Next Article in Journal
Role of Protein Kinases in Hedgehog Pathway Control and Implications for Cancer Therapy
Next Article in Special Issue
Proteomic Analysis Implicates Vimentin in Glioblastoma Cell Migration
Previous Article in Journal
Latent Cytomegalovirus Infection in Female Mice Increases Breast Cancer Metastasis
Previous Article in Special Issue
The ‘Ins and Outs’ of Early Preclinical Models for Brain Tumor Research: Are They Valuable and Have We Been Doing It Wrong?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of SVZ Stem Cells in Glioblastoma

by
Christine Altmann
1,
Stefanie Keller
1 and
Mirko H. H. Schmidt
1,2,3,*
1
Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
2
German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt/55131 Mainz, Germany
3
German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(4), 448; https://doi.org/10.3390/cancers11040448
Submission received: 27 February 2019 / Revised: 22 March 2019 / Accepted: 26 March 2019 / Published: 29 March 2019
(This article belongs to the Special Issue Glioblastoma: State of the Art and Future Perspectives)

Abstract

:
As most common primary brain cancer, glioblastoma is also the most aggressive and malignant form of cancer in the adult central nervous system. Glioblastomas are genetic and transcriptional heterogeneous tumors, which in spite of intensive research are poorly understood. Over the years conventional therapies failed to affect a cure, resulting in low survival rates of affected patients. To improve the clinical outcome, an important approach is to identify the cells of origin. One potential source for these are neural stem cells (NSCs) located in the subventricular zone, which is one of two niches in the adult nervous system where NSCs with the capacity of self-renewal and proliferation reside. These cells normally give rise to neuronal as well as glial progenitor cells. This review summarizes current findings about links between NSCs and cancer stem cells in glioblastoma and discusses current therapeutic approaches, which arise as a result of identifying the cell of origin in glioblastoma.

1. Introduction

Glioblastoma (GB) is the most frequent form of brain tumor in adults and is associated with a poor prognosis and a short median patient survival [1]. Limited therapeutic options, combined with a poor response to currently used therapies, increased the pressure to discover new genetic, epigenetic and molecular pathways involved in GB to create new therapies. One of the most significant questions in GB research is aiming at the hierarchical organization and the cell of origin. Conventional theories state that cancer arises from an accumulation of somatic mutations, resulting in uncontrolled proliferation as well as selective growth advantage [2,3]. Most commonly, cancer occurs in epithelial tissues [4]. Whether a tumor originates from a differentiated cell, which regains the ability to proliferate, or whether it originates from a stem cell, which already has the capacity to proliferate, is not fully resolved, and depends on the tissue and the tumor itself. The existence of brain tumor propagating cells (BTPCs) and their molecular, genetic, and epigenetic footprint could open new ways of therapeutic approaches. In the last years, diverse tumors could be retraced to mutations in stem cells [4] and various studies have suggested that NSCs might be the cells of origin of GB, including mutated astrocyte-like NSCs from the SVZ [5,6,7,8]. Recent studies reported from clinics and mouse models that glioblastoma arise from migration of mutated astrocyte-like NSCs from the SVZ [8].

2. Glioblastoma

2.1. General Facts

Glioma is an umbrella term, compromising around 30 percent of all brain tumors that are thought to grow from intrinsic glia cells. As an umbrella term glioma consolidates different types of tumors including ependymoma, astrocytoma, and oligodendroglioma, which vary in their symptoms, aggressiveness, malignancy, and treatment strategy.
Glioblastoma multiforme (GB) belongs to the category of astrocytoma, is the most common and most aggressive of all malignant glial tumor in adults [1], and is less common in children [9]. Based on the World Health Organization classification, GB is the most malignant form of glioma and is classified as a grade IV tumor (ICD-O 9440/3) [10]. GB can be divided into primary (arising de novo) or secondary (developed from a pre-existing tumor) intrinsic brain tumor, however, 90% of all GB are primary [9]. Specific mutations in the gene of isocitrate dehydrogenase (IDH) 1/2 are characteristic for secondary glioblastomas, which are more frequent in younger patients. High invasiveness of GB is recorded, with tumor cells mainly spreading into distinct brain regions, whereas metastasis into other organs is infrequent [1].
Diagnosis of GB comes with a poor prognosis with high morbidity and mortality [1]. The median survival of patients diagnosed with GB and treated with the common medication is only 12 to 15 months [1]. GB can occur in each age group; however, most of the patients are between 45–75 years old. Gliomas are mainly located in the cerebral cortex of adult brains, with 40% in the frontal lobe, followed by the temporal lobe (29%), the parietal lobe (14%), the occipital lobe (3%) and 14 % of gliomas are positioned in deeper brain structures [11].

2.2. Genetic Alterations

GB features a complex pathogenesis that involves mutations and alterations of several key cellular pathways, associated with cell proliferation, angiogenesis, migration, and survival [9]. However, the lack of effective therapies increases the importance to understand pathogenesis in detail. Cellular signaling pathways involved in GB are reviewed in [9,12].
The most common mutations in GB are found in p53 (85.3–87%) [13,14], the epidermal growth factor receptor (EGFR) (45–57%) [13,14,15], the platelet-derived growth factor receptor (PDGFR) (60%) [16,17] the mouse double minute homolog 2 (10–15%) (MDM2) [18], the phosphatase and tensin homolog (PTEN) gene (20–34%) [19,20], the RTK/Ras/PI3K signaling pathway (86–89.6%) [13,14] and in the pRB signaling pathway (77–78.9%) [13,16].

2.3. Conventional Therapy

The current therapy of GB is limited and inefficient and focuses on surgical resection of as much of the tumorigenic tissue as possible with subsequent radiation- and chemotherapy, hereby mostly using oral alkylating agent temozolomide [21]. However, this therapy strategy is insufficient and no adequate cure for GB was described, yet. One major disadvantage of GB is the tissue-distribution pattern of the tumors resulting from dispersion of the tumors cells in the neighboring brain tissue [22,23]. This characteristic hinders complete surgical resection, increases recurrence rate and thereby reduces healing abilities.
Despite ongoing research, survival of GB patients could not be increased in the last years. However, there are rare case reports about long-term survival and zero recurrence of single patients with GB, which are explained by young age, as well as aggressive and complete surgical removal [24,25]. These cases, as rarely as they are, rise the hope that GB can be cured and show that more research effort and innovative treatment approaches are desperately needed to better understand the tumor development and progression.

