Abstract
Glioblastoma is the most common type of primary brain tumor in adults and is among the most lethal and least successfully treated solid tumors. Recently, research into the area of stem cells in brain tumors has gained momentum. However, due to the relatively new and novel hypothesis that a subpopulation of cancer cells in each malignancy has the potential for tumor initiation and repopulation, the data in this area of research are still in its infancy. This review article is aimed at attempting to bring together research carried out so far in order to build an understanding of glioblastoma stem cells (GSCs). Initially, we consider GSCs at a morphological and cellular level, and then discuss important cell markers, signaling pathways and genetics. Furthermore, we highlight the difficulties associated with what some of the evidence indicates and what collectively the studies contribute to further defining the interpretation of GSCs.
Keywords: cell marker, genetics, glioblastoma, morphology, signaling pathway, stem cells
INTRODUCTION
Recently, research into the area of stem cells in brain tumors has gained momentum. Indeed, the first evidence for the existence of any subpopulation of cancer stem cells (CSCs) within a tumor came in 1994 from studying acute myeloid leukemia (92). As such, due to the relatively new and novel experimental evidence that CSCs exist and have the potential for tumor initiation and repopulation, the data in this area of research are still in its infancy. Thus, it is the purpose of this review to attempt to bring together some of the research in order to attempt to define the characteristic features of stem cells in glioblastomas, covering the morphological features, cellular biology and genetics.
There are two main reasons why a review is useful. First, interest in this area of oncology has increased dramatically in recent years. This is evidential from the amount of research being produced and published on the subject of glioblastoma stem cells (GSCs). Using a simple keyword search of “glioblastoma stem cell” in key journal search engines produced thousands of related articles in 2011: ScienceDirect, 5541; MEDLINE, 7266; and PubMed, 832. There was a significant increase compared with the same search dated up to and including 1992: ScienceDirect, 496; MEDLINE, 832; PubMed, 44.
Second, glioblastoma is the most common type of primary brain tumor in adults and is among the most lethal and least successfully treated solid tumors (46), occurring in 2–3 cases per 100 000 people in Europe and North America (167). The median survival time of patients with glioblastoma multiforme (GBM) following treatment is 12 months, with only 3–5% of patients surviving longer than 5 years 32, 56. Compared with the advances in the treatment of other types of tumors, the poor prognosis for GBM patients has improved minimally over decades, underscoring the challenges and difficulties in effectively detecting and treating these fatal cancers (27).
Such a poor prognosis has several causes, including the extensive dissemination of tumor cells, making total surgical resection almost, if not completely, impossible; the tumor is highly refractory to available chemotherapeutic and radiation treatments and there is still insufficient understanding of glioblastomas. This review article is therefore aimed at attempting to bring together research carried out so far in order to build an understanding of GSCs.
The nomenclature of stem cells in glioblastomas remains unresolved and thus far there are no standardized criteria for identifying them. There are a number of terms used to denote the same concept including GSC, glioma tumor initiating cells and brain tumor stem‐like cells. The terminology reflects some shared characteristics with normal stem cells, especially adult somatic stem cells, including the capacities for self‐renewal, differentiation and maintained proliferation (153). Furthermore, it is known that cancer stem cells are distinct from the cell of origin, indeed the phenotype of the cell of origin may differ substantially from that of the cancer stem cell (165). It is suggested that cancer stem cells do not necessarily originate from the transformation of normal stem cells, but rather from restricted progenitors or more differentiated cells that have acquired self‐renewing capacity (166). In this article, the term GSC is used.
MORPHOLOGICAL AND CELLULAR BIOLOGY OF GSCs, AND DIFFICULTY IN LOCATING GSC NICHES
Table 1 shows that there are a number of morphological and biological features that can distinguish GSCs. Despite the strong evidence for the existence of the three main areas of neuronal production, more recently there have been several reports of adult neurogenesis in other regions of the brain, including the striatum 33, 102, the amygdala 13, 41, the hypothalamus 74, 176 and the substantia nigra (183), as well as in the brain stem (11) and olfactory tubercle (12). However, there are also studies which have failed to confirm these findings 26, 30, 44 or only confirmed the finding following damage or pharmacological intervention (57). If the origin of neural stem cells can be located, then a more precise strike could be dealt when delivering treatment for glioblastomas. However, there are two main problems which would still have to be resolved, even if neural stem cell niches are located.
Table 1.
Morphological and biological characteristics of GSCs. Abbreviations: GBM = glioblastoma multiforme; GSC = glioblastoma stem cell
| Feature | Evidence of existence in GSCs | Reference |
|---|---|---|
| Origin of glioblastomas | Cerebral hemispheres, typically at the cortical/subcortical interface and spread through the white matter, occasionally to the opposite hemisphere through the corpus callosum. GBM cells are extremely infiltrative, often migrating along the basement membrane of blood vessels or along myelinated white matter. | 14, 101 |
| Origin of neural stem cells | Three sites of neuronal production in the adult mammalian brain; the subgranular zone of the dentate gyrus of the hippocampus, the subventricular zone which lines the lateral walls of the ventricles and the subependymal zone of the spinal cord | 2, 47, 48, 81 |
| Migrating ability | Possess the ability to migrate and differentiate after transplantation into a rodent brain. It has also been shown that neuronal precursors can migrate to a damaged environment within the brain which has resulted from ischemia or the presence of a tumor. | 1, 67 |
| Cellular biology | GSCs have many of the same characteristics of normal stem cells such as the formation of floating neurospheres, more G0/G1 phase cells, more resistance to hypoxia, irradiations and some chemotherapeutics, higher colony‐formation ability, and high capacity to develop into multiple lineages. | (124) |
| Histology | Indistinguishable from normal stem cells | (165) |
| Karyotype analysis | GSCs have been shown to have aneuploidy of specific autosomes and sex chromosomes; specifically monosomy 1, trisomy 7 and 22, and disomy X. | (49) |
| Proteins | Express high levels of ATP‐binding cassette transporters and proteins related to drug resistance, such as P‐glycoprotein, multidrug resistant protein (MRP)‐1, MRP3, MRP5, breast cancer drug‐resistance protein, and glutathione‐S‐transferase | 124, 180 |
| Enzymes | Expression of matrix metalloproteinase‐13 was specifically expressed in GSCs, indicating they have highly invasive potential. | (76) |
| Number of GSCs needed for tumor formation | Analysis of mouse brains following CD133+ engraftment revealed that as few as 100 CD133+ cells were sufficient for the formation of human brain tumors in mice. | (143) |
First, neural stem cells differ according to the region of brain in which they are located. The detection of a genetic signature specific to cerebella vs. neocortical neural stem cells suggests that morphologically indistinguishable stem cells from different brain regions are unique (138). Two observable differences in neural progenitor cells (NPCs) which relate to their origin are: (i) NPCs from the rostral part of the brain tended to proliferate faster than those from the caudal part; and (ii) the NPCs from the diencephalon and mesencephalon gave rise to more tyrosine hydroxylase‐positive neurons than did those from the telencephalon and rhombencephalon (70). If neural stem cells differ according to the region of the brain in which they are located, then it may also be the case that GSCs also differ according to the region of brain in which they are located. Thus it may be that, depending on where the glioblastoma precisely originates, certain types of GSCs may have specific mutations which are unique to their location of origin.
Second, evidence suggests that neural stem cells can migrate, a feature which may also be attributed to GSCs. The extent of this is considerable as it has also been shown that mesenchymal stem cells (which can transdifferentiate into neural cells in vitro under the influence of matrix molecules and growth factors present in neurogenic niches) have the potential to migrate long distances, such as from the subventricular zone (SVZ) toward the olfactory bulb through the rostral migratory stream (34). However, evidence shows that the migration mode of SVZ progenitor cells can be controlled by environmental cues and it was further proposed that subcortical white matter tracts present structural and/or biochemical properties favorable to progenitor cell migration (25). Manipulation of the local environment of stem cell niches and therefore GSC niches could provide a gateway into controlling both the initial damage and future spread of glioblastomas.
However, despite the difficulties associated with pinpointing GSC niches, recently, there have been a collection of reports which indicate two particular areas where GSCs may be found. Bao et al was the first to show that GSCs develop a special relationship with the surrounding vasculature, as it was found that CD 133+ human glioblastoma cells consistently secrete high levels of vascular endothelial growth factor (VEGF) and that this might contribute to their tumor‐initiating capacity (6). Soon after, Calabrese et al demonstrated that GSC fractions were located next to capillaries in brain tumors and that endothelial cells interact selectively with these GSCs in culture and supply secreted factors that maintain these cells in self‐renewing and undifferentiated states (22). They also found that targeting a brain tumor with bevacizumab (a drug that blocks angiogenesis) reduces the number of cancer stem cells in treated tumors (22).
Furthermore, Heddleston et al, in examining the role of hypoxia in regulating tumor cell plasticity, found that a hypoxic environment promotes the self‐renewal capability of the stem and nonstem population (65). It was also found to promote a more stem‐like phenotype in the nonstem population, with increased neurosphere formation as well as up‐regulation of important stem cell factors, such as OCT4, NANOG and c‐MYC (65). A more recent study has also shown that in exposing GSCs to a hypoxic state (1% oxygen) for 72 h resulted in increased expression of multiple genes including lysyl oxidase (LOX), VEGF, hypoxia inducible gene 2 (HIG2) and Krüpple‐like factor 4 (Klf4) (9). Klf4 is particularly important as it plays an important role in maintaining embryonic stem cells and preventing their differentiation. Klf4 also contributes to the down‐regulation of p53 transcription. The study by Bar et al also goes on to show that by using digoxin to target the hypoxic response in GBM, via the inhibition of hypoxia inducible factor (HIF), growth of glioma cell lines in vitro and in vivo can be slowed, suggesting that GSC responses to hypoxia can be suppressed (9). Seidel et al also showed that hypoxia plays a key role in the regulation of the tumor stem cell phenotype through HIG2 alpha, indicating that tumor cells are also maintained within a hypoxic niche and could thus provide a GSC niche (136).
CELL MARKERS OF GSCs
Much of the research regarding cell markers has been directed toward CD133, as seen in Table 2. Early research showed that within glioblastomas, a subpopulation of cells existed. This subpopulation which represented a minority of the tumor was identified by expression of the cell surface marker CD133. It was confirmed as a brain tumor stem cell in 2003 and it was the first marker of GSCs (142). Following its discovery, CD133 (a pentaspan transmembrane glycoprotein cell surface molecule) has been widely used as a marker for enrichment of a GSC population (26). As such, CD133 continues to be used as the primary cell marker of GSCs. In reviewing Table 2, it is of note that there is still limited data on cell markers of GSCs, thus it is difficult to conclude that any one marker will provide greater benefit in aiding the location of GSCs. Furthermore, as Table 2 shows, in ascertaining the percentage of GSCs that express marker per tumor, all samples used have been cultured in some way which further adds difficulty when making comparisons between cell markers.
Table 2.
