can be used as predictors of early stage LAC and poor disease-free intervals. In addition, this work validates the relevance of the CSC hypothesis in LAC.
Collapse
Affiliation(s)
- Lourdes Cortes-Dericks
- Department of Clinical Research, Division of General Thoracic Surgery, University Hospital Bern, Bern, Switzerland
| | | | | | | | | |
Collapse
|
23
|
Marsh JC, Wendt JA, Herskovic AM, Diaz A, Gielda BT, Byrne RW. High-Grade Glioma Relationship to the Neural Stem Cell Compartment: A Retrospective Review of 104 Cases. Int J Radiat Oncol Biol Phys 2012; 82:e159-65. [DOI: 10.1016/j.ijrobp.2010.08.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 07/08/2010] [Accepted: 08/19/2010] [Indexed: 12/18/2022]
|
24
|
Abstract
Cells with certain attributes of very immature astroglial cells and their radial precursors can act as stem and/or progenitor cells during developmental and persistent neurogenesis. Neural stem/progenitor cells both express and are affected by a variety of developmentally regulated macromolecules and growth factors, and such signaling or recognition molecules are being uncovered through extensive genomic and proteomic studies, as well as tested using in vitro/in vivo cell growth bioassays. Glycosylated molecules are appreciated as distinct signaling molecules during morphogenesis in a variety of tissues and organs, with glycoconjugates (glycoproteins, glycolipids, and glycosaminoglycans) serving as mediators for the interactions of cells with each other and their substrates, to confer growth and differentiation cues to precursor cells in search of identity. Neurogenic astrocytes and associated glycoconjugates, especially extracellular matrix molecules, are discussed in the context of neurogenesis and stem/progenitor cell growth, fate choice, and differentiation.
Collapse
Affiliation(s)
- Dennis A Steindler
- Department of Neuroscience, The Evelyn F. and William L. McKnight Brain Institute, The University of Florida, Gainesville, FL, USA.
| |
Collapse
|
25
|
Abstract
Pancreatic cancer stem-like cells are described by membrane expression of CD24, CD44 and ESA (epithelial-specific antigen) and their capacity to grow as spheres in a serum-free medium containing well-defined growth factors. The capacity of a panel of four pancreatic cancer cell lines (PANC-1, CFPAC-1, PancTu-1 and PSN-1) to form spheres was tested. All cell lines with the exception of PancTu-1 developed spheres. Phenotypically, the sphere-growing cells showed an increased in vitro invasion capability. Both gene and protein expressions of markers of metastases [CXCR4 (CXC chemokine receptor 4), OPN (osteopontin) and CD44v6] and components of active hedgehog pathway signalling were assessed. Spheres clearly demonstrated increased expression of the above-mentioned markers when compared with their adherent counterpart. With the aim of identifying a minimum set of markers able to separate cells that have the capacity to form spheres from those incapable of forming spheres, a PCA (principal component analysis) of the multidimensional dataset was performed. Although PCA of the ‘accepted’ stemness genes was unable to separate sphere-forming from sphere-incapable cell lines, the addition of the ‘aggressiveness’ marker CD44v6 allowed a clear differentiation. Moreover, inoculation of the spheres and the adherent cells in vivo confirmed the superior aggressiveness (proliferation and metastasis) of the spheres over the adherent cells. In conclusion, the present study suggests that the sphere-growing cell population is not only composed of cells displaying classical stem membrane markers but also needs CD44v6-positive cells to successfully form spheres. Our results also emphasize the potential therapeutic importance of pathways such as CXCR4 and hedgehog for pancreatic cancer treatment.