3. Neural Stem Cell Niches in the Adult Brain

Neural stem cells (NSCs), a subpopulation of astroglial cells, are self-renewing cells with the capacity to differentiate into multiple neural cell types like neurons and glial cells (astrocytes and oligodendrocytes) (reviewed in [26]). During development, NSCs are obligatory for the formation of the nervous system. They are most active in this period; however, since 1992 it is described that NSCs can also be found in the adult brain. Here, small populations of NSCs are located in specific stem cell niches that divide occasionally to generate differentiated cells including neurons (neurogenesis) and glial cells (gliogenesis) [27,28].

3.1. Adult Neurogenesis

NSCs have a relative slow division rate and generate progenitor cells, which are able to differentiate into one of the three major cell lineages of the brain: neurons, astrocytes and oligodendrocytes [26,29,30]. The two neurogenic niches in the adult rodent brain, which contain NSCs and produce new neural cells, are the subventricular zone (SVZ) of the lateral ventricle and the dentate gyrus of the hippocampus [31,32]. While adult neurogenesis in the SVZ of humans is widely accepted [33,34], it is still under discussion in the hippocampus [33,35,36,37]. Recently, Sorrells et al. showed that neurogenesis in the dentate gyrus declines sharply in children and that in the adult brain they could not detect any new neurons [36]. However, shortly after this publication, Boldrini et al. published a manuscript stating that neurogenesis still persists throughout life [37]. Thus, it is still controversial if the potential to produce new neurons still exists in the adult brain; but many studies show that it is drastically decreased as compared to embryonic stages [5,33,38,39]. During the last decades adult neurogenesis was also described in other mammalian brain regions, including the hypothalamus, the cortex, the striatum, and the amygdala [40,41,42,43]. Furthermore, an increase of neurogenesis in the adult brain was reported after injury and in disease [44,45,46,47,48].
The impact and consequence of adult neurogenesis in humans is largely unknown and can only be estimated. However, many studies suggest that the generation of new neurons in the adult brain could be important for learning and memory, degradation as well as regeneration processes underlying aging, injury and diseases, including dementia and cancer [49,50,51,52,53,54].

3.2. Stem Cells in the Subventricular Zone

The SVZ is the largest neurogenic niche in the adult mammalian brain. It is located at the border of both lateral cerebral ventricles, fitting in between the ependyma and the parenchyma of the striatum. The NSCs in the SVZ are found in an astrocytic ribbon in the sub-ependymal zone. They are surrounded by ependymal cells, vascular endothelial cells, astrocytes, and oligodendrocytes [5], which are important to support the stem cells and to control the proliferation rate [55,56]. Next to the SVZ, the brain parenchyma is located, which is mostly composed of differentiated neurons and glia cells.
In adult rodents, the SVZ contains four major cell types: ependymal cells, NSCs, fast proliferation precursors and neuroblasts. The ependymal cells form a monolayer-border to the ventricle. This layer is followed by the other three cell types, which are not arranged in layers and keep close contact to the ependymal layer. Astrocyte-like NSCs have an apical cilium, which extends into the ventricle lumen and might influence cell proliferation and differentiation [57,58,59]. These astrocyte-like NSCs (type-B cells) occasionally give rise to multipotent intermediate progenitors (type-C cells), which correspond to transit-amplifying cells that further divide to generate neuroblasts (type-A cells) (reviewed in detail in [60]). In rodents, these neuroblasts migrate in chains along the rostral migratory stream (RMS) to the cortical layer of the olfactory bulb (OB) [61,62], where they differentiate into interneurons and integrate into the pre-existing neural circuitry [61,62,63,64].
NSCs in the adult human SVZ were first described in 2004 by Sanai et al. [5], followed by several groups, which identified and isolated human NSCs from the anterior SVZ (Figure 1) [65,66,67]. In the human SVZ, there are some differences in the anatomical organization and in the behavior of NSCs compared to rodents:
(I) The human SVZ features a hypocellular gap between the ependymal layer and the neurogenic astrocytes, which mainly contains ependymal and astrocytic expansions [5,38,68]. This layer is missing in rodents.
(II) The SVZ in humans can be separated into four distinct layers, whereas the rodent SVZ is not separated into layers. The first layer (ependymal layer, layer I) lines the ventricle and is composed of ependymal cells that possess several apical microvilli and basal expansions [38]. This layer is followed by the hypocellular gap forming the second layer (hypocellular layer, layer II) which is characterized by the absence of cell bodies and has an unknown function. As it mainly contains astrocytic and ependymal processes, one theory would be that these are forming connections in layer II [60,69,70]. The third layer (astrocytic ribbon, layer III) mainly contains the astrocyte-like NSCs and neuroblasts [5], whereas the forth layer, the transitional zone (layer IV) mainly contains myelinated axons and oligodendrocytes [38].
(III) In the human SVZ, multipotent transit-amplifying cells (in rodents: type-C cells) are missing. Only NSCs and neuroblasts can be found in the human SVZ.
(IV) The migration of neural progenitor cells from the SVZ along the RMS to the olfactory bulb is still under discussion and the migration of non-neuronal progenitor cells is at least reduced [5,33,34,67,71], whereas migration of these in rodents is verified. In the OB, mostly new oligodendrocytes from the SVZ are needed to maintain the myelin sheath of the neurons [72,73]. The fate of newborn neurons is unclear. It was shown by Ernst et al. in 2014 that in the adult human brain, new neurons from the SVZ are integrated not into the OB but in form of interneurons into the striatum [74]. Thus, the SVZ is still considered as an important pool of neuronal and glial progenitor cells in the adult mammalian brain, which provides an opportunity for neuroregenerative repair, learning, and memory [75,76,77,78,79]. However, this pool was also implicated in other conditions such as injury, neurodegeneration and cancer [80,81,82,83,84].

4. Cancer Stem Cells

Gradually, many studies have reported that tumor cells are not homogenous, but that a tumor consists of a variety of cell types forming a hierarchical organization containing slowly dividing cancer stem cells (CSCs) rapidly dividing precursor cells and non-dividing differentiated cells [85,86]. This cellular hierarchy in tumors was identified by genetic, molecular, epigenetic as well as behavioral variations of the cells. But what is the origin of these different tumor cells?