Cell markers of GSCs, percentage expression per sample size and percentage of GSCs that express marker per tumor. Abbreviations: GSC = glioblastoma stem cell; N/A = data not available
| Cell marker | Sample size | Percentage of expression per sample size | Percentage of GSCs that express marker per tumor | Methodology of preparing specimens | Study |
|---|---|---|---|---|---|
| Cell surface markers | |||||
| CD133 | 25 | N/A | 11.2–18.4 | Analyzed using flow cytometry | (113) |
| 4 | N/A | 19–29 | In vitro primary sphere formation assays performed on all uncultured cells prior to flow cytometric quantification | (143) | |
| 24 | 54 | 1.7–47.1 | Enzymatically dissociated into single cells and cultured; sorted by flow cytometric analysis | (146) | |
| 7 | N/A | 2.0–94.4 | Biopsies were dissociated into single cells and cultured; sorted by flow cytometric analysis. | (164) | |
| 16 | 75 | N/A | Immunohistochemical staining using mouse antihuman CD133/1‐biotin MAb, CD133/2‐biotin MAb, and CD133/1MAb antibody. Signals were counted in eight random fields of each single tumor specimen. | (159) | |
| 125 | 78.4 | N/A | Immunohistochemical staining using anti‐Nestin monoclonal antibody. The number of positive‐staining cells showing immunoreactivity on the cell membranes and cytoplasm in 10 representative microscopic fields was counted and the percentage of positive cells was calculated. | (182) | |
| 47 | 96 | N/A | Immunohistochemical staining using mouse monoclonal anti‐CD133 antibody | (181) | |
| Integrin α6 | 7 | 100 | 1.2–41.1 | Biopsy was used immediately after dissociation or after transient xenograft passage in immunocompromised mice. Cells were sorted by fluorescence‐activated cell sorting or magnetic bead separation | (94) |
| SSEA/CD15 | 24 | 96 | 1.4–84.2 | Enzymatically dissociated into single cells and cultured; sorted by flow cytometric analysis | (146) |
| 7 | N/A | 3.0–15.2 | Biopsies were dissociated into single cells and cultured; sorted by flow cytometric analysis | (164) | |
| L1CAM | 3 | 100 | 3.8–6.3 | Matched glioma cell populations were isolated and cultured and then subjected to fluorescence‐activated cell sorting | (7) |
| CXCR4 | 7 | 100 | 1.1–40.2 | Biopsies were dissociated into single cells and cultured, followed by flow cytometric analysis | (164) |
| A2B5 | 25 | N/A | 57.9–65.5 | Analyzed using flow cytometry | (113) |
| Intracellular markers | |||||
| Nestin | 125 | 82.4 | N/A | Immunohistochemical staining using anti‐Nestin monoclonal antibody. The number of positive‐staining cells showing immunoreactivity on the cell membranes and cytoplasm in 10 representative microscopic fields was counted and the percentage of positive cells was calculated. | (182) |
| 7 | 100 | 10–97.5 | Samples were cultured in Dulbecco's modified Eagle's medium; immunocytochemistry was used for quantification of the cells positive for a specific marker | (52) | |
| Sox‐2 | 7 | 100 | 31–85.5 | Samples were cultured in Dulbecco's modified Eagle's medium; immunocytochemistry was used for quantification of the cells positive for a specific marker | (52) |
| 11 | 90 | N/A | Immunohistochemistry was performed; the proportion of Sox2‐positive cells was determined by counting all cell nuclei as well as nuclei stained for Sox2 in three randomly selected high‐power fields in the tumor core of each sample | (134) | |
A problem, however, in using CD133 as the gold standard cell marker for glioblastomas, is that many normal cells express CD133 as well. This fact potentially limits the use of CD133 as a target, as the ability to discriminate GSCs from non‐GSCs is not absolute (27). This was confirmed by Tchoghandjian et al who isolated stem cells from glioblastomas expressing the surface marker A2B5 (157). This subpopulation was then divided into two groups using the CD133 antigen into A2B5+/CD133+ and A2B5+/CD133− stem cells, both of which were able to generate tumors when grafted in to nude mice. A2B5 is a cell surface ganglioside that marks neural precursor cells in the adult human brain (112). As Table 2 shows, A2B5 is also a useful GSC marker, especially it would seem in the absence of CD133.
Furthermore, the point of course in attaining a reliable GSC marker is so that it can be used as a target in clinical therapy. Worryingly, it has been found in at least one study that when GBM patients are classified into two groups (CD133‐high group where the CD133+ cell ratio is <3% and CD133‐low group where the CD133− cell ratio is >3%), the CD133‐low GBMs showed more aggressive morphologies as determined by magnetic resonance imaging and unique gene expression patterns that were related to a worse prognosis (82). Kim et al also found no relationship between the level of stem cell marker expression of CD133, SSEA or nestin and prognostic implications, and in fact found that patients with GBM who were CD133+ had a slightly longer survival rate than did the CD133− group (89). A study by Wang et al showed that using CD133− cells derived from six different patients was tumorgenic when implanted into rat brains and from these tumors CD133 positive cells could be obtained (169). Therefore, it is likely that a more primitive cancer stem cell phenotype precedes the CD133+ GSCs.
As with the cell marker CD133, as Table 2 indicates, SSEA‐1 does not enrich for a GSC population in every tumor either. The antigen SSEA‐1 is a fructose‐containing trisaccharide, which is highly expressed on embryonic stem cells in the developing brain and adult SVZ (24). SSEA‐1+ cells have increased expression of stem cell genes, such as Sox‐2 and Bmi1, and are capable of self‐renewal and multilineage differentiation (146). Furthermore, preliminary data have shown that in some tumors, both SSEA‐1−/CD133+ cells and SSEA‐1+/CD133− cells are much more tumorigenic as compared with SSEA‐1−/CD133− cells (146). Therefore, like A2B5, SSEA‐1 should prove to be a useful enrichment marker for GSCs especially for those that are CD133 negative.
One of the more recent cell markers to be analyzed is CXCR4 and the limited data collected so far on this particular cell marker are shown in Table 2. CXCR4 is a G‐protein coupled chemokine receptor which is known to be widely expressed on different cell types (43). Within glioblastomas, CXCR4 expression has been found to increase with increasing grade and colocalizes to regions within these tumors where its interaction may contribute to angiogenesis (128). Furthermore, cancer cells with high levels of CXCR4 are more likely to metastasize (4). It has only been recently reported that CXCR4 is overexpressed in primary glioblastoma progenitor cells and the stimulation of CXCR4 specific ligand, CXCL12, induces a significant proliferative response in the cancer stem cell but not in corresponding differentiated cells (38). Therefore, the role of CXCR4 as a cell marker for GSCs may be more significant than Table 2 suggests.
In the same light as CXCR4, there has been limited investigation on the cell marker L1CAM. This particular marker is a cell adhesion molecule derived from the immunoglobulin family (126). The protein cell marker has been found to interact with several extracellular matrix proteins as well as epidermal growth factor receptor, neuropilin‐1, and α5β1 and αvβ3 integrins 104, 133. Interestingly, Cheng et al (28) found that although a subpopulation of cancer cells inside tumors expressed L1CAM, the invasive fronts of primary GBM showed many more cells with high levels of L1CAM. Immunofluorescent staining further confirmed that many more L1CAM‐positive cells were localized in the invasive fronts than the center region of GBM (28).
Also to date, limited data have been collected on the integrin α6, as shown in Table 2. Integrins are heterodimeric proteins with alpha and beta subunits (35) which make up a large family of receptors that mediate the adhesive interactions of cells (107). Similarly to the L1CAM protein, integrin α6 can mostly be found on the invasive fronts of GBM. Specifically, it was found that 60% of integrin α6‐positive cells were located within 5 µm of a blood vessel as compared with only 10% of total tumor cells (94).
Finally, as indicated by Table 2, intracellular markers Sox‐2 and nestin show promise as reliable GSC markers, but their use is limited because they are intracellular, making the study of live cells difficult. To use either of these markers while cells are still alive and viable would require the use of the reporter system. Furthermore, the most recent evidence suggests that Sox‐2 may not be a reliable GSC marker because it can be found in high frequencies even in low‐grade gliomas and was not found to be linked to patient survival either (168). Evidence by Wan et al throws into doubt the usefulness of the findings that Sox‐2 expression can be found in many glioblastoma tumors as Table 2 highlights. Sox‐2 is a member of the Sry‐related High Mobility Group box family of transcription factors (61) and whose overexpression can induce pluripotency in both mice and human somatic cells 154, 155. More recently, Gangemi et al showed that silencing of the Sox2 in freshly derived GSC stopped proliferation and the resulting cells' tumorigenicity in immunodeficient mice (52). Interestingly, a study by Sun and Zhang showed that arsenic trioxide can induce the apoptosis of GSCs, at least partly through down‐regulation of Sox‐2 (152). Therefore, Sox‐2 usefulness as a GSC marker may no longer be as credible as once perceived, but despite this, its importance in glioblastoma formation is yet to be fully understood and therefore is not to be underestimated.
Nestin is also an intracellular marker, but shows more promise as a reliable marker of GSCs. Nestin is an intermediate filament protein involved in the organization of the cytoskeleton, but it has also been implicated in cell signaling, organogenesis and cell metabolism (45). Nestin was one of the first cell markers to be detected originally by the monoclonal antibody Rat.401 and by immunochemistry and double‐label fluorescence‐activated cell sorting (FACS), where it was shown to be expressed in rat CNS stem cells 42, 68, 96.
Table 2 incorporates cell markers which have been measured according to whether or not any of the GSCs within a number of tumors expresses the particular marker and also how many of those GSCs within a single tumor express the marker. Those cell markers included, however, are by no means exhaustive and other markers of GSCs (which have not been measured in the same way as those present in Table 2) have been found as potential markers for GSCs. For example, Kondo et al used the fluorescent dye Hoechst 33342 and flow cytometry to distinguish a side population (a subpopulation of cells that is distinct in biological characteristics compared with the main population) from glioma cell lines. Kondo et al note that this method of isolating the GSCs provides a simple and general strategy for identifying the cells (91). Similarly, Harris et al reported that GSCs are enriched in the side population cells and that these cells show enhanced self‐renewal and multipotentiality compared with nonside population cells from the same tumor (64). The side population was sorted from the nonside population by staining tumorsphere cells with Hoechst 33342 dye and sorting via FACS (64). However, more recently Broadley et al show that sphere formation in GBM cell lines and primary GBM cells enriches for a CSC‐like phenotype of increased self‐renewal gene expression in vitro and increased tumor initiation in vivo (16). However, the side population was absent from all sphere cultures. Direct isolation of the side population from the GBM lines did not enrich for stem‐like activity in vitro, and tumor‐initiating activity was lower in sorted side population compared with non‐\side population and parental cells (16).
Other GSC markers that have been used include musashi, an RNA binding protein which has been shown to be expressed in glioblastomas and is known in mammals to control neural stem cell homeostasis, differentiation and tumorigenesis, but expression was significantly lower than that of nestin when compared in the same study (149). In a study by Kang and Kang, GBM cancer cells were exposed to 1,3‐bis(2chloroethyl)‐1‐nitrosourea (BCNU) and it was found that a small population of cells survived and proliferated. Some of these subpopulations contained stem‐like cells, which expressed CD133, CD117, CD90, CD71 and CD45 cell‐surface markers and had the capacity for multipotency and when transplanted into mice induced tumors (86). Furthering their studies, Kang et al found that radio‐resistant subpopulations of GBM stem cells also expressed the cell surface marker CD117 as well as CD133, CD71 and CD45 (87).