Collapse
|
26
|
CD133 positive embryonal rhabdomyosarcoma stem-like cell population is enriched in rhabdospheres. PLoS One 2011; 6:e19506. [PMID: 21602936 PMCID: PMC3094354 DOI: 10.1371/journal.pone.0019506] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/30/2011] [Indexed: 12/25/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified in a number of solid tumors, but not yet in rhabdomyosarcoma (RMS), the most frequently occurring soft tissue tumor in childhood. Hence, the aim of this study was to identify and characterize a CSC population in RMS using a functional approach. We found that embryonal rhabdomyosarcoma (eRMS) cell lines can form rhabdomyosarcoma spheres (short rhabdospheres) in stem cell medium containing defined growth factors over several passages. Using an orthotopic xenograft model, we demonstrate that a 100 fold less sphere cells result in faster tumor growth compared to the adherent population suggesting that CSCs were enriched in the sphere population. Furthermore, stem cell genes such as oct4, nanog, c-myc, pax3 and sox2 are significantly upregulated in rhabdospheres which can be differentiated into multiple lineages such as adipocytes, myocytes and neuronal cells. Surprisingly, gene expression profiles indicate that rhabdospheres show more similarities with neuronal than with hematopoietic or mesenchymal stem cells. Analysis of these profiles identified the known CSC marker CD133 as one of the genes upregulated in rhabdospheres, both on RNA and protein levels. CD133(+) sorted cells were subsequently shown to be more tumorigenic and more resistant to commonly used chemotherapeutics. Using a tissue microarray (TMA) of eRMS patients, we found that high expression of CD133 correlates with poor overall survival. Hence, CD133 could be a prognostic marker for eRMS. These experiments indicate that a CD133(+) CSC population can be enriched from eRMS which might help to develop novel targeted therapies against this pediatric tumor.
Collapse
|
27
|
Dong L, Qi N, Ge RM, Cao CL, Lan F, Shen L. Overexpression of CD133 promotes the phosphorylation of Erk in U87MG human glioblastoma cells. Neurosci Lett 2010; 484:210-4. [DOI: 10.1016/j.neulet.2010.08.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 08/15/2010] [Accepted: 08/19/2010] [Indexed: 12/19/2022]
|
28
|
Abstract
Glioblastoma multiforme is an extremely aggressive and clinically unresponsive form of cancer. Transformed neoplastic neural stem cells, resistant to chemotherapy and radiation therapy, are thought to be responsible for the initial tumor formation and the recurrence of disease following surgical resection. These stem cells express multidrug resistance markers along with CD133. We show that ectopic overexpression of CD133 in rat C6 glioma cells leads to significant reluctance to undergo apoptosis from camptothecin and doxorubicin. Although p53 was upregulated in CD133-overexpressing glioma cells treated with DNA-damaging agents, apoptosis seems to be p53 independent. At least one ABC transporter, rat P-glycoprotein/ABCB1, was upregulated by 62% in CD133(+) cells with a corresponding increase in activity. Thus, the combination of higher P-glycoprotein mRNA transcription and elevated transporter activity seems to contribute to the protection from cytotoxic reagents. In conclusion, previous investigators have reported that resilient cancer stem cells coexpress CD133 and ABC transporters with increased reluctance toward apoptosis. Our data suggest that CD133 may contribute to the observed resistance to apoptosis of CD133(+) cancer stem cells.
Collapse
Affiliation(s)
- James M Angelastro
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
29
|
Annabi B, Doumit J, Plouffe K, Laflamme C, Lord-Dufour S, Béliveau R. Members of the low-density lipoprotein receptor-related proteins provide a differential molecular signature between parental and CD133+ DAOY medulloblastoma cells. Mol Carcinog 2010; 49:710-717. [PMID: 20564348 DOI: 10.1002/mc.20645] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the low-density lipoprotein receptor-related protein (LRP) family are involved in metabolic stress and resistance phenotypes of cancer cells. New breakthroughs in brain cancer therapy have exploited that molecular signature and proved that efficient delivery of therapeutic agents involve LRP-mediated mechanisms. We performed gene expression profiling of CD133, a cell surface cancer stem cell marker, and of LRP in response to in vitro nutrient deprivation. We