4.1. Cancer Stem Cell Theories

The transformation of a cell into a tumorigenic cell includes multiple mutations. There are two prominent theories about the origin of cancer cells. One property of at least some cancer cells is the ability to divide. However, do cancer cells acquire this ability or do they arise from a stem cell population that already possesses this ability?
The first theory about the origin of CSCs states that any body cell can become a cancer stem cell by mutation, meaning that already differentiated, somatic cells become tumorigenic. Therefore, an accumulation of mutations is needed in oncogenes (gain of function) or tumor suppressor genes (loss of function), which regulate cell growth, to transform somatic cells into CSCs [87,88,89]. These mutations occur through replication errors or DNA damage, combined with a missing or incorrect repair mechanism [90]. However, many critics on this conventional theory emerged in the last decades, because of the unlikelihood that various mutations occur in one mature (non-dividing) cell with a limited lifespan. These critics favor the second theory, which is called cancer stem cell theory. This theory is based on the self-renewal ability of stem cells or progenitor cells and states that CSCs arise through oncogenic mutation in stem cells. The remaining of this review will deal with this second cancer stem cell theory, including the influence of CSCs on glioblastoma, their origin in the SVZ as well as possible treatment therapies targeting CSCs.
The idea of stem cells derived CSCs was minted by studies using human leukemia cancer cells, which were transferred into immunodeficient mice [91,92]. When characterizing these cells, the authors found that the cells were quite heterogeneous and only a minor portion had the potential of producing leukemia in mice. This suggests that not all cancer cells but only the slowly dividing stem cells have the potential to reproduce the tumor itself [91,93]. Another study addressed breast cancer cells and described the heterogeneous phenotype of the cells. Only a limited number of cells in the tumor displayed tumorigenic potential which they identified by cell surface markers (CD44+ CD24−/low lineage) [94]. Thus, targeting these cells by cancer therapy would be most promising.
In addition to their tumorigenic properties and extensive proliferative potential, CSCs share various qualities with normal stem cells: (I) The capacity of multipotency, meaning the ability to differentiate into multiple lineages, self-renewal, and the capacity to divide into either new stem cells or into differentiated cells [86]. (II) A low self-renewal rate and rare occurrence (only one in a million cells) [92,93,95,96]. (III) A strict control by their microenvironment to regulate the balance between proliferation and cell death [97,98]. (IV) The usage of similar signaling pathways [85].

4.2. Neural Brain Tumor Propagating Cells

The hypothesis of CSCs can also be extended to brain tumors, here referred to as brain tumor propagating cells (BTPCs), however, with some minor deviations. As discussed above, stem cells are scarce in the adult brain and can only grow in protective stem cells niches, including the hippocampus and the SVZ [31,32]. These NSCs already possess the ability to proliferate and thus they could transform more easily and rapidly into BTPCs than any other post-mitotic neural cell in the brain [99]. After certain variations, neural precursor cells could become BTPCs (Figure 2). However, other than their offspring, NSCs normally do not leave their neurogenic niches. One hypothesis would be that BTPCs originate from a mutation or deregulation that enables the NSCs to migrate and leave the niche. This exit and a subsequent dysregulation of the stem cell might result in unpredictable proliferation and thus tumorigenesis [100,101,102,103].
Like other stem cells, NSCs have the ability of self-renewal and by asymmetric cell division, another NSC and differentiated daughter cell is created. To ensure that the number of NSCs remains constant, proliferation is strictly regulated by intrinsic and extrinsic factors from proliferative cells themselves, neighboring cells, as well as the adjacent blood vessels (reviewed in [104]). Disruption/mutation of these regulatory mechanisms could result in creation of a BTPC. However, brain tumors are not found in the SVZ itself, but rather in the cerebral cortex of adult brains. Thus, besides various tumorigenic mutations, the mutated neural precursor cells need a signal to migrate into other brain regions.
Tumorigenic mutations in migratory SVZ neural precursor cells would be of advantage, because of their ability to migrate over long distances [61,62]. However, in the adult human brain, migration of neuroblasts to the OB is limited, which raises the question if these cells still have the ability to migrate [33,34,67]. Brain injuries, like ischemia, increase the number of neural precursors and some NSCs migrate into the lesion site to increase regeneration, suggesting that the NSCs themselves are able to migrate [105,106]. Furthermore, BTPCs could migrate along tumor microtubes, which have been described as ultra-long membrane protrusions combining BTPC niches scattered in the brain [107,108]. Those BTPC niches are similar to NSC niches with a specific protective microenvironment composed of specific cell types. Based on their location within the tumor and their composition, they are mostly described as peri-vascular, necrotic and hypoxic niches [101,109]. BTPCs in peri-vascular niches are closely located to endothelial cells and express vascular endothelial growth factor VEGF, a well-known growth factor which regulates angiogenesis [110]. Those endothelial cells include capillaries, venules, lymph vessels or arterioles, whereby in case of arterioles, the niche can be directly specified as peri-arteriolar niche [111]. In hypoxic niches, the level of hypoxia-inducible factor HIF is increased, which can increase pro-angiogenic growth factors [112,113]. Most recently, a combined concept was presented, comprising all BTPC niches to one integral hypoxic peri-arteriolar niche model (reviewed in [114]).
Over the years, various studies on mouse models of brain tumors suggested that the cell of origin for brain tumors are NSCs located in the neurogenic niches [6,115,116,117,118]. These NSCs further proliferate in other brain regions, forming a tumor, which consist of BTPCs and more differentiated cancer cells. In the following the cancer stem cell theory is discussed in the context of glioblastoma.

5. The Cell of Origin in Glioblastoma

Over the years of glioblastoma research, various theories emerged about the cell of origin, including tissue-specific stem cells like NSCs or committed precursor cells, like astrocyte precursor cells (APCs) and oligodendrocyte precursor cells (OPCs) [119,120].