SIGNALING PATHWAYS COMMONLY FOUND IN GSCs
Key signaling pathways as potential GSC targets are illustrated in Table 3 and indicate the receptors which can activate them and the genes they affect. Understanding the signaling pathways can give direction to pharmacological studies. The current chemotherapy drug is temozolomide (TMZ), an alkylating agent that is taken orally and readily penetrates the blood–brain barrier (114). It is often given at low doses concomitant with radiotherapy, followed by adjuvant standard doses. This aggressive treatment schedule results in an increase of the 2‐year survival rate from 10.4% with radiotherapy alone to 26.5% (150). Recent research has shed some light on why TMZ is not as effective as first thought. Shi et al has shown that microRNAs (small noncoding RNA molecules that regulate protein expression by cleaving or repressing the translation of target mRNAs), specifically mi‐RNA‐125b overexpression, might confer GSC resistance to TMZ as mi‐RNA‐125b is overexpressed in GSCs (141). Following the inhibition of mRNA‐125‐b in GSCs, the expression of the anti‐apoptotic protein Bcl‐2 was decreased, while the expression of the proapoptptic protein Bax and cytochrome c release from mitochondria was increased (141). Currently, there is no pharmacological cure for glioblastoma tumors and as surgical resection and radiotherapy are inadequate in being able to remove such a diffuse tumor, further developments in targeting GSC populations with chemotherapy treatments remain a priority.
Table 3.
Signaling cascades commonly found in GSCs and their role within the cell. Abbreviations: Akt = v‐akt murine thymoma viral oncogene; BAX = B‐cell lymphoma 2 associated X protein; BCL‐XL = B‐cell lymphoma extra large; BCL = B‐cell lymphoma; BCL‐2 = B‐cell lymphoma 2; BMP = bone morphogenetic proteins; Bmi‐1 = B lymphoma Mo‐MLV insertion region 1 homolog; BMPR = bone morphogenetic protein receptor; CDK4 = cyclin‐dependant kinase 4; c‐Myc = myelocytomatosis; CSL = citrate synthase‐like; EGFR = epidermal growth factor receptor; Fra = fos related antigen; GSC = glioblastoma stem cell; Hes = hairy and enhancer of split 1; Hey = Hairy/enhancer‐of‐split related with YRPW; IGFBP‐2 = insulin‐like growth factor‐binding protein‐2; IGF = insulin‐like growth factor; MCL‐1 = myeloid cell leukemia‐1; PDGF‐β = platelet derived growth factor‐beta; NMDA = N‐Methyl‐D‐aspartic acid; NPC = neural precursor cell; NSC = neural stem cell; Rtk = receptor tyrosine kinase; IRS1 = insulin receptor substrate 1; WNT = Wingless‐type MMTV integration site family
| Signaling pathway | Receptor(s) | Target gene(s) | Overall role of signaling pathway | Reference |
|---|---|---|---|---|
| Notch signaling | 300 kDa single‐pass short‐range transmembrane receptor | Converted CSL becomes activator of transcription for Hes and Hey families, cyclin D and c‐myc | Essential for the maintenance and fate determination in somatic stem and progenitor cells by promoting self‐renewal and repressing differentiation | 17, 78, 93, 123, 130, 139, 156 |
| Stat3 | Ciliary neurotrophic factor family cytokine receptor | BCL‐2 family including BCL‐2, BCL‐X, BAX and MLC‐1 | Controls major cellular responses, including cell proliferation and survival. Also, the JAK‐STAT3 signaling pathway plays an essential role in the maintenance of NPCs by fibroblast growth factor 2. | 21, 31, 54, 125 |
| Rtk‐Akt | Receptor tyrosine kinases | EGFR | Activation of EGFR promotes the growth of both astrocyte precursors and neural stem cells. | 3, 95, 144 |
| Transforming growth factor beta | Serine/threonine kinase complex membrane receptor | PDGF‐β | Oncogenic stimulus in glioblastoma growth through induction of angiogenesis, immune evasion and invasion | 20, 118, 173 |
| Hedgehog‐gli signaling | Surface membrane receptor patched | Gli‐1, Cyclin D, Cyclin E, IRS1 | Regulates the behavior of stem cells particularly clonogenicity, survival, tumorigenicity and proliferation | 29, 37, 72, 88, 106, 132, 137, 175 |
| BMP | BMPR1A; BMPR1B; BMPR2 | Activin response element | Mediate a wide range of biological responses in NSCs including proliferation, differentiation and apoptosis. Instructive role in stem cell niche which favors the acquisition of the astroglial fate. BMP4 may inhibit GSCs by decreasing symmetric cell cycles that generate two GSCs on division, trigger differentiation or block proliferation | 5, 98, 115, 120 |
| Rac1‐Pak | NMDA | Cyclin D1 | Maintenance of stemness and malignant properties of glioma stem‐like cells | 83, 158, 178 |
| Wnt‐β‐catenin | Low‐density lipoprotein receptor related protein 5, 6, 10; Frizzle receptor | Wnt 1, 5a, 10b, 13; Fra‐1; c‐Myc; Cyclin D1 | Induces neuronal differentiation of neural precursor cells and promotes proliferation. It is also an important regulator of stem cell self‐renewal. | 63, 71, 79, 84, 90 |
| IGFBP‐2 | IGF‐receptor | Cyclin D1, CDK4, Cyclin E, Bmi‐1, | GSC promoting factor and a broad regulator of GSC resistance to lethal stress induced by genotoxic agents or small molecule inhibitors. Stimulates the migration of stem cells | 10, 73 |
In the RTK‐Akt signaling pathway, Soeda et al showed tyrosine kinase inhibitors (AG1478 and gefitinib) which are relatively selective for epidermal growth factor receptor (EGFR) kinase, eradicated the self‐renewal capacity of GSCs and the proliferation and differentiation of CD133+ cells (145). Eyler et al used a phosphatidylinositol ether lipid analog as an inhibitor of Akt and found that it decreased GSC migration and invasion and also increased the survival of immuno‐compromised mice bearing human glioma xenografts by 22 days (39). Thus, there is increasing evidence that by inhibiting a signaling pathway involved in GBM development, such as the Akt pathway, glioblastoma development can at least be hindered. Gallia et al also used an inhibitor of the Akt pathway, the small polymer molecule A‐443654, finding that it inhibited the growth of GSC lines and extended the survival of intracranial glioma tumor bearing mice by an average of 11 days when they received the tumor and polymer on the same day and an average of 6 days when they received the polymer 4 days after receiving the tumor (50).
More recently, Griffero et al evaluated the in vitro sensitivity of human glioma tumor initiating cells to (EGFR) kinase inhibitors and found that they are responsive to anti‐EGFR drugs erlotinib and gefitinib, but phosphatase and tensin homolog expression and Akt inhibition seem to be necessary for such effect (58). An atypical approach of using eckol, a phlorotannin component of some brown algae, has been used to suppress the stemness and the malignance of glioma stem‐like cell populations (75). Hyun et al found that eckol inhibited the phosphoinositide 3‐kinase‐Akt and Ras‐RAF‐1Erk signaling pathways, which also made glioma stem‐like cells more sensitive to anticancer treatment such as ionizing radiation and temozolomide (75).
Research has shown that CXCR4 can signal through the Akt and RAS pathways 117, 163, 174. Redjal et al used the CXCR4 inhibitor AMD3100 in combination with BCNU on GBM cell lines and found it resulted in synergistic antitumor efficacy in vitro and also resulted in tumor regression in vivo showing increased apoptosis and decreased proliferation of mice with established orthograft tumors (127). Furthermore, CXCR4 is the known receptor for stromal cell derived factor‐1 and has also been shown to mediate neural stem cell migration toward injury or tumors in a murine brain (135). Schulte et al confirmed CXCR4 overexpression in GSC lines by real‐time polymerase chain reaction analysis, and then used a CXCR4 receptor blockade, AMD3100, for in vivo treatment of glioblastoma growth in the brains of nude mice and found that it caused inhibition of expansive tumor growth by 58.3% (135).
It has been suggested that Notch may play a particularly important role in cancer stem cells and it has been shown to be critically implicated in stem cell fate determination and cancer in GSCs 121, 122, 142. Notch proteins are transmembrane receptors that mediate short‐range cellular communication through interaction with ligands (Jagged‐1, ‐2 and Delta‐like‐1, ‐2, ‐3, ‐4). The activation of Notch requires sequential proteolytic cleavage by the gamma secretase complex to release its intracellular domain from membrane to nucleus. Notch signaling promotes the proliferation of normal neural stem cells and is required for the maintenance of neural progenitors both in vitro and in vivo (108). A study by Wang et al showed that inhibition of the Notch pathway with gamma‐secretase inhibitors (GSIs) renders the GSCs more sensitive to radiation at clinically relevant doses (170). Indeed, inhibition of the Notch signaling pathway with GSIs has been the most extensively used approach to target Notch (93). However, it has been noted that GSIs affect all Notch receptors, causing unwanted side effects, the most common being metaplasia of goblet cells in the small intestine (147).
A study by Li et al of U87 human glioblastoma cells and glioma stem cells 0308 interestingly found that microRNA‐34a (miRNA‐34a), a transcriptional target of p53, down‐regulated notch‐1 and notch‐2 expression in glioma cells (97). The transient transfection of miRNA‐34a in glioma stem cell lines strongly inhibited cell proliferation, cell cycle progression, cell survival and cell invasion, showing potential as a therapeutic agent for GSCs (97). It should be noted that miRNA‐34a has multiple predicted targets among which are many oncogenes, and therefore the proposed inhibition of malignant activity may not have been strictly caused by the down‐regulation of notch‐1 and notch‐2. More recently, Campos et al used all‐trans‐retinoic acid (ATRA), a known potent modulator of cellular differentiation and proliferation, as a treatment on stem‐like glioma cells and found that expression of Notch‐1 was reduced on mRNA and protein levels after differentiation (23). Following implantation into mouse brains with undifferentiated stem‐like glioma cells, there was higher Notch‐1 expression as compared with samples which were inoculated with differentiated stem‐like glioma cells (23). On post‐mortem examination of the brains of the mice, it was reported that those which had been implanted with undifferentiated cells developed tumors which were more than seven times larger and showing a highly invasive growth pattern, compared with those mice which were implanted with differentiated cells, despite being implanted with 10 times fewer cells (23).
Further work on the use of ATRA with GSC has been carried out by Ying et al. The group used ATRA treatment to induce changes in morphology, induce growth arrest at G1/G0 to S transition, decrease cyclin D1 expression and increase p27 expression in GSCs which were grown as nonadherent neurospheres in growth factor supplemented serum‐free medium (177). As with Campos et al, it was also found that several Notch pathway components were significantly down‐regulated by ATRA (177). Furthermore, the importance of the Notch signaling pathway cannot be underestimated, as indicated by a study by Wang et al which showed that either notch1 or notch2 protected GSCs against radiation (170), indicating that currently, attaining the full benefits of radiotherapy for GBM treatment is only likely to occur once significant advances have been made pharmacologically.