found that CD133 was selectively induced in serum-starved DAOY medulloblastoma cells but not in U87MG glioblastoma cells. Such CD133 induction was correlated to increases in LRP-1 and LRP-1b gene and protein expression. When a specific CD133(+) DAOY cell population was sorted from parental DAOY, we found increases in LRP-5 and LRP-8. Uptake of alpha(2)-macroglobulin, a specific LRP-1/1b ligand, was increased in serum-starved parental DAOY cells but not in CD133(+) DAOY cells, and receptor-associated protein (RAP), which binds to all cell surface LRPs, was able to compete for that uptake. Conversely, RAP binding was increased in serum-starved parental DAOY but alpha(2)-macroglobulin was unable to compete for such uptake. Strategies aiming at targeting cancer stem cell metabolic adaptative responses, such as that through LRP differential expression within the brain tissue microenvironmental niche, can now be envisioned.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
30
|
AC133 expression in egyptian children with acute leukemia: impact on treatment response and disease outcome. J Pediatr Hematol Oncol 2010; 32:286-93. [PMID: 20224439 DOI: 10.1097/mph.0b013e3181c80c08] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
AC133 antigen is expressed restrictively in the immature subset of the CD34 cells. Hence, it is expected to be a valuable prognostic marker in acute leukemia. Sixty Egyptian children with acute leukemia were enrolled into this prospective study divided into 2 groups: 30 acute myeloblastic leukemia (AML) and 30 acute lymphoblastic leukemia (ALL) patients. Flow cytometric assessment of AC133 expression was performed on CD34 blast cells. AC133 was expressed in 66.7% and 40% of AML and ALL patients, respectively. AC133-positive expression was not associated with any of the studied standard prognostic factors. In AML, 80% of patients with poor clinical outcome (relapse or death) were positive for AC133 expression, whereas, all ALL patients who developed resistance as well as those who displayed poor clinical outcome had AC133-positive expression (P<0.05). Patients with positive AC133 expression had significantly shorter overall and disease-free survival times compared with AC133-negative patients in both ALL (P<0.001) and AML (P<0.05) groups. AC133 expression percentage was a reliable poor prognostic marker in ALL patients (P<0.0001). AC133-positive expression is an independent poor prognostic factor in childhood acute leukemia and could characterize a group of patients with resistance to standard chemotherapy, as well as high incidence of relapse and death.
Collapse
|
31
|
Achanta P, Roman NIS, 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 PMCID: PMC2981502 DOI: 10.2174/187152010790909290] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
| | - Neda I Sedora Roman
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
- University of Puerto Rico School of Medicine, Office A-873, PO BOX 365067, San Juan PR 00936-5067
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
| |
Collapse
|
32
|
Abstract
OBJECTIVES Gliomas are an important form of brain cancer, with high mortality rate. Mathematical models are often used to understand and predict their behaviour. However, using current modeling techniques one must choose between simulating individual cell behaviour and modeling tumours of clinically significant size. MATERIALS AND METHODS We propose a hybrid compartment-continuum-discrete model to simulate glioma growth and malignant cell invasion. The discrete portion of the model is capable of capturing intercellular interactions, including cell migration, intercellular communication, spatial cell population heterogeneity, phenotype differentiation, epigenetic events, proliferation, and apoptosis. Combining this with a compartment and continuum model allows clinically significant tumour sizes to be evaluated. RESULTS AND CONCLUSIONS This model is used to perform multiple simulations to determine sensitivity to changes in important model parameters, specifically, the fundamental length parameter, necrotic cell degradation rate, rate of cell migration, and rate of phenotype transformation. Using these values, the model is able to simulate tumour growth and invasion behaviour, observed clinically. This mathematical model provides a means to simulate various tumour development scenarios, which may lead to a better understanding of how altering fundamental parameters can influence neoplastic progression.