5.1. NSCs as the Cells of Origin in Glioblastoma

Gradually, several groups identified that glioblastomas are organized hierarchically and contain tumorigenic cells, which could correspond to BTPCs [121,122,123,124,125] with the basic BTPC and NSC characteristics like multi-lineage potency, self-renewal capacity and the ability of tumor initiation and migration [122,123,126,127,128]. Another evidence for an involvement of undifferentiated BTPCs in GB was provided by discovering that the cells share molecular pathways with NSCs, e.g., the Notch receptor activation. Notch 1 along with its signaling partners is important for NSC maintenance [129] and glioma cell survival [130]. Furthermore, BTPCs and NSCs both express characteristic genes, which can be found in human tumor tissue [125], including Nestin [131,132,133,134,135,136], CD133 [133,137], Sox [132], Musashi1 [136,138], Olig1/2 [139,140], Ras [141], Akt [141], and GFAP [142,143]. However, these genes are not exclusively expressed in NSCs and BTPCs, but can be found in various other cell types, e.g., GFAP is expressed in astrocytes and radial glial cells. Also, the inactivation of certain tumor suppression genes is similar, e.g., PTEN, which increases proliferation of NSCs as well as of tumor cells [144].
One of the neurogenic niches, the SVZ, came into the focus of BTPC research. Already in the early 21st century, GB was linked to the NSCs of the SVZ. Ignatova et al. isolated cancer cells from human glioblastoma, took them in culture and discovered that these tumors contain neurosphere-forming cells [121]. Those BTPC niches Comparing the structural organization of these tumor-derived spheres to those derived from adult human SVZ highlighted a similar hierarchy, concerning the general organization and an outward gradient of differentiation [145]. This gradient in neurospheres is believed to be important for maintaining the stem cell pool. However, for a long time, data from mouse models could not provide a direct link between SVZ NSCs and GB BTPCs.
One novel factor involved in both GB and SVZ NSCs is the extracellular matrix protein epidermal growth factor-like protein 7 (EGFL7), which was previously described to be secreted by endothelial cells as well as NSCs in the SVZ [146,147,148,149,150]. Loss of EGFL7 increased proliferation of NSCs and decreased cell differentiation in the SVZ, mainly via Notch signaling [146,148]. Additionally, the interaction of Notch 1 and EGFR increased survival of GB cells [147] via promoting angiogenesis [15]. The influence of EGFL7 in angiogenesis [149] also plays an important role in GB [150]. EGFL7 is secreted by glioma blood vessels and increases angiogenesis in GB, suggesting inhibition of EGFL7 as a possible treatment strategy [150].
Analyses of the survival of patients with GB revealed that it is crucial if the tumor has a direct connection to the SVZ [151,152]. Patients bearing tumors which were in contact to the SVZ had a shortened survival period [151,153,154,155], but the size of these tumors was not altered [156] and thereby did not influence the survival time [152]. Potentially, a contact to the SVZ might provide the tumor with a pool of tumorigenic stem cells, therefore increasing the growth and invasiveness of the tumor. Proteomic analyses by Gollapalli et al. revealed significant alterations in acute phase proteins (e.g., proteins-hemopexin, alpha-1-antichymotrypsin), lipid carrying proteins (e.g., apolipoprotein A1), cytoskeletal proteins (e.g., brain acid soluble protein 1, thymosin beta 4), lipid binding proteins, chaperones, and regulating proteins between tumors with SVZ contact and without [157]. However, gene expression studies showed that there is no evidence that tumors with contact to the SVZ are more likely to be stem-cell derived than tumors without any contact to the SVZ [153]. Following this, two scenarios were suggested: [I] Tumors without contact are not derived from SVZ NSCs or [II] NSCs from the SVZ migrate into other brain regions and only then start to form a tumor. Various studies are pointing to the second scenario [8,158], although it could not be explained why the survival of the patients is altered.
The origin of the tumor can influence the malignance and survival of the patients. A study in mice showed the induction of GB originating in NSCs of the SVZ increases tumor development and resistance to cancer drugs, giving a hind to the aggressiveness and the malignance of glioblastoma [159]. Furthermore, chronic inflammation in the SVZ could facilitate and accelerate mutations in NSCs and thus contribute to the transformation into BTPCs (reviewed in [99]). The SVZ represents a unique inflammatory niche, which differs significantly from other brain regions, due to close contact to the cerebrospinal fluid (CSF).
Recently, Lee et al. showed direct genetic evidence that brain BTPCs arise from cells of the SVZ using human patients and glioblastoma mouse models [8]. The authors performed single-cell sequencing and laser microdissection on tumor free SVZ tissue and tumor tissue and found a clonal relationship of driver mutations between the SVZ and GB-derived tissue. This is the first proof that SVZ NSCs indeed are the cell of origin in glioblastoma. Furthermore, the authors could show that astrocyte-like NSCs leave the SVZ and migrate into distant brain regions to form gliomas. Restoration of homeobox protein EMX2 expression, which is normally decreased in cancer tissue, but is also expressed in NSCs of the SVZ, affects the cell cycle in glioblastoma, leading to a cell cycle arrest and cell death [160]. A recent study of the tumorigenic potential in different stages of neuronal progenitor cells showed that the tumorigenic potential decreases with an increasing lineage restriction, hinting that mutations likely occur in early stages of neurogenesis, e.g., in NSCs [161]. Taken together, these latest reports on the involvement of SVZ derived NSCs in GB are a huge step to discover new therapeutic targets for the treatment of GB patients.

5.2. Committed Precursor Cells as the Cell of Origin in Glioblastoma

Another theory about the cell of origin states that glioblastoma BTPCs arise from committed precursor cells, like APCs or OPCs. Proliferating multipotent NSCs can create committed neuronal precursor cells that may further divide and differentiate into mature neurons and committed glial precursor cells (GPCs) that can further differentiate into OPCs to generate mature oligodendrocytes and into APCs to generate mature astrocytes [162,163].
For a long time, astrocytes were believed to be the only proliferating cells in the adult brain [164] und thus, were subject in glioblastoma research, also because of high GFAP levels in glioma tissue [165]. However, these mature astrocytes would need to dedifferentiate to become tumorigenic, which is possible but unlikely [166,167]. Furthermore, GFAP is also expressed by radial glial cells [168] and NSCs of the adult SVZ [55].
Lindberg et al. and Hide et al. introduced OPCs as the cell of origin by specific mouse models to study OPCs in glioblastoma development, like the MADM-based lineage tracing model to mutate sporadically p53/Nf1 [120,169]. Hide et al. suggested that transformation of both OPCs and NSCs could lead to formation of BTPCs with tumorigenic properties [120]. Additionally, Liu et al. noted that it is important to analyze premalignant stages of tumors to identify the cell of origin, because the tumor cell could acquire plasticity and veil their origin [119]. Further, they demonstrated that OPCs, but not NSC or any other NSC-derived lineage, show aberrant growth prior to malignancy. In OPCs are some overlapping marker expressions, like PDGFRα and NG2, which are involved in development of OPCs and are altered in glioma [13,170,171]. OPCs might form a stem cell niche at the tumor border, increasing chemo-radioresistance and promoting recurrence [171].
Taken together, the current data suggests that BTPCs might develop from various stem or progenitor cells, which needs to be considered when developing treatment strategies. NSCs, ASCs, OPCs, and GPCs might all be the cell of origin, which lead to the development of GB. However, most recent studies using state of the art techniques clearly point to an involvement of NSCs in GB [8].

6. Brain Tumor Propagating Cells as Target for Glioblastoma Treatment

Traditional glioma treatments include chemotherapy, radiation, and surgery. However, these approaches are limited due to multiple factors like the development of therapy resistance coming with a decreasing sensitivity of tumor cells towards ionizing radiation, the unspecific targeting of both cancer and healthy brain cells, as well as the recurrence of the tumor after treatment. Recent advances suggest that the failure of therapies might be caused by the assumption that the tumor consists of a homogenous group of cells. In fact, the tumor is distinguishable into many subgroups of cells varying on genetics, epigenetics and phenotype as well as their capabilities to replicate and migrate. Thus, elimination of BTPCs is a promising alternative to specifically target the cell of origin of the tumor, which raises the possibility to prevent tumor proliferation, tumor cell differentiation and BTPC regeneration.