Clement et al tested for the possible role of hedgehog‐gli signaling in cancer stem cells, using glioma stem cell cultures, which self‐renew and mimic the original tumor after transplantation into the brains of immunocompromised mice (29). Treatment with cyclopamine decreased their proliferation and increased apoptosis in a concentration‐dependent manner (29). It was also argued that due to the lack of any obvious secondary effects in cyclopamine‐treated adult mice, it was suggested that such treatment may spare normal quiescent stem cells in their niches, likely allowing regeneration of any damage to normal adult tissues after cessation of treatment (29). Similarly, Bar et al found that cyclopamine caused a 40%–60% reduction in growth of adherent glioma lines which largely expressed hedgehog‐gli signaling (8). When the GBM‐derived neurospheres were treated with cyclopamine and then dissociated and seeded in media lacking the inhibitor, no new neurospheres formed, suggesting that clonogenic cancer stem cells had been depleted (8). Consistent with this hypothesis, the stem‐like fraction in gliomas marked by both aldehyde dehydrogenase activity and Hoechst dye excretion was significantly reduced or eliminated by cyclopamine (8). More recently, Ulasov et al found that the therapeutic effect of TMZ was enhanced by Notch and Hedgehog pathway pharmacological antagonism with GSi‐1 and cyclopamine (162). Importantly, simultaneous treatment involving TMZ with both GSi‐1 and cyclopamine led to a significant increase in CD133+ glioma cytotoxicity than treatment with any of these agents alone (162).
Other notable pharmacological developments include the use of GSC lines derived from right temporal GBMs, in which Sherry et al found that inhibition of STAT3 in these cells with either small molecular inhibitors or RNAi resulted in inhibition of growth and neurosphere formation although they did not undergo apoptosis (140). Wei et al also showed that STAT3 expression in GSCs can be inhibited with a STAT3 short hairpin RNA or the small molecule inhibitor WP1066 (172). More recently, Ma et al investigated the role of celecoxib, a selective COX‐2 inhibitor, in enhancing the therapeutic effects of radiation on CD133+ glioblastoma cells (103). They found that high levels of COX‐2 protein were detected in CD133+ but not in the CD133− glioblastoma cells and further demonstrated that 30 µM celecoxib was able to enhance the effect of radiation in inhibiting colony formation and increasing apoptosis in CD133+ glioblastoma cells (103).
Despite recent advances in novel pharmacological developments aimed at the various signaling pathways which exist in GSCs, one of the major difficulties in developing any pharmacological treatment for GBM is that cell signaling cascades differ depending on the GBM and it is clear from the studies that not all signaling pathways are active or required to produce GBM. Indeed, in certain signaling cascades, there is still debate regarding their existence in grade IV GBM tumors. The hedgehog‐gli signaling pathway, for example, has been shown to be active in more than 50% of GBM (8). When Bar et al used Gli‐1 as a marker for the hedgehog pathway, it was shown to be highly expressed in five out of nine primary GBM and in four of seven GBM cell lines. However, other research demonstrated in 13 adult gliomas that hedgehog‐gli pathway was operational and active within grades II and III gliomas, but not grade IV GBM (137). It is likely that hedgehog signaling is operational in only some brain tumors, as demonstrated by Xu et al who showed there are both hedgehog‐gli signaling‐dependent and ‐independent brain tumors characterized by a cancer stem cell zone of Gli‐1 positive cells (175). This indicates that within any given GBM tumor, there are most likely a number of different types of GSCs operating, each with its own unique and multivariant signaling cascades.
GENE MUTATIONS IN GSCs
The genetic research of GSCs, which can be seen in Table 4, clearly shows that there are a number of mutations which are involved in the development of brain tumors. So far there is no evidence that any one individual gene can induce glioblastoma tumor formation. Studies have shown that the activation of certain genes may also require the specific activation of another gene, combinational gene activation, in order to induce glioblastoma formation 69, 105. This evidence highlights the importance of combined gene mutations which have to occur synergistically in order to achieve a desired effect. It is possible that many genes and signaling pathways require similar co‐operators in order for their effect to take place and therefore a more holistic observation of cellular activity may be required in order to better understand genetic and signaling interaction.
Table 4.
Gene mutations, mechanism of action and involvement in glioblastoma development. Abbreviations: Akt = v‐akt murine thymoma viral oncogenes; Arf = ADP‐ribosylation factor; Bmi‐1 = B lymphoma Mo‐MLV insertion region 1 homolog; ECM = extra cellular matrix; EGFR = epidermal growth factor receptor; GBM = glioblastoma multiforme; mTOR = mammalian target of rapamycin; NPC = neural precursor cell; NSC = neural stem cell; OPC = oligodendrocyte precursor cell; PDGF‐β = platelet derived growth factor‐beta; PTEN = phosphatase and tensin homolog; TGF‐β = transforming growth factor beta; Ras = RAt Sarcoma
| Gene | Action in neural stem cell | Involvement in glioblastoma development | Reference |
|---|---|---|---|
| p53 | Negative regulation of NSC proliferation. Mediator of cellular responses to DNA damage, stimulating and increasing the levels of Cdk inhibitor Cdkn1A | Deficiency induces high levels of platelet derived growth factor receptor expression, deregulation of the Rb‐mediated cell cycle regulatory pathway, high levels of Cdk4 expression and cyclin D1 over expression. | 99, 116, 171 |
| Ras and Akt | Ras gene codes for proteins whose primary role is to assemble transient signaling complexes at the membrane that activate signal transduction pathways coordinating transcription, cell shape and migration, endocytosis, cell survival and cell cycle progression. Akt is a downstream target for receptor tyrosine kinases that are regulated by stimuli such as growth factors and insulin, and therefore plays a major role in metabolism, cell growth and cell survival. | Ras, if mutated prevents the conversion of Ras‐GTP back to Ras‐GDP, stimulating continuous cell growth due to the consistent stimulation of a signal transduction pathway for cell proliferation. Akt activity in tumors correlated with phosphorylation of S6RP and 4E‐BP1 downstream of mTOR. Transfer of genes encoding for Ras and Akt together to neural progenitors induces high grade gliomas. | 15, 53, 80, 105, 116 |
| p16INK4a and p19ARF | p16INK4a inhibits cyclin D‐dependent kinases, regulates the ability of the retinoblastoma protein to control G1 exit. p19ARF is a negative regulator of Mdm2 function, can activate p53 in response to particular oncogenic signals, thereby inducing cell cycle arrest or apoptosis, depending on the biological context. | Loss of both p16INK4a and p19ARF in combination with epidermal growth factor receptor pathway activation provokes a common high‐grade glioma phenotype regardless of special cell or origin. | 85, 95 |
| Bmi‐1 transcription factor | A core component of the Polycomb group proteins which are epigenetic chromatin modifiers involved in heritable gene repression and maintenance of stem cell self‐renewal and progenitor proliferation and this has been primarily attributed to its ability to repress the Ink4a‐Arf locus | Bmi‐1 inactivation causes a change in the expression of genes involved in the TGF‐β signal transduction pathway, ECM remodeling and cell cycle control. Bmi‐1 can cause tumor development in an Ink4a/Arf‐independent manner. | 18, 19, 109, 151, |
| PDGF‐β | Exerts its stimulatory effects on cell growth and motility by binding to two related protein tyrosine kinase receptors | Deregulated PDGF‐β expression has been shown to cause glioma‐like tumors by retroviral insertion into newborn mice. | 66, 110 |
| PTEN phosphatase | Negative control of NSC proliferation. | Frequently mutated in aggressive high‐grade gliomas. | 59, 131, 161 |
| EGFR membrane receptor | EGF‐dependent NSC proliferation/maintenance | Transduction of Ink/Arf‐/‐ in NSC or astrocytes with constitutively active EGFR induces high‐grade glioma in mice. Enhance cell proliferation by promoting P13K/Akt signaling; inhibit cell cycle regulators including 27 KIP1 and increase expression of anti‐apoptotic proteins such as Bcl‐XL and unregulates vascular endothelial growth factor, interleukin‐8 and matrix metalloproteinase 13 expression | 51, 95, 148, 160 |
| High‐mobility group A1 | Developmental regulator of stem cell self renewal and Ink4a/Arf expression | Important role in the proliferation, invasion and differentiation of GBM | 40, 111 |
Indeed there is growing evidence that there is no single universal GSC type. Phillips et al reported classification of three main glioblastoma subtypes defined by gene expression signature. Three subtypes included one which expressed neurodevelopmental genes which was associated with a better prognosis, and two other subtypes characterized by their resemblance to either highly proliferative cell lines or tissues of mesenchymal origin, both being associated with a rapid rate of cell division (119). Similarly, Guenther et al established glioblastoma stem‐like cell lines from nine different glioblastomas which had been maintained for up to 3 years and although established under identical conditions two distinct subtypes arose. Using gene expression profiling and considering neurobiological criteria, four cell lines (cluster 1) shared similar gene expression patterns associated with neural development and displayed a full stem‐like phenotype, whereas five cell lines (cluster 2) shared a different gene expression pattern and displayed only restricted stem‐like phenotype (62). Interestingly, most of the cluster 2 cell lines showed no detectable CD133+ cells, although these cell lines did contain cells expressing the Sox2 and nestin markers. More recently, a study by Lottaz et al compared the gene expression profiles of 17 GSC lines and distinguished two subgroups; type I GSC lines display “proneural” signature genes and resemble fetal neural stem cells lines, whereas type II lines show “mesenchymal” transcriptional profiles similar to adult neural stem cells (100). Clearly then, GSCs are not represented by one single phenotype or genetic signature but rather a mosaic, which may be typical of the environment in which they develop.
Furthermore, it has been shown that specific combinations of gene deletions lead to different tumors. Bachoo et al found that combined loss of p16INK4a and p19ARF, but not of p53, p16INK4a or p19ARF, enables astrocyte dedifferentiation in response to EGFR activation (3). Similarly, Jacques et al deleted Rb/p53, Rb/p53/PTEN or PTEN/p53 in adult subventricular stems and in ectopically neurografted stem cells (77). It was found that recombination of PTEN/p53 gave rise to gliomas whereas deletion of Rb/p53 or Rb/p53/PTEN generated primitive neuroectodermal tumors. It would seem, therefore, that stem cells which have activated oncogenes and inactivated tumor suppressor genes give rise to tumors of specific phenotypes depending on the combination of mutations.