Collapse
Affiliation(s)
- M L Tanaka
- Department of Orthopaedic Surgery, Wake Forest University, Winston-Salem, North Carolina, USA
| | | | | |
Collapse
|
33
|
Boivin D, Labbé D, Fontaine N, Lamy S, Beaulieu É, Gingras D, Béliveau R. The Stem Cell Marker CD133 (Prominin-1) is Phosphorylated on Cytoplasmic Tyrosine-828 and Tyrosine-852 by Src and Fyn Tyrosine Kinases. Biochemistry 2009; 48:3998-4007. [DOI: 10.1021/bi900159d] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Dominique Boivin
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - David Labbé
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - Nicolas Fontaine
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - Sylvie Lamy
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - Édith Beaulieu
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - Denis Gingras
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
| | - Richard Béliveau
- Laboratoire de Médecine Moléculaire, Chemistry Department, Université du Québec à Montréal, C.P. 8888, succ. centre-ville, Montréal, Québec, Canada H3C 3P8
- Holder of the “Chaire en prévention et traitement du cancer” from Université du Québec à Montréal and of the Claude-Bertrand Chair in Neurosurgery from Université de Montréal
| |
Collapse
|
34
|
Abstract
The tolerance of normal brain tissues limits the radiation dose that can be delivered safely during cranial radiotherapy, and one of the potential complications that can arise involves cognitive impairment. Extensive laboratory data have appeared recently showing that hippocampal neurogenesis is significantly impacted by irradiation and that such changes are associated with altered cognitive function and involve, in part, changes in the microenvironment (oxidative stress and inflammation). Although there is considerable uncertainty about exactly how these changes evolve, new in vitro and in vivo approaches have provided a means by which new mechanistic insights can be gained relevant to the topic. Together, the data from cell culture and animal-based studies provide complementary information relevant to a potentially serious complication of cranial radiotherapy and should enhance our understanding of the tolerance of normal brain after cranial irradiation.
Collapse
Affiliation(s)
- John R Fike
- Brain and Spinal Injury Center, University of California, San Francisco, CA 94110-0899, USA.
| | | | | |
Collapse
|
35
|
Abstract
The trophoblastic theory of cancer, proposed in the early 1900s by Dr John Beard, may not initially seem relevant to current cancer models and treatments. However, the underpinnings of this theory are remarkably similar to those of the cancer stem cell (CSC) theory. Beard noticed that a significant fraction of germ cells never reach their final destination as they migrate during embryonic development from the hindgut to the germinal ridge. In certain situations, upon aberrant stimulation, these vagrant germ cells are able to generate tumors. Simplistically, the CSC theory surmises that a small population of tumorigenic cells exists, which initiate and maintain tumors, and these cells have a likely origin in normal stem cells. Both these theories are based on the potential of a single primitive cell to form a tumor. This has a major implication for cancer therapy, in that only a small percentage of cells need to be targeted to ablate a tumor.
Collapse
Affiliation(s)
- Angela R Burleigh
- Department of Pathology and Laboratory Medicine, University of British Columbia, British Columbia, Canada.
| |
Collapse
|
36
|
Annabi B, Rojas-Sutterlin S, Laflamme C, Lachambre MP, Rolland Y, Sartelet H, Béliveau R. Tumor environment dictates medulloblastoma cancer stem cell expression and invasive phenotype. Mol Cancer Res 2008; 6:907-916. [PMID: 18567795 DOI: 10.1158/1541-7786.mcr-07-2184] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neural precursor surface marker CD133 is thought to be enriched in brain cancer stem cells and in radioresistant DAOY medulloblastoma-derived tumor cells. Given that membrane type-1 matrix metalloproteinase (MT1-MMP) expression is a hallmark of highly invasive, radioresistant, and hypoxic brain tumor cells, we sought to determine whether MT1-MMP and other MMPs could regulate the invasive phenotype of CD133(+) DAOY cells. We found that when DAOY medulloblastoma or U87 glioblastoma cells were implanted in nude mice, only those cells specifically implanted in the brain environment generated CD133(+) brain tumors. Vascular endothelial growth factor and basic fibroblast growth factor gene expression increases in correlation with CD133 expression in those tumors. When DAOY cultures were induced to generate in vitro neurosphere-like cells, gene expression of CD133, MT1-MMP, MMP-9, and MDR-1 was induced and correlated with an increase in neurosphere invasiveness. Specific small interfering RNA gene silencing of either MT1-MMP or MMP-9 reduced the capacity of the DAOY monolayers to generate neurospheres and concomitantly abrogated their invasive capacity. On the other hand, overexpression of MT1-MMP in DAOY triggered neurosphere-like formation which was further amplified when cells were cultured in neurosphere medium. Collectively, we show that both MT1-MMP and MMP-9 contribute to the invasive phenotype during CD133(+) neurosphere-like formation in medulloblastoma cells. Increases in MMP-9 may contribute to the opening of the blood-brain barrier, whereas increased MT1-MMP would promote brain tumor infiltration. Our study suggests that MMP-9 or MT1-MMP targeting may reduce the formation of brain tumor stem cells.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal, CP 8888, Succursale Centre-ville, Montreal, Quebec, Canada H3C 3P8