6.1. Complications in Glioblastoma Therapy

Due to specific BTPC characteristics, like slow cell division rate, self-renewal properties, high capacity for DNA repairing and high expression of drug transporters, the identification and targeting of this cell population represents a challenge to this day. Moreover, BTPCs are capable of developing resistance mechanisms in multiple ways complicating conventional drug efficacies. High expression of ATP-binding cassette drug transporters can impede cytotoxic agents to enter the cell [172], resulting in resistance to different chemotherapeutic drugs including the commonly used alkylating agent temozolomide [173,174] and increasing the risk of tumor recurrence after the treatment [175]. Besides the chemo-resistance, BTPCs are capable of developing a radio-resistance by an increase in the activation of the DNA repair machinery, which is promoted by the expression of stem cell marker CD133 [176]. This combined chemo- and radio-resistance hampers a successful treatment and therefore many patients require combinational therapeutic strategies to improve the survival.
Another way how BTPCs escape especially surgery is by forming stem cell niches and using ultra-long membrane protrusions, tumor microtubes, which can be found in various brain tumors and can be used as migration routes for cells located in BTPCs niches scattered in the brain. Disconnecting these tumor microtubes was suggested as a new target for treatment of GB [107,108].
The brain and especially brain tumors are always considered as extremely difficult for treatment, due to the blood–brain barrier (BBB). The BBB normally hinders harmful substances and toxins to enter the brain via different cellular and molecular components as well as divers transport systems. However, the location of the SVZ at the border to the lateral ventricle introduces a new aspect to the system, the CSF, which is secreted by the choroid plexus, forming the blood–cerebrospinal fluid barrier (CSFB). This barrier is functionally distinct and is not as tight as the BBB; mostly all non-cellular substances can enter the CSF [177]. It raises an opportunity for future drugs to enter the brain via the CSF and thereby increase the range of possible treatment molecules. This new approach was already successfully applied to medulloblastoma, another brain tumor type [178].

6.2. BTPCs as Target in Glioblstoma

Over the years, different approaches to target BTPCs were developed and tested (Figure 3), mostly in combination with traditional treatments and with more or less successful outcome [179,180]. There are three major therapeutic strategies for targeting BTPCs: (I) Targeting specific cell surface markers, signaling pathways or BTPCs microenvironment, (II) the induction of apoptosis or stem cell differentiation, as well as inhibition of autophagy and (III) the application of vaccines or epigenetic drugs.

6.2.1. Targeting Specific Cell Surface Markers of Signaling Pathways or the Microenvironment of BTPCs

The most prominent marker to target the cell surface with antibody-drug conjugates is CD133, which is expressed not only in brain tumors [124], but also in other cancer types like lung cancer [181] or colorectal cancer [182]. The expression of CD133, alone or together with proliferation marker Ki67, is correlated to a glioma patients’ prognosis with poor clinical outcome [137,183]. However, only recent studies found a population of BTPCs that does not express CD133 and displays a lower proliferation rate compared to the CD133-positive cell population, with equal tumorigenic characteristics [184]. Thus, targeting CD133-positive cells alone could spare some proliferating BTPCs and thereby increase the reoccurrence rate, but targeting CD133-positive cells could be combined with other treatment strategies.
Targeting aberrant signaling pathways is a common approach in all forms of cancer. As for many cancer cell types, Notch signaling, the Wnt/β-catenin pathway and the PI3K/Akt cascade are among the pathways, which are significantly up-regulated and in the focus for targeting BTPCs in glioblastoma [185,186,187]. As an example, the upregulation of the Notch pathway in glioma plays a significant role in increasing tumorigenesis by promoting proliferation and cell survival. Evidence emerged that it also directly regulates maintenance and cell differentiation of BTPCs [130,188]. Application of γ-secretase inhibitors to impair Notch signaling led to decreased growth of glioma, but also induced both neuronal and astrocytic differentiation in the stem cell niches and thereby decreased the number of CD133-positive BTPCs [189,190].
Another way to target BTPCs is to inhibit or modify the signaling from their microenvironment that sustains the growth of the cells and is necessary for fundamental processes e.g., the regulation of hypoxia and angiogenesis. These processes are closely linked, with angiogenesis being regulated through hypoxia. The hypoxia-inducible factor (HIF) is regulating numerous genes, including vascular endothelial growth factor (VEGF) expression. The overexpression of HIF-1α and HIF-2α have been linked to poor prognosis in many cancer types [191]. HIF-2α-positive cells are in close contact to BTPCs, which indicates that BTPCs might be able to self-regulate their maintenance through communication with surrounding cells and support of angiogenesis [113]. Different agents to target HIF proteins are in development with some being tested in clinical trials [191] and it was also previously shown that bevacizumab, a VEGF neutralizing antibody, could specifically inhibit the proangiogenic effects of BTPCs [192]. A combinational therapy, targeting both hypoxic cancer cells and angiogenesis, may therefore represent a promising strategy in depleting BTPCs.

6.2.2. The Induction of Apoptosis, Autophagy or Stem Cell Differentiation

A further approach to diminish the number of BTPCs and to erase the tumors origin is the induction of apoptosis. Apoptosis includes a complex signaling network and the evasion of this system is crucial for the stem cell survival as well as tumor development. Agents to manipulate apoptotic pathways include the inhibition of tumor necrosis factor related apoptosis inducing ligand (TRAIL) and bortezomib, a proteasome inhibitor, which, given in combination, could trigger stem cell death in glioblastoma [193]. Also, both EGFR inhibitors, AG1478 and gefitinib, did not only decrease cell proliferation, but also induced apoptosis of BTPCs [194].
Autophagy, a process of degradation and recycling of cellular components, has also been studied in glioblastoma BTPCs, especially in the context of drug-therapy resistance [195]. Modulation of the autophagic process provides an opportunity to increase cell death and to interfere with the cell cycle in BTPCs. Impairment of the autophagic flux decreases the cell self-renewal capacity of BTPCs. Furthermore, it was reported that chemotherapy increases autophagy in cancer cells [196] and that combining cytotoxic drugs and autophagy inhibitors (e.g., chloroquine or quinacrine) increase sensitivity of BTPCs [197]. Combination of bevacizumab or temozolomide with autophagy inhibitor chloroquine increased efficiency of the chemotherapy and affected survival of BTPCs [198,199]. Another novel and promising approach is the autophagy inhibitor quinacrine, which is able to cross the blood-brain barrier and increases the responsiveness of BTPCs to temozolomide and thereby death of BTPCs [200].
Stem cell differentiation also displays a promising strategy to limit the number of cells with tumorigenic potential and to produce cells with higher sensitivity to radio- and chemotherapy. Among the anti-cancer drugs affecting cell differentiation are bone morphogenetic proteins (BMP), retinoic acid and histone deacetylase (HDAC) inhibitors [201]. The application of BMP has been shown to block glioma development and to reduce CD133-positive cells, probably by manipulating symmetric cell cycles that generate new stem cells [202].