RELATIONSHIP BETWEEN IMMUNOSUPPRESSION AND ONCOGENESIS RELATIVE TO GLIOBLASTOMAS
A relationship must exist between immunosuppression and oncogenesis, and for GSCs it might be that they sustain neoplastic growth through immunoevasion and/or some kind of immunomodulatory function. This correlates with the fact that many immunocompromised patients are at a higher risk of developing cancer (60). GSCs can disrupt tumour immunosurveillance and result in both ineffective adaptive and innate immune responses (55). There are a plethora of immunosuppressive mechanisms associated with malignant gliomas, where patients have demonstrated significant impaired immune function (179). These mechanisms include inhibition of antigen‐presenting cells, direct inhibition of B cells and T cells, and the immunosuppressive effects have been shown to be mediated by chemokines such as transforming growth factor beta, prostaglandin E2 and certain gangliosides (36). A recent report by Rodrigues et al (129) also found that GSCs suppress activation of microglial cells, which in turn suppress T‐cell activity by two secreting immunosuppressive factors, interleukin‐10 and Fas‐ligand. Thus, GSCs contribute to immunosuppression, and given the poor prognoses associated with high‐grade gliomas, there is a therapeutic need in order to improve survival rates. Owing to the fact that the immune system must play a key role in the development of GBM, it would be highly desirable to develop an immunotherapeutic treatment strategy. More importantly, it may be necessary to take a multifaceted approach to treatment whereby two or more immunotherapeutic strategies are adopted, and this approach will be more beneficial to GSCs due to the inherent heterogeneous population of cells with different proliferative potential.
CONCLUSION
The continual growth in research and new findings on glioblastomas and the causative subpopulation of stem cells highlight both its importance and the challenge it presents. This type of cancer introduces a number of unique problems, including locating the origin of stem cells which become tumorigenic not least because of a lack of a global cell marker for this type of tumor. The variability in signaling pathways and genetic mutations would indicate that at least a few, if not many, of these are active within a single GSC population, indicating that any pharmacological treatment will have to be broad enough to cover all active tumorigenic signaling pathways and mutations in order to be successful. Thus, the evidence seems to propose that glioblastomas are derived from multiple stem cell lineages, whereby there are subpopulations within the subpopulation of GSCs. This would help to explain the variety of combinations and permutations of cell markers, signaling cascades, and genetic mutations found within single tumor samples and across all samples from different patients. Furthermore, the nutrient‐rich brain matter provides an optimum environment that allows for the extraordinary proliferative capacity of GSCs to develop, meaning an early diagnostic marker is crucial. Future studies may want to focus on identifying any differences between cancer stem cells which make up the subpopulation of a glioblastoma and consider if it is possible to provide treatments which are multidimensional in their approach.
There are no conflicts of interest.
REFERENCES
- 1. Aboody KS, Brown A, Rainov NG, Bower KA, Liu SX, Yang W et al (2000) Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc Natl Acad Sci U S A 97:12846–12851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Alvarez‐Buylla A, Garcia‐Verdugo JM (2002) Neurogenesis in adult subventricular zone. J Neurosci 22:629–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Bachoo RM, Maher EA, Ligon KL, Sharpless NE, Chan SS, You MJ et al (2002) Epidermal growth factor receptor and Ink4a/Arf: convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis. Cancer Cells 1:269–277. [DOI] [PubMed] [Google Scholar]
- 4. Bajetto A, Barbieri F, Dorcaratto A, Barbero S, Daga A, Porcile C et al (2006) Expression of CXC chemokine receptors 1–5 and their ligands in human glioma tissues: role of CXCR4 and SDF1 in glioma cell proliferation and migration. Neurochem Int 49:423–432. [DOI] [PubMed] [Google Scholar]
- 5. Baker JC, Harland RM (1997) From receptor to nucleus: the Smad pathway. Curr Opin Genet Dev 7:467–473. [DOI] [PubMed] [Google Scholar]
- 6. Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB et al (2006) Stem cell‐like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res 66:7843–7848. [DOI] [PubMed] [Google Scholar]
- 7. Bao S, Wu Q, Li Z, Sathornsumetee S, Wang H, McLendon RE et al (2008) Targeting cancer stem cells through L1CAM suppresses glioma growth. Cancer Res 68:6043–6048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W et al (2007) Cyclopamine‐mediated hedgehog pathway inhibition depletes stem‐like cancer cells in glioblastoma. Stem Cells 25:2524–2533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bar EE, Lin A, Mahairaki V, Matsui W, Eberhart CG (2010) Hypoxia increases the expression of stem‐cell markers and promotes clonogenicity in glioblastoma neurospheres. Am J Pathol 177:1491–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bartling B, Koch A, Simm A, Scheubel R, Silber R‐E, Santos AN (2010) Insulin‐like growth factor binding proteins‐2 and‐4 enhance the migration of human CD34(−)/CD133(+) hematopoietic stem and progenitor cells. Int J Mol Med 25:89–96. [PubMed] [Google Scholar]
- 11. Bauer S, Hay M, Amilhon B, Jean A, Moyse E (2005) In vivo neurogenesis in the dorsal vagal complex of the adult rat brainstem. Neuroscience 130:75–90. [DOI] [PubMed] [Google Scholar]
- 12. Bedard A, Levesque M, Bernier PJ, Parent A (2002) The rostral migratory stream in adult squirrel monkeys: contribution of new neurons to the olfactory tubercle and involvement of the antiapoptotic protein Bcl‐2. Eur J Neurosci 16:1917–1924. [DOI] [PubMed] [Google Scholar]
- 13. Bernier PJ, Bedard A, Vinet J, Levesque M, Parent A (2002) Newly generated neurons in the amygdala and adjoining cortex of adult primates. Proc Natl Acad Sci U S A 99:11464–11469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Bernstein JJ, Woodard CA (1995) Glioblastoma cells do not intravasate into blood‐vessels. Neurosurgery 36:124–132. [DOI] [PubMed] [Google Scholar]
- 15. Bleau A‐M, Hambardzumyan D, Ozawa T, Fomchenko EI, Huse JT, Brennan CW, Holland EC (2009) PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem‐like cells. Stem Cells 4:226–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Broadley KWR, Hunn MK, Farrand KJ, Price KM, Grasso C, Miller RJ et al (2011) Side population is not necessary or sufficient for a cancer stem cell phenotype in glioblastoma multiforme. Stem Cells 29:452–461. [DOI] [PubMed] [Google Scholar]
- 17. Brou C, Logeat F, Gupta N, Bessia C, LeBail O, Doedens JR et al (2000) A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin‐metalloprotease TACE. Mol Cell 5:207–216. [DOI] [PubMed] [Google Scholar]
- 18. Bruggeman SWM, Valk‐Lingbeek ME, van der Stoop PPM, Jacobs JJL, Kieboom K, Tanger E et al (2005) Ink4a and Arf differentially affect cell proliferation and neural stem cell self‐renewal in Bmi1‐deficient mice. Genes Dev 19:1438–1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bruggeman SWM, Hulsman D, Tanger E, Buckle T, Blom M, Zevenhoven J et al (2007) Bmi1 controls tumor development in an ink4a/Arf‐independent manner in a mouse model for glioma. Cancer Cells 12:328–341. [DOI] [PubMed] [Google Scholar]
- 20. Bruna A, Darken RS, Rojo F, Ocana A, Penuelas S, Arias A et al (2007) High TGF beta‐Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF‐B gene. Cancer Cells 11:147–160. [DOI] [PubMed] [Google Scholar]
- 21. Buettner R, Mora LB, Jove R (2002) Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention. Clin Cancer Res 8:945–954. [PubMed] [Google Scholar]
- 22. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B et al (2007) A perivascular niche for brain tumor stem cells. Cancer Cells 11:69–82. [DOI] [PubMed] [Google Scholar]
- 23. Campos B, Wan F, Farhadi M, Ernst A, Zeppernick F, Tagscherer KE et al (2010) Differentiation therapy exerts antitumor effects on stem‐like glioma cells. Clin Cancer Res 16:2715–2728. [DOI] [PubMed] [Google Scholar]
- 24. Capela A, Temple S (2002) LeX/ssea‐1 is expressed by adult mouse CNS stem cells, identifying them as nonependymal. Neuron 35:865–875. [DOI] [PubMed] [Google Scholar]
- 25. Cayre M, Bancila M, Virard I, Borges A, Durbec P (2006) Migrating and myelinating potential of subventricular zone neural progenitor cells in white matter tracts of the adult rodent brain. Mol Cell Neurosci 31:748–758. [DOI] [PubMed] [Google Scholar]
- 26. Chen Y, Ai Y, Slevin JR, Maley BE, Gash DM (2005) Progenitor proliferation in the adult hippocampus and substantia nigra induced by glial cell line‐derived neurotrophic factor. Exp Neurol 196:87–95. [DOI] [PubMed] [Google Scholar]
- 27. Cheng L, Bao S, Rich JN (2010) Potential therapeutic implications of cancer stem cells in glioblastoma. Biochem Pharmacol 80:654–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cheng L, Wu Q, Guryanova OA, Huang Z, Huang Q, Rich JN, Bao S (2011) Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun 406:643–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Altaba ARI (2007) HEDGEHOG‐GLI1 signaling regulates human glioma growth, cancer stem cell self‐renewal, and tumorigenicity. Curr Biol 17:165–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Cooper O, Isacson O (2004) Intrastriatal transforming growth factor alpha delivery to a model of Parkinson's disease induces proliferation and migration of endogenous adult neural progenitor cells without differentiation into dopaminergic neurons. J Neurosci 24:8924–8931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Darnell JE, Kerr IM, Stark GR (1994) Jak‐Stat pathways and transcriptional activation in response to IFNS and other extracellular signalling proteins. Science 264:1415–1421. [DOI] [PubMed] [Google Scholar]
- 32. Das P, Puri T, Jha P, Pathak P, Joshi N, Suri V et al (2011) A clinicopathological and molecular analysis of glioblastoma multiforme with long‐term survival. J Clin Neurosci 18:66–70. [DOI] [PubMed] [Google Scholar]