| | | | | | | | | | | | | |
Collapse
|
37
|
Wu A, Oh S, Wiesner SM, Ericson K, Chen L, Hall WA, Champoux PE, Low WC, Ohlfest JR. Persistence of CD133+ cells in human and mouse glioma cell lines: detailed characterization of GL261 glioma cells with cancer stem cell-like properties. Stem Cells Dev 2008; 17:173-84. [PMID: 18271701 DOI: 10.1089/scd.2007.0133] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The concept of cancer stem cells suggests that there are malignant stem-like cells within a tumor that are responsible for tumor renewal and resistance to cytotoxic therapies. Studies have identified glioma stem-like cells that extrude Hoechst 33342 dye, representing a double-negative "side population" (SP) thought to be selectively resistant to drug therapy. A CD133+ stem cell-like subpopulation has been isolated from a human glioma that was enriched for tumor-initiating cells. It is unknown whether CD133+ cells with similar phenotype persist in established glioma cell lines, or if CD133 is a marker of glioma stem-like cells in rodents. We investigated whether CD133+ and SP cells existed in the GL261 cell line, a syngeneic mouse glioma model that is widely used for preclinical and translational research. Intracerebral injection of less than 100 CD133+ GL261 cells formed tumors, whereas it required 10,000 CD133(-) cells to initiate a tumor. CD133+ GL261 cells expressed nestin, formed tumor spheres with high frequency, and differentiated into glial and neuronal-like cells. Similar to GL261, seven human glioma cell lines analyzed also contained a rare CD133+ population. Surprisingly, we found that CD133+ GL261 cells did not reside in the SP, nor did the majority ( approximately 94%) of CD133+ human glioma cells. These results demonstrate that the expression of CD133 in murine glioma cells is associated with enhanced tumorigenicity and a stem-like phenotype. This study also reveals a previously unrecognized level of heterogeneity in glioma cell lines, exposing several populations of cells that have characteristics of cancer stem cells.
Collapse
Affiliation(s)
- Anhua Wu
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lichtenauer UD, Shapiro I, Geiger K, Quinkler M, Fassnacht M, Nitschke R, Rückauer KD, Beuschlein F. Side population does not define stem cell-like cancer cells in the adrenocortical carcinoma cell line NCI h295R. Endocrinology 2008; 149:1314-22. [PMID: 18063677 DOI: 10.1210/en.2007-1001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence suggests the existence of a stem cell-like subpopulation of cells in hematological and solid tumor entities, which determine the malignant phenotype of a given tumor through their proliferative potential and chemotherapy resistance. A recently used technique for the isolation of this cell population is through exclusion of the vital dye Hoechst 33342, which defines the so-called side population (SP). Herein we demonstrate the presence of SP cells in a variety of adrenal specimens, including primary cultures of human adrenocortical tumors and normal adrenal glands as well as established human and murine adrenocortical cancer cell lines by fluorescence-activated cell sorter analysis and confocal microscopy. On a functional level, SP cells from the human adrenocortical tumor cell line NCI h295R revealed an expression pattern consistent with a less differentiated phenotype, including lower expression of steroidogenic enzymes such as steroid acute regulatory protein (StAR) and side-chain cleavage enzyme (P450scc) in comparison with non-SP cells. However, proliferation between SP and non-SP cells did not differ (105.6 +/- 18.1 vs. 100.0 +/- 3.5%). Furthermore, re-sorting and tracing experiments revealed the capacity for both cell types to give rise to the original SP- and non-SP-containing cell population. Similarly to the baseline growth kinetics, no survival benefit was evident in SP cells after treatment with cytotoxic agents commonly used in adrenocortical carcinomas. Taken together, these findings provide evidence that Hoechst dye exclusion, in contrast to what has been reported for other tumor entities, is not a major tumor stem cell defining marker in adrenocortical NCI h295R tumor cells.
Collapse
Affiliation(s)
- Urs D Lichtenauer
- Institute of Molecular Medicine and Cell Research, Centre for Systems Biology, Albert-Ludwigs-University Freiburg, D-79085 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|