6.2.3. The Application of Vaccines or Epigenetic Drugs

HDAC inhibitors are belonging to the category of epigenetic drugs and target reversible histone acetylation and methylation. HDAC inhibitor vorinostat is approved by the Food and Drug Administration [203] and is blocking HDAC SirT1 (silencing information regulator), which both increased apoptosis and cell differentiation of CD133-positive glioma cells [204].
Immune cell therapy has a lot of potential and receives increasing attention in the field of cancer treatments. The dendritic cell vaccine ICT-107 consists of six synthetic peptides derived from tumor-associated and on glioma BTPCs overexpressed antigens (AIM-2, MAGE1, TRP-2, gp100, HER2/neu, and IL-13Rα2). It is currently tested in phase III studies and it was already reported that ICT-107 treatment decreased CD133-positive cells and prolonged survival of the patients [205].

6.2.4. Radiation of the SVZ

Mutated tumor cells might reside in the SVZ and other BTPCs niches, forming a reservoir of BTPCs leading to increased tumor recurrence and decreased outcome of GB patients. An approach to overcome distribution of tumor cells as well as radio resistance would be to decrease the number of BTPCs in the SVZ by radiation. Studies in this field however report conflicting results, depending on the dose of radiation and the size of the resected area [128,206,207,208,209,210,211,212,213,214,215].
Besides controversy of the benefit of this treatment, side effects are difficult to estimate. Delivering high doses of radiation to healthy SVZ tissue could decrease quality of life by loss of memory. One recent study on a large group of patients could not find any improvement in survival [216]. This might indicate that BTPCs either do not reside in the SVZ or that there are other reservoirs of BTPCs (BTPC niches) outside the SVZ which lead to a regrow of the tumors.
All these therapeutical approaches are only a small selection of the broad range of agents under development and examination to target BTPCs in GB and how this, especially in combination with classical therapies, may bring benefits in erasing cancer stem cells for a successful treatment of GB patients.

7. Conclusions

Each year, 240,000 cases of glioma are recorded worldwide, with GB accounting for most of these cases. As the most aggressive and malignant form of primary brain cancer, GB leads to thousands of deaths per year. The current standard care of therapy consists of a combination of surgery, radiation, and chemotherapy but the therapeutical success remains poor. This may be due to invasive tumor cells infiltrating neighboring brain regions, intra-tumoral heterogeneity and the capacity of distinct tumor cell populations to develop therapy resistance mechanisms. In this review we summarized various studies, which show that major reasons for the almost inevitable tumor recurrence are BTPCs, which are often spared by conventional therapy. It is suggested that these cells originate from astrocytic-like NSCs in the SVZ with the ability to leave the niche and to migrate over long distances. Through division into either differentiated cells or new BTPCs, they can provide an infinite pool of tumor cells and, additionally, the cells’ chemo- and radio-resistance in turn leads to resistance of recurrent GB to standard therapy. The identification of BTPCs and their origin raises hope to identify new molecular, epigenetic and genetic characteristics for the development of combination therapies to erase tumor cells as well as BTPCs. Current research focuses on multiple ways to target BTPCs including targeting specific cell surface markers, signaling pathways or the microenvironment, inducing either apoptosis or cell differentiation, inhibiting autophagy or applying vaccines or epigenetic drugs. Some of the agents are already investigated in clinical trials, showing promising outcomes. Further research is still needed to verify the latest results and technical advantages will help to discover specific GB BTPC treatment to terminate tumor stem cell proliferation and thereby increase the survival of GB patients. Taken together, this leads towards the development of a cure for this dreadful disease.

Author Contributions

C.A. and S.K. wrote and edited the manuscript and M.H.H.S. edited the manuscript.

Funding

S.K. is funded by the German Research Foundation (DFG) Collaborative Research Center 1292, project TP09.