- 33. Dayer AG, Cleaver KM, Abouantoun T, Cameron HA (2005) New GABAergic interneurons in the adult neocortex and striatum are generated from different precursors. J Cell Biol 168:415–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Delcroix GJR, Jacquart M, Lemaire L, Sindji L, Franconi F, Le Jeune J‐J, Montero‐Menei CN (2009) Mesenchymal and neural stem cells labeled with HEDP‐coated SPIO nanoparticles: in vitro characterization and migration potential in rat brain. Brain Res 1255:18–31. [DOI] [PubMed] [Google Scholar]
- 35. Demetriou MC, Pennington ME, Nagle RB, Cress AE (2004) Extracellular alpha 6 integrin cleavage by urokinase‐type plasminogen activator in human prostate cancer. Exp Cell Res 294:550–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Ehtesham M, Black KL, Yu JS (2004) Recent progress in immunotherapy for malignant glioma: treatment strategies and results from clinical trials. Cancer Control 11:192–207. [DOI] [PubMed] [Google Scholar]
- 37. Ehtesham M, Sarangi A, Valadez JG, Chanthaphaychith S, Becher MW, Abel TW et al (2007) Ligand‐dependent activation of the hedgehog pathway in glioma progenitor cells. Oncogene 26:5752–5761. [DOI] [PubMed] [Google Scholar]
- 38. Ehtesham M, Mapara KY, Stevenson CB, Thompson RC (2009) CXCR4 mediates the proliferation of glioblastoma progenitor cells. Cancer Lett 274:305–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Eyler CE, Foo W‐C, Lafiura KM, McLendon RE, Hjelmeland AB, Rich JN (2008) Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells 26:3027–3036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Fan H, Guo H, Zhang IY, Liu B, Luan L, Xu S et al (2011) The different HMGA1 expression of total population of glioblastoma cell line U251 and glioma stem cells isolated from U251. Brain Res 1384:9–14. [DOI] [PubMed] [Google Scholar]
- 41. Fowler CD, Liu Y, Ouimet C, Wang Z (2002) The effects of social environment on adult neurogenesis in the female prairie vole. J Neurobiol 51:115–128. [DOI] [PubMed] [Google Scholar]
- 42. Frederiksen K, McKay RDG (1988) Proliferation and differentiation of rat neuroepotheilial precursor cells invivo . J Neurosci 8:1144–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Fricker SP, Anastassov V, Cox J, Darkes MC, Grujic O, Idzan SR et al (2006) Characterization of the molecular pharmacology of AMD3100: a specific antagonist of the G‐protein coupled chemokine receptor, CXCR4. Biochem Pharmacol 72:588–596. [DOI] [PubMed] [Google Scholar]
- 44. Frielingsdorf H, Schwarz K, Brundin P, Mohapel P (2004) No evidence for new dopaminergic neurons in the adult mammalian substantia nigra. Proc Natl Acad Sci U S A 101:10177–10182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Fuchs E, Weber K (1994) Intermediate filaments—structure, dynamics, function, and disease. Annu Rev Biochem 63:345–382. [DOI] [PubMed] [Google Scholar]
- 46. Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A et al (2007) Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21:2683–2710. [DOI] [PubMed] [Google Scholar]
- 47. Gage FH (1998) Discussion point—stem cells of the central nervous system. Curr Opin Neurobiol 8:671–676. [DOI] [PubMed] [Google Scholar]
- 48. Gage FH (2002) Neurogenesis in the adult brain. J Neurosci 22:612–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S et al (2004) Isolation and characterization of tumorigenic, stem‐like neural precursors from human glioblastoma. Cancer Res 64:7011–7021. [DOI] [PubMed] [Google Scholar]
- 50. Gallia GL, Tyler BM, Hann CL, Siu IM, Giranda VL, Vescovi AL et al (2009) Inhibition of Akt inhibits growth of glioblastoma and glioblastoma stem‐like cells. Mol Cancer Ther 8:386–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Gan HK, Kaye AH, Luwor RB (2009) The EGFRvIII variant in glioblastoma multiforme. J Clin Neurosci 16:748–754. [DOI] [PubMed] [Google Scholar]
- 52. Gangemi RMR, Griffero F, Marubbi D, Perera M, Capra MC, Malatesta P et al (2009) SOX2 silencing in glioblastoma tumor‐initiating cells causes stop of proliferation and loss of tumorigenicity. Stem Cells 27:40–48. [DOI] [PubMed] [Google Scholar]
- 53. Germano I, Swiss V, Casaccia P (2010) Primary brain tumors, neural stem cell, and brain tumor cancer cells: where is the link? Neuropharmacology 58:903–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Ghosh MK, Sharma P, Harbor PC, O Rahaman S, Haque SJ (2005) PI3K‐AKT pathway negatively controls EGFR‐dependent DNA‐binding activity of Stat3 in glioblastoma multiforme cells. Oncogene 24:7290–7300. [DOI] [PubMed] [Google Scholar]
- 55. Gilbert CA, Ross AH (2009) Cancer stem cells: cell culture, markers, and targets for new therapies. J Cell Biochem 108:1031–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Goetz CG (2007) Textbook of Clinical Neurology. WB Saunders: Philadelphia. [Google Scholar]
- 57. Gould E (2007) Opinion—how widespread is adult neurogenesis in mammals? Nat Rev Neurosci 8:481–488. [DOI] [PubMed] [Google Scholar]
- 58. Griffero F, Daga A, Marubbi D, Capra MC, Melotti A, Pattarozzi A et al (2009) Different response of human glioma tumor‐initiating cells to epidermal growth factor receptor kinase inhibitors. J Biol Chem 284:7138–7148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Groszer M, Erickson R, Scripture‐Adams DD, Lesche R, Trumpp A, Zack JA et al (2001) Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo. Science 294:2186–2189. [DOI] [PubMed] [Google Scholar]
- 60. Grulich AE, van Leeuwen MT, Falster M, Vajdic CM (2007) Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta‐analysis. Lancet 370:59–67. [DOI] [PubMed] [Google Scholar]
- 61. Gubbay J, Collignon J, Koopman P, Capel B, Economou A, Munsterberg A et al (1990) A gene‐mapping to the sex‐determined region of the mouse Y‐chromosome is a member of a novel family of embryonically expressed genes. Nature 346:245–250. [DOI] [PubMed] [Google Scholar]
- 62. Guenther HS, Schmidt NO, Phillips HS, Kemming D, Kharbanda S, Soriano R et al (2008) Glioblastoma‐derived stem cell‐enriched cultures form distinct subgroups according to molecular and phenotypic criteria. Oncogene 27:2897–2909. [DOI] [PubMed] [Google Scholar]
- 63. Han L, Yang Y, Yue X, Huang K, Liu X, Pu P et al (2010) Inactivation of PI3K/AKT signaling inhibits glioma cell growth through modulation of beta‐catenin‐mediated transcription. Brain Res 1366:9–17. [DOI] [PubMed] [Google Scholar]
- 64. Harris MA, Yang H, Low BE, Mukherje J, Guha A, Bronson RT et al (2008) Cancer stem cells are enriched in the side population cells in a mouse model of glioma. Cancer Res 68:10051–10059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Heddleston JM, Li ZZ, McLendon RE, Hjelmeland AB, Rich JN (2009) The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle 8:3274–3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Heldin CH, Ostman A, Ronnstrand L (1998) Signal transduction via platelet‐derived growth factor receptors. BBA-Rev Cancer 1378:F79–F113. [DOI] [PubMed] [Google Scholar]
- 67. Hemmati HD, Nakano I, Lazareff JA, Masterman‐Smith M, Geschwind DH, Bronner‐Fraser M, Kornblum HI (2003) Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A 100:15178–15183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Hockfield S, McKay RDG (1985) Identification of major cell classes in the developing mammalian nervous‐system. J Neurosci 5:3310–3328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Holland EC, Celestino J, Dai CK, Schaefer L, Sawaya RE, Fuller GN (2000) Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nat Genet 25:55–57. [DOI] [PubMed] [Google Scholar]
- 70. Horiguchi S, Takahashi J, Kishi Y, Morizane A, Okamoto Y, Koyanagi M et al (2004) Neural precursor cells derived from human embryonic brain retain regional specificity. J Neurosci Res 75:817–824. [DOI] [PubMed] [Google Scholar]
- 71. Howng SL, Wu CH, Cheng TS, Sy WD, Lin PCK, Wang CH, Hong YR (2002) Differential expression of Wnt genes, beta‐catenin and E‐cadherin in human brain tumors. Cancer Lett 183:95–101. [DOI] [PubMed] [Google Scholar]
- 72. Hsieh A, Ellsworth R, Hsieh D (2011) Hedgehog/GLI1 regulates IGF dependent malignant behaviors in glioma stem cells. J Cell Physiol 226:1118–1127. [DOI] [PubMed] [Google Scholar]
- 73. Hsieh D, Hsieh A, Stea B, Ellsworth R (2010) IGFBP2 promotes glioma tumor stem cell expansion and survival. Biochem Biophys Res Commun 397:367–372. [DOI] [PubMed] [Google Scholar]
- 74. Huang LY, DeVries GJ, Bittman EL (1998) Photoperiod regulates neuronal bromodeoxyuridine labeling in the brain of a seasonally breeding mammal. J Neurobiol 36:410–420. [DOI] [PubMed] [Google Scholar]
- 75. Hyun K‐H, Yoon C‐H, Kim R‐K, Lim E‐J, An S, Park M‐J et al (2011) Eckol suppresses maintenance of sternness and malignancies in glioma stem‐like cells. Toxicol Appl Pharmacol 254:32–40. [DOI] [PubMed] [Google Scholar]
- 76. Inoue A, Takahashi H, Harada H, Kohno S, Ohue S, Kobayashi K et al (2010) Cancer stem‐like cells of glioblastoma characteristically express MMP‐13 and display highly invasive activity. Int J Oncol 37:1121–1131. [DOI] [PubMed] [Google Scholar]
- 77. Jacques TS, Swales A, Brzozowski MJ, Henriquez NV, Linehan JM, Mirzadeh Z et al (2010) Combinations of genetic mutations in the adult neural stem cell compartment determine brain tumour phenotypes. EMBO J 29:222–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Jalali A, Bassuk AG, Kan L, Israsena N, Mukhopadhyay A, McGuire T, Kessler JA (2011) HeyL promotes neuronal differentiation of neural progenitor cells. J Neurosci Res 89:299–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Jeong Y‐H, Sekiya M, Hirata M, Ye M, Yamagishi A, Lee S‐M et al (2010) The low‐density lipoprotein receptor‐related protein 10 is a negative regulator of the canonical Wnt/beta‐catenin signaling pathway. Biochem Biophys Res Commun 392:495–499. [DOI] [PubMed] [Google Scholar]
- 80. Jhanwar‐Uniyal M, Albert L, McKenna E, Karsy M, Rajdev P, Braun A, Murali R (2011) Deciphering the signaling pathways of cancer stem cells of glioblastoma multiforme: role of Akt/mTOR and MAPK pathways. Adv Enzyme Regul 51:164–170. [DOI] [PubMed] [Google Scholar]
- 81. Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Frisen J (1999) Identification of a neural stem cell in the adult mammalian central nervous system. Cell 96:25–34. [DOI] [PubMed] [Google Scholar]
- 82. Joo KM, Kim SY, Jin X, Song SY, Kong DS, Lee JI et al (2008) Clinical and biological implications of CD133‐positive and CD133‐negative cells in glioblastomas. Lab Invest 88:808–815. [DOI] [PubMed] [Google Scholar]
- 83. Joyce D, Bouzahzah B, Fu MF, Albanese C, D'Amico M, Steer J et al (1999) Integration of Rac‐dependent regulation of cyclin D1 transcription through a nuclear factor‐kappa B‐dependent pathway. J Biol Chem 274:25245–25249. [DOI] [PubMed] [Google Scholar]
- 84. Kalani MYS, Cheshier SH, Cord BJ, Bababeygy SR, Vogel H, Weissman IL et al (2008) Wnt‐mediated self‐renewal of neural stem/progenitor cells. Proc Natl Acad Sci U S A 105:16970–16975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Kamijo T, Bodner S, van de Kamp E, Randle DH, Sherr CJ (1999) Tumor spectrum in ARF‐deficient mice. Cancer Res 59:2217–2222. [PubMed] [Google Scholar]
- 86. Kang MK, Kang SK (2007) Tumorigenesis of chemotherapeutic drug‐resistant cancer stem‐like cells in brain glioma. Stem Cells Dev 16:837–847. [DOI] [PubMed] [Google Scholar]
- 87. Kang MK, Hur BI, Ko MH, Kim CH, Cha SH, Kang SK (2008) Potential identity of multi‐potential cancer stem‐like subpopulation after radiation of cultured brain glioma. BMC Neurosci 9:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Kenney AM, Rowitch DH (2000) Sonic hedgehog promotes G(1) cyclin expression and sustained cell cycle progression in mammalian neuronal precursors. Mol Cell Biol 20:9055–9067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kim KJ, Lee KH, Kim HS, Moon KS, Jung TY, Jung S, Lee MC (2011) The presence of stem cell marker‐expressing cells is not prognostically significant in glioblastomas. Neuropathology 31:494–502. [DOI] [PubMed] [Google Scholar]
- 90. Kleber M, Sommer L (2004) Wnt signaling and the regulation of stem cell function. Curr Opin Cell Biol 16:681–687. [DOI] [PubMed] [Google Scholar]
- 91. Kondo T, Setoguchi T, Taga T (2004) Persistence of a small subpopulation of cancer stem‐like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A 101:781–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Cacerescortes J et al (1994) A cell initiating human acute myeliod‐leukemia after transplanation into scid mice. Nature 367:645–648. [DOI] [PubMed] [Google Scholar]
- 93. Lathia JD, Mattson MP, Cheng A (2008) Notch: from neural development to neurological disorders. J Neurochem 107:1471–1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Lathia JD, Gallagher J, Heddleston JM, Wang J, Eyler CE, MacSwords J et al (2010) Integrin Alpha 6 regulates glioblastoma stem cells. Stem Cells 6:421–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lee J, Kotliarova S, Kotliarov Y, Li AG, Su Q, Donin NM et al (2006) Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum‐cultured cell lines. Cancer Cells 9:391–403. [DOI] [PubMed] [Google Scholar]
- 96. Lendahl U, Zimmerman LB, McKay RDG (1990) CNS stem cells express a new class of intermediate filament protein. Cell 60:585–595. [DOI] [PubMed] [Google Scholar]
- 97. Li Y, Guessous F, Zhang Y, DiPierro C, Kefas B, Johnson E et al (2009) MicroRNA‐34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res 69:7569–7576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Lim DA, Tramontin AD, Trevejo JM, Herrera DG, Garcia‐Verdugo JM, Alvarez‐Buylla A (2000) Noggin antagonizes BMP signaling to create a niche for adult neurogenesis. Neuron 28:713–726. [DOI] [PubMed] [Google Scholar]
- 99. Llaguno SA, Chen J, Kwon C‐H, Jackson EL, Li Y, Burns DK et al (2009) Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse models. Cancer Cells 15:45–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lottaz C, Beier D, Meyer K, Kumar P, Hermann A, Schwarz J et al (2010) Transcriptional profiles of CD133(+) and CD133(−) glioblastoma‐derived cancer stem cell lines suggest different cells of origin. Cancer Res 70:2030–2040. [DOI] [PubMed] [Google Scholar]
- 101. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A et al (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol (Berl) 114:97–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Luzzati F, De Marchis S, Fasolo A, Peretto P (2006) Neurogenesis in the caudate nucleus of the adult rabbit. J Neurosci 26:609–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ma HI, Chiou SH, Hueng DY, Tai LK, Huang PI, Kao CL et al (2011) Celecoxib and radioresistant glioblastoma‐derived CD133(+) cells: improvement in radiotherapeutic effects laboratory investigation. J Neurosurg 114:651–662. [DOI] [PubMed] [Google Scholar]
- 104. Maness PF, Schachner M (2007) Neural recognition molecules of the immunoglobulin superfamily: signaling transducers of axon guidance and neuronal migration. Nat Neurosci 10:19–26. [DOI] [PubMed] [Google Scholar]
- 105. Marumoto T, Tashiro A, Friedmann‐Morvinski D, Scadeng M, Soda Y, Gage FH, Verma IM (2009) Development of a novel mouse glioma model using lentiviral vectors. Nat Med 15:110–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Ming JE, Roessler E, Muenke M (1998) Human developmental disorders and the Sonic hedgehog pathway. Mol Med Today 4:343–349. [DOI] [PubMed] [Google Scholar]
- 107. Miranti CK, Brugge JS (2002) Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol 4:E83–E90. [DOI] [PubMed] [Google Scholar]
- 108. Mizutani K, Yoon K, Dang L, Tokunaga A, Gaiano N (2007) Differential Notch signalling distinguishes neural stem cells from intermediate progenitors. Nature 449:351–355. [DOI] [PubMed] [Google Scholar]
- 109. Molofsky AV, He SH, Bydon M, Morrison SJ, Pardal R (2005) Bmi‐1 promotes neural stem cell self‐renewal and neural development but not mouse growth and survival by repressing the p16(Ink4a) and P19(Arf) senescence pathways. Genes Dev 19:1432–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Niklasson M, Bergström T, Zhang X‐Q, Gustafsdottir SM, Sjögren M, Edqvist P‐H et al (2010) Enlarged lateral ventricles and aberrant behavior in mice overexpressing PDGF‐B in embryonic neural stem cells. Exp Cell Res 316:2779–2789. [DOI] [PubMed] [Google Scholar]
- 111. Nishino J, Kim I, Chada K, Morrison SJ (2008) Hmga2 promotes neural stem cell self‐renewal in young but not old mice by reducing p16(Ink4a) and p19(Arf) expression. Cell 135:227–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G, Jiang L et al (2003) Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat Med 9:439–447. [DOI] [PubMed] [Google Scholar]
- 113. Ogden AT, Waziri AE, Lochhead RA, Fusco D, Lopez K, Ellis JA et al (2008) Identification of A2B5(+)CD133‐tumor‐initiating cells in adult human gliomas. Neurosurgery 62:505–514. [DOI] [PubMed] [Google Scholar]
- 114. Ostermann S, Csajka C, Buclin T, Leyvraz S, Lejeune F, Decosterd LA, Stupp R (2004) Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients. Clin Cancer Res 10:3728–3736. [DOI] [PubMed] [Google Scholar]
- 115. Panchision DM, McKay RDG (2002) The control of neural stem cells by morphogenic signals. Curr Opin Genet Dev 12:478–487. [DOI] [PubMed] [Google Scholar]
- 116. Pasternak JJ (2005) An Introduction to Human Molecular Genetics. John Wiley and Sons: Hoboken, NJ. [Google Scholar]
- 117. Peng SB, Peek V, Zhai Y, Paul DC, Lou QY, Xia XL et al (2005) Akt activation, but not extracellular signal—regulated kinase activation, is required for SDF‐1 alpha/CXCR4‐mediated migration of epitheloid carcinoma cells. Mol Cancer Res 3:227–236. [DOI] [PubMed] [Google Scholar]
- 118. Penuelas S, Anido J, Prieto‐Sanchez RM, Folch G, Barba I, Cuartas I et al (2009) TGF‐beta increases glioma‐initiating cell self‐renewal through the induction of LIF in human glioblastoma. Cancer Cells 15:315–327. [DOI] [PubMed] [Google Scholar]
- 119. Phillips HS, Kharbanda S, Chen RH, Forrest WF, Soriano RH, Wu TD et al (2006) Molecular subclasses of high‐grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cells 9:157–173. [DOI] [PubMed] [Google Scholar]
- 120. Piccirillo SGM, Reynolds BA, Zanetti N, Lamorte G, Binda E, Broggi G et al (2006) Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour‐initiating cells. Nature 444:761–765. [DOI] [PubMed] [Google Scholar]
- 121. Purow BW, Haque RM, Noel MW, Su Q, Burdick MJ, Lee J et al (2005) Expression of Notch‐1 and its ligands, Delta‐like‐1 and Jagged‐1, is critical for glioma cell survival and proliferation. Cancer Res 65:2353–2363. [DOI] [PubMed] [Google Scholar]
- 122. Purow BW, Sundaresan TK, Burdick MJ, Kefas BA, Comeau LD, Hawkinson MP et al (2008) Notch‐1 regulates transcription of the epidermal growth factor receptor through p53. Carcinogenesis 29:918–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Pursglove SE, Mackay JP (2005) CSL: a notch above the rest. Int J Biochem Cell Biol 37:2472–2477. [DOI] [PubMed] [Google Scholar]
- 124. Qiang L, Yang Y, Ma Y‐J, Chen F‐H, Zhang L‐B, Liu W et al (2009) Isolation and characterization of cancer stem like cells in human glioblastoma cell lines. Cancer Lett 279:13–21. [DOI] [PubMed] [Google Scholar]
- 125. Rahaman SO, Vogelbaum NA, Haque SJ (2005) Aberrant stat3 signaling by interleukin‐4 in malignant glioma cells: involvement of IL‐13R alpha 2. Cancer Res 65:2956–2963. [DOI] [PubMed] [Google Scholar]
- 126. Raveh S, Gavert N, Ben‐Ze'ev A (2009) L1 cell adhesion molecule (L1CAM) in invasive tumors. Cancer Lett 282:137–145. [DOI] [PubMed] [Google Scholar]
- 127. Redjal N, Chan JA, Segal RA, Kung AL (2006) CXCR4 inhibition synergizes with cytotoxic chemotherapy in gliomas. Clin Cancer Res 12:6765–6771. [DOI] [PubMed] [Google Scholar]
- 128. Rempel SA, Dudas S, Ge SG, Gutierrez JA (2000) Identification and localization of the cytokine SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis in human glioblastoma. Clin Cancer Res 6:102–111. [PubMed] [Google Scholar]
- 129. Rodrigues JC, Gonzalez GC, Zhang L, Ibrahim G, Kelly JJ, Gustafson MP et al (2010) Normal human monocytes exposed to glioma cells acquire myeloid‐derived suppressor cell‐like properties. Neuro-Oncol 12:351–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Ronchini C, Capobianco AJ (2001) Induction of cyclin D1 transcription and CDK2 activity by Notch(ic): implication for cell cycle disruption in transformation by Notch(ic). Mol Cell Biol 21:5925–5934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Ruiz i Altaba A, Sanchez P, Dahmane N (2002) Gli and Hedgehog in cancer: tumours, embryos and stem cells. Nat Rev Cancer 2:361–372. [DOI] [PubMed] [Google Scholar]
- 132. Ruiz i Altaba A, Stecca B, Sanchez P (2004) Hedgehog‐Gli signaling in brain tumors: stem cells and paradevelopmental programs in cancer. Cancer Lett 204:145–157. [DOI] [PubMed] [Google Scholar]