Acknowledgments

We thank members of the Schmidt lab for all their support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wen, P.Y.; Kesari, S. Malignant gliomas in adults. N. Engl. J. Med. 2008, 359, 492–507. [Google Scholar] [CrossRef]
  2. Greaves, M.; Maley, C.C. Clonal evolution in cancer. Nature 2012, 481, 306–313. [Google Scholar] [CrossRef]
  3. Merlo, L.M.; Pepper, J.W.; Reid, B.J.; Maley, C.C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 2006, 6, 924–935. [Google Scholar] [CrossRef] [PubMed]
  4. Blanpain, C. Tracing the cellular origin of cancer. Nat. Cell Biol. 2013, 15, 126. [Google Scholar] [CrossRef] [PubMed]
  5. Sanai, N.; Tramontin, A.D.; Quinones-Hinojosa, A.; Barbaro, N.M.; Gupta, N.; Kunwar, S.; Lawton, M.T.; McDermott, M.W.; Parsa, A.T.; Manuel-Garcia Verdugo, J.; et al. Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration. Nature 2004, 427, 740–744. [Google Scholar] [CrossRef]
  6. Alcantara Llaguno, S.; Chen, J.; Kwon, C.H.; Jackson, E.L.; Li, Y.; Burns, D.K.; Alvarez-Buylla, A.; Parada, L.F. Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model. Cancer Cell 2009, 15, 45–56. [Google Scholar] [CrossRef] [PubMed]
  7. Tomasetti, C.; Li, L.; Vogelstein, B. Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science 2017, 355, 1330–1334. [Google Scholar] [CrossRef]
  8. Lee, J.H.; Lee, J.E.; Kahng, J.Y.; Kim, S.H.; Park, J.S.; Yoon, S.J.; Um, J.-Y.; Kim, W.K.; Lee, J.-K.; Park, J.; et al. Human glioblastoma arises from subventricular zone cells with low-level driver mutations. Nature 2018, 560, 243–247. [Google Scholar] [CrossRef] [PubMed]
  9. Pearson, J.R.D.; Regad, T. Targeting cellular pathways in glioblastoma multiforme. Signal Transduct. Target. Ther. 2017, 2, 17040. [Google Scholar] [CrossRef] [PubMed]
  10. Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Burger, P.C.; Jouvet, A.; Scheithauer, B.W.; Kleihues, P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007, 114, 97–109. [Google Scholar] [CrossRef]
  11. Larjavaara, S.; Mantyla, R.; Salminen, T.; Haapasalo, H.; Raitanen, J.; Jaaskelainen, J.; Auvinen, A. Incidence of gliomas by anatomic location. Neuro Oncol. 2007, 9, 319–325. [Google Scholar] [CrossRef]
  12. Aldape, K.; Zadeh, G.; Mansouri, S.; Reifenberger, G.; von Deimling, A. Glioblastoma: Pathology, molecular mechanisms and markers. Acta Neuropathol. 2015, 129, 829–848. [Google Scholar] [CrossRef]
  13. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef]
  14. Philpott, C.; Tovell, H.; Frayling, I.M.; Cooper, D.N.; Upadhyaya, M. The NF1 somatic mutational landscape in sporadic human cancers. Hum. Genom. 2017, 11, 13. [Google Scholar] [CrossRef]
  15. Keller, S.; Schmidt, M.H.H. EGFR and EGFRvIII promote angiogenesis and cell invasion in glioblastoma: Combination therapies for an effective treatment. Int. J. Mol. Sci. 2017, 18, 1295. [Google Scholar] [CrossRef]
  16. Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
  17. Ozawa, T.; Brennan, C.W.; Wang, L.; Squatrito, M.; Sasayama, T.; Nakada, M.; Huse, J.T.; Pedraza, A.; Utsuki, S.; Yasui, Y.; et al. PDGFRA gene rearrangements are frequent genetic events in PDGFRA-amplified glioblastomas. Genes Dev. 2010, 24, 2205–2218. [Google Scholar] [CrossRef]
  18. Shapiro, W.R.; Green, S.B.; Burger, P.C.; Mahaley, M.S., Jr.; Selker, R.G.; VanGilder, J.C.; Robertson, J.T.; Ransohoff, J.; Mealey, J., Jr.; Strike, T.A.; et al. Randomized trial of three chemotherapy regimens and two radiotherapy regimens and two radiotherapy regimens in postoperative treatment of malignant glioma. Brain Tumor Cooperative Group Trial 8001. J. Neurosurg. 1989, 71, 1–9. [Google Scholar] [CrossRef]
  19. Duerr, E.M.; Rollbrocker, B.; Hayashi, Y.; Peters, N.; Meyer-Puttlitz, B.; Louis, D.N.; Schramm, J.; Wiestler, O.D.; Parsons, R.; Eng, C.; et al. PTEN mutations in gliomas and glioneuronal tumors. Oncogene 1998, 16, 2259–2264. [Google Scholar] [CrossRef]
  20. Smith, J.S.; Tachibana, I.; Passe, S.M.; Huntley, B.K.; Borell, T.J.; Iturria, N.; O’Fallon, J.R.; Schaefer, P.L.; Scheithauer, B.W.; James, C.D.; et al. PTEN mutation, EGFR amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J. Natl. Cancer Inst. 2001, 93, 1246–1256. [Google Scholar] [CrossRef]
  21. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  22. Holland, E.C. Glioblastoma multiforme: The terminator. Proc. Natl. Acad. Sci. USA 2000, 97, 6242–6244. [Google Scholar] [CrossRef]
  23. Vescovi, A.L.; Galli, R.; Reynolds, B.A. Brain tumour stem cells. Nat. Rev. Cancer 2006, 6, 425. [Google Scholar] [CrossRef] [PubMed]
  24. Caruso, R.; Pesce, A.; Wierzbicki, V. A very rare case report of long-term survival: A patient operated on in 1994 of glioblastoma multiforme and currently in perfect health. Int. J. Surg. Case Rep. 2017, 33, 41–43. [Google Scholar] [CrossRef]
  25. Kumar, A.; Deopujari, C.; Karmarkar, V. A case of glioblastoma multiforme with long term survival: Can we predict the outcome? Turk. Neurosurg. 2012, 22, 378–381. [Google Scholar] [CrossRef] [PubMed]
  26. Joyner, A.L.; Zervas, M. Genetic inducible fate mapping in mouse: Establishing genetic lineages and defining genetic neuroanatomy in the nervous system. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 2006, 235, 2376–2385. [Google Scholar] [CrossRef]
  27. Reynolds, B.A.; Weiss, S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 1992, 255, 1707–1710. [Google Scholar] [CrossRef]
  28. Reynolds, B.A.; Weiss, S. Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. Dev. Biol. 1996, 175, 1–13. [Google Scholar] [CrossRef]
  29. Simitzi, C.; Karali, K.; Ranella, A.; Stratakis, E. Controlling the outgrowth and functions of neural stem cells: The effect of surface topography. Chem. Phys. Chem. 2018, 19, 1143–1163. [Google Scholar] [CrossRef]
  30. Das, S.; Srikanth, M.; Kessler, J.A. Cancer stem cells and glioma. Nat. Clin. Pract. Neurol. 2008, 4, 427. [Google Scholar] [CrossRef] [PubMed]
  31. Lois, C.; Alvarez-Buylla, A. Proliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and glia. Proc. Natl. Acad. Sci. USA 1993, 90, 2074–2077. [Google Scholar] [CrossRef]