- 133. Schmid RS, Maness PF (2008) L1 and NCAM adhesion molecules as signaling coreceptors in neuronal migration and process outgrowth. Curr Opin Neurobiol 18:245–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Schmitz M, Temme A, Senner V, Ebner R, Schwind S, Stevanovic S et al (2007) Identification of SOX2 as a novel glioma‐associated antigen and potential target for T cell‐based immunotherapy. Br J Cancer 96:1293–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Schulte A, Gunther HS, Phillips HS, Kemming D, Martens T, Kharbanda S et al (2011) A distinct subset of glioma cell lines with stem cell‐like properties reflects the transcriptional phenotype of glioblastomas and overexpresses CXCR4 as therapeutic target. Glia 59:590–602. [DOI] [PubMed] [Google Scholar]
- 136. Seidel S, Garvalov BK, Wirta V, von Stechow L, Schanzer A, Meletis K et al (2010) A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain 133:983–995. [DOI] [PubMed] [Google Scholar]
- 137. Shahi MH, Lorente A, Castresana JS (2008) Hedgehog signalling in medulloblastoma, glioblastoma and neuroblastoma. Oncol Rep 19:681–688. [PubMed] [Google Scholar]
- 138. Sharma MK, Mansur DB, Reifenberger G, Perry A, Leonard JR, Aldape KD et al (2007) Distinct genetic signatures among pilocytic astrocytomas relate to their brain region origin. Cancer Res 67:890–900. [DOI] [PubMed] [Google Scholar]
- 139. Sharma VM, Calvo JA, Draheim KM, Cunningham LA, Hermance N, Beverly L et al (2006) Notch1 contributes to mouse T‐cell leukemia by directly inducing the expression of c‐myc. Mol Cell Biol 26:8022–8031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Sherry MM, Reeves A, Wu JLK, Cochran BH (2009) STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 27:2383–2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Shi L, Zhang S, Feng K, Wu F, Wan Y, Wang Z et al (2012) MicroRNA‐125b‐2 confers human glioblastoma stem cells resistance to temozolomide through the mitochondrial pathway of apoptosis. Int J Oncol 40:119–129. [DOI] [PubMed] [Google Scholar]
- 142. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB (2003) Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821–5828. [PubMed] [Google Scholar]
- 143. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T et al (2004) Identification of human brain tumour initiating cells. Nature 432:396–401. [DOI] [PubMed] [Google Scholar]
- 144. Smith JS, Tachibana I, Passe SM, Huntley BK, Borell TJ, Iturria N et al (2001) PTEN mutation, EGFR amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst 93:1246–1256. [DOI] [PubMed] [Google Scholar]
- 145. Soeda A, Inagaki A, Oka N, Ikegame Y, Aoki H, Yoshimura S et al (2008) Epidermal growth factor plays a crucial role in mitogenic regulation of human brain tumor stem cells. J Biol Chem 283:10958–10966. [DOI] [PubMed] [Google Scholar]
- 146. Son MJ, Woolard K, Nam D‐H, Lee J, Fine HA (2009) SSEA‐1 is an enrichment marker for tumor‐initiating cells in human glioblastoma. Stem Cells 4:440–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Stockhausen M‐T, Kristoffersen K, Poulsen HS (2010) The functional role of Notch signaling in human gliomas. Neuro-Oncol 12:199–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Stockhausen M‐T, Broholm H, Villingshoj M, Kirchhoff M, Gerdes T, Kristoffersen K et al (2011) Maintenance of EGFR and EGFRvIII expressions in an in vivo and in vitro model of human glioblastoma multiforme. Exp Cell Res 317:1513–1526. [DOI] [PubMed] [Google Scholar]
- 149. Strojnik T, Røsland GV, Sakariassen PO, Kavalar R, Lah T (2007) Neural stem cell markers, nestin and musashi proteins, in the progression of human glioma: correlation of nestin with prognosis of patient survival. Surg Neurol 68:133–143. [DOI] [PubMed] [Google Scholar]
- 150. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996. [DOI] [PubMed] [Google Scholar]
- 151. Subkhankulova T, Zhang X, Leung C, Marino S (2010) Bmi1 directly represses p21(Waf1/Cip1) in Shh‐induced proliferation of cerebellar granule cell progenitors. Mol Cell Neurosci 45:151–162. [DOI] [PubMed] [Google Scholar]
- 152. Sun H, Zhang S (2011) Arsenic trioxide regulates the apoptosis of glioma cell and glioma stem cell via down‐regulation of stem cell marker Sox2. Biochem Biophys Res Commun 410:692–697. [DOI] [PubMed] [Google Scholar]
- 153. Tabatabai G, Weller M (2011) Glioblastoma stem cells. Cell Tissue Res 343:459–465. [DOI] [PubMed] [Google Scholar]
- 154. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–676. [DOI] [PubMed] [Google Scholar]
- 155. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–872. [DOI] [PubMed] [Google Scholar]
- 156. Talora C, Campese AF, Bellavia D, Felli MP, Vacca A, Gulino A, Screpanti I (2008) Notch signaling and diseases: an evolutionary journey from a simple beginning to complex outcomes. BBA-Mol Basis Dis 1782:489–497. [DOI] [PubMed] [Google Scholar]
- 157. Tchoghandjian A, Baeza N, Colin C, Cayre M, Metellus P, Beclin C et al (2010) A2B5 cells from human glioblastoma have cancer stem cell properties. Brain Pathol 20:211–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Tejada‐Simon MV, Villasana LE, Serrano F, Klann E (2006) NMDA receptor activation induces translocation and activation of Rac in mouse hippocampal area CA1. Biochem Biophys Res Commun 343:504–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Thon N, Damianoff K, Hegermann J, Grau S, Krebs B, Schnell O et al (2010) Presence of pluripotent CD133(+) cells correlates with malignancy of gliomas. Mol Cell Neurosci 43:51–59. [DOI] [PubMed] [Google Scholar]
- 160. Tropepe V, Sibilia M, Ciruna BG, Rossant T, Wagner EF, van der Kooy D (1999) Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev Biol 208:166–188. [DOI] [PubMed] [Google Scholar]
- 161. Tunici P, Bissola L, Lualdi E, Pollo B, Cajola L, Broggi G et al (2004) Genetic alterations and in vivo tumorigenicity of neurospheres derived from an adult glioblastoma. Mol Cancer 3:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Ulasov IV, Nandi S, Dey M, Sonabend AM, Lesniak MS (2011) Inhibition of sonic hedgehog and notch pathways enhances sensitivity of CD133(+) glioma stem cells to temozolomide therapy. Mol Med 17:103–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Vicente‐Manzanares M, Rey M, Jones DR, Sancho D, Mellado M, Rodriguez‐Frade JM et al (1999) Involvement of phosphatidylinositol 3‐kinase in stromal cell‐derived factor‐1 alpha‐induced lymphocyte polarization and chemotaxis. J Immunol 163:4001–4012. [PubMed] [Google Scholar]
- 164. Vik‐Mo EO, Sandberg C, Joel M, Stangeland B, Watanabe Y, Mackay‐Sim A et al (2011) A comparative study of the structural organization of spheres derived from the adult human subventricular zone and glioblastoma biopsies. Exp Cell Res 317:1049–1059. [DOI] [PubMed] [Google Scholar]
- 165. Visvader JE (2011) Cells of origin in cancer. Nature 469:314–322. [DOI] [PubMed] [Google Scholar]
- 166. Visvader JE, Lindeman GJ (2008) Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8:755–768. [DOI] [PubMed] [Google Scholar]
- 167. Voyant C, Roustit R, Tatje J, Baffi K, Leschi D, Braincon J (2011) Therapeutic potential of atmospheric neutrons. Rep Pract Oncol Radiother 16:21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Wan F, Herold‐Mende C, Campos B, Centner FS, Dictus C, Becker N et al (2011) Association of stem cell‐related markers and survival in astrocytic gliomas. Biomarkers 16:136–143. [DOI] [PubMed] [Google Scholar]
- 169. Wang J, Sakariassen PO, Tsinkalovsky O, Immervoll H, Boe SO, Svendsen A et al (2008) CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int J Cancer 122:761–768. [DOI] [PubMed] [Google Scholar]
- 170. Wang J, Wakeman TP, Lathia JD, Hjelmeland AB, Wang X‐F, White RR et al (2010) Notch promotes radioresistance of glioma stem cells. Stem Cells 28:17–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Wang Y, Yang J, Zheng H, Tomasek GJ, Zhang P, McKeever PE et al (2009) Expression of mutant p53 proteins implicates a lineage relationship between neural stem cells and malignant astrocytic glioma in a murine model. Cancer Cells 15:514–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Wei J, Barr J, Kong LY, Wang YT, Wu A, Sharma AK et al (2010) Glioblastoma cancer‐initiating cells inhibit T‐cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther 9:67–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Wick W, Naumann U, Weller M (2006) Transforming growth factor‐beta: a molecular target for the future therapy of glioblastoma. Curr Pharm Des 12:341–349. [DOI] [PubMed] [Google Scholar]
- 174. Wu MH, Chen Q, Li D, Li XL, Li XY, Huang C et al (2008) LRRC4 inhibits human glioblastoma cells proliferation, invasion, and proMMP‐2 activation by reducing SDF‐1 alpha/CXCR4‐mediated ERK1/2 and Akt signaling pathways. J Cell Biochem 103:245–255. [DOI] [PubMed] [Google Scholar]
- 175. Xu Q, Yuan X, Liu G, Black KL, Yu JS (2008) Hedgehog signaling regulates brain tumor‐initiating cell proliferation and portends shorter survival for patients with PTEN‐coexpressing glioblastomas. Stem Cells 26:3018–3026. [DOI] [PubMed] [Google Scholar]
- 176. Xu Y, Tamamaki N, Noda T, Kimura K, Itokazu Y, Matsumoto N et al (2005) Neurogenesis in the ependymal layer of the adult rat 3rd ventricle. Exp Neurol 192:251–264. [DOI] [PubMed] [Google Scholar]
- 177. Ying M, Wang S, Sang Y, Sun P, Lal B, Goodwin CR et al (2011) Regulation of glioblastoma stem cells by retinoic acid: role for Notch pathway inhibition. Oncogene 30:3454–3467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Yoon C‐H, Hyun K‐H, Kim R‐K, Lee H, Lim E‐J, Chung H‐Y et al (2011) The small GTPase Rac1 is involved in the maintenance of stemness and malignancies in glioma stem‐like cells. FEBS Lett 585:2331–2338. [DOI] [PubMed] [Google Scholar]
- 179. Young HF, Sakalas R, Kaplan AM (1976) Inhibition of cell‐mediated immunity in patients with brain tumors. Surg Neurol 5:19–23. [PubMed] [Google Scholar]
- 180. Yu S, Ping Y, Yi L, Zhou Z, Chen H, Yao X et al (2008) Isolation and characterization of cancer stem cells from a human glioblastoma cell line U87. Cancer Lett 265:124–134. [DOI] [PubMed] [Google Scholar]
- 181. Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N et al (2008) Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res 14:123–129. [DOI] [PubMed] [Google Scholar]
- 182. Zhang M, Song T, Yang L, Chen R, Wu L, Yang Z, Fang J (2008) Nestin and CD133: valuable stem cell‐specific markers for determining clinical outcome of glioma patients. J Exp Clin Cancer Res 27:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Zhao M, Momma S, Delfani K, Carlen M, Cassidy RM, Johansson CB et al (2003) Evidence for neurogenesis in the adult mammalian substantia nigra. Proc Natl Acad Sci U S A 100:7925–7930. [DOI] [PMC free article] [PubMed] [Google Scholar]