  32. Eriksson, P.S.; Perfilieva, E.; Bjork-Eriksson, T.; Alborn, A.M.; Nordborg, C.; Peterson, D.A.; Gage, F.H. Neurogenesis in the adult human hippocampus. Nat. Med. 1998, 4, 1313–1317. [Google Scholar] [CrossRef]
  33. Sanai, N.; Nguyen, T.; Ihrie, R.A.; Mirzadeh, Z.; Tsai, H.H.; Wong, M.; Gupta, N.; Berger, M.S.; Huang, E.; Garcia-Verdugo, J.M.; et al. Corridors of migrating neurons in the human brain and their decline during infancy. Nature 2011, 478, 382–386. [Google Scholar] [CrossRef]
  34. Wang, C.; Liu, F.; Liu, Y.Y.; Zhao, C.H.; You, Y.; Wang, L.; Zhang, J.; Wei, B.; Ma, T.; Zhang, Q.; et al. Identification and characterization of neuroblasts in the subventricular zone and rostral migratory stream of the adult human brain. Cell Res. 2011, 21, 1534–1550. [Google Scholar] [CrossRef]
  35. Roy, N.S.; Wang, S.; Jiang, L.; Kang, J.; Benraiss, A.; Harrison-Restelli, C.; Fraser, R.A.; Couldwell, W.T.; Kawaguchi, A.; Okano, H.; et al. In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus. Nat. Med. 2000, 6, 271–277. [Google Scholar] [CrossRef]
  36. Sorrells, S.F.; Paredes, M.F.; Cebrian-Silla, A.; Sandoval, K.; Qi, D.; Kelley, K.W.; James, D.; Mayer, S.; Chang, J.; Auguste, K.I.; et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature 2018, 555, 377–381. [Google Scholar] [CrossRef]
  37. Boldrini, M.; Fulmore, C.A.; Tartt, A.N.; Simeon, L.R.; Pavlova, I.; Poposka, V.; Rosoklija, G.B.; Stankov, A.; Arango, V.; Dwork, A.J.; et al. Human hippocampal neurogenesis persists throughout aging. Cell Stem Cell 2018, 22, 589–599. [Google Scholar] [CrossRef]
  38. Quiñones-Hinojosa, A.; Sanai, N.; Soriano-Navarro, M.; Gonzalez-Perez, O.; Mirzadeh, Z.; Gil-Perotin, S.; Romero-Rodriguez, R.; Berger, M.S.; Garcia-Verdugo, J.M.; Alvarez-Buylla, A. Cellular composition and cytoarchitecture of the adult human subventricular zone: A niche of neural stem cells. J. Comp. Neurol. 2006, 494, 415–434. [Google Scholar] [CrossRef]
  39. Guerrero-Cazares, H.; Gonzalez-Perez, O.; Soriano-Navarro, M.; Zamora-Berridi, G.; Garcia-Verdugo, J.M.; Quinones-Hinojosa, A. Cytoarchitecture of the lateral ganglionic eminence and rostral extension of the lateral ventricle in the human fetal brain. J. Comp. Neurol. 2011, 519, 1165–1180. [Google Scholar] [CrossRef]
  40. Mitchell, B.D.; Emsley, J.G.; Magavi, S.S.; Arlotta, P.; Macklis, J.D. Constitutive and induced neurogenesis in the adult mammalian brain: Manipulation of endogenous precursors toward CNS repair. Dev. Neurosci. 2004, 26, 101–117. [Google Scholar] [CrossRef]
  41. Gould, E. How widespread is adult neurogenesis in mammals? Nat. Rev. Neurosci. 2007, 8, 481–488. [Google Scholar] [CrossRef]
  42. Fowler, C.D.; Liu, Y.; Wang, Z. Estrogen and adult neurogenesis in the amygdala and hypothalamus. Brain Res. Rev. 2008, 57, 342–351. [Google Scholar] [CrossRef]
  43. Vessal, M.; Darian-Smith, C. Adult neurogenesis occurs in primate sensorimotor cortex following cervical dorsal rhizotomy. J. Neurosci. 2010, 30, 8613–8623. [Google Scholar] [CrossRef]
  44. Parent, J.M. The role of seizure-induced neurogenesis in epileptogenesis and brain repair. Epilepsy Res. 2002, 50, 179–189. [Google Scholar] [CrossRef]
  45. Parent, J.M.; Vexler, Z.S.; Gong, C.; Derugin, N.; Ferriero, D.M. Rat forebrain neurogenesis and striatal neuron replacement after focal stroke. Ann. Neurol. 2002, 52, 802–813. [Google Scholar] [CrossRef]
  46. Hou, S.W.; Wang, Y.Q.; Xu, M.; Shen, D.H.; Wang, J.J.; Huang, F.; Yu, Z.; Sun, F.Y. Functional integration of newly generated neurons into striatum after cerebral ischemia in the adult rat brain. Stroke 2008, 39, 2837–2844. [Google Scholar] [CrossRef]
  47. Danilov, A.I.; Covacu, R.; Moe, M.C.; Langmoen, I.A.; Johansson, C.B.; Olsson, T.; Brundin, L. Neurogenesis in the adult spinal cord in an experimental model of multiple sclerosis. Eur. J. Neurosci. 2006, 23, 394–400. [Google Scholar] [CrossRef]
  48. Chi, L.; Ke, Y.; Luo, C.; Li, B.; Gozal, D.; Kalyanaraman, B.; Liu, R. Motor neuron degeneration promotes neural progenitor cell proliferation, migration, and neurogenesis in the spinal cords of amyotrophic lateral sclerosis mice. Stem Cells 2006, 24, 34–43. [Google Scholar] [CrossRef]
  49. Drapeau, E.; Montaron, M.F.; Aguerre, S.; Abrous, D.N. Learning-induced survival of new neurons depends on the cognitive status of aged rats. J. Neurosci. 2007, 27, 6037–6044. [Google Scholar] [CrossRef]
  50. Pereira, A.C.; Huddleston, D.E.; Brickman, A.M.; Sosunov, A.A.; Hen, R.; McKhann, G.M.; Sloan, R.; Gage, F.H.; Brown, T.R.; Small, S.A. An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc. Natl. Acad. Sci. USA 2007, 104, 5638–5643. [Google Scholar] [CrossRef]
  51. Saxe, M.D.; Battaglia, F.; Wang, J.W.; Malleret, G.; David, D.J.; Monckton, J.E.; Garcia, A.D.; Sofroniew, M.V.; Kandel, E.R.; Santarelli, L.; et al. Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc. Natl. Acad. Sci. USA 2006, 103, 17501–17506. [Google Scholar] [CrossRef] [PubMed]
  52. Aimone, J.B.; Wiles, J.; Gage, F.H. Potential role for adult neurogenesis in the encoding of time in new memories. Nat. Neurosci. 2006, 9, 723–727. [Google Scholar] [CrossRef] [PubMed]
  53. Mu, Y.; Gage, F.H. Adult hippocampal neurogenesis and its role in Alzheimer’s disease. Mol. Neurodegener. 2011, 6, 85. [Google Scholar] [CrossRef]
  54. Hamilton, L.K.; Joppe, S.E.; Cochard, L.M.; Fernandes, K.J. Aging and neurogenesis in the adult forebrain: What we have learned and where we should go from here. Eur. J. Neurosci. 2013, 37, 1978–1986. [Google Scholar] [CrossRef] [PubMed]
  55. Lim, D.A.; Alvarez-Buylla, A. Interaction between astrocytes and adult subventricular zone precursors stimulates neurogenesis. Proc. Natl. Acad. Sci. USA 1999, 96, 7526–7531. [Google Scholar] [CrossRef]
  56. Lim, D.A.; Tramontin, A.D.; Trevejo, J.M.; Herrera, D.G.; Garcia-Verdugo, J.M.; Alvarez-Buylla, A. Noggin antagonizes BMP signaling to create a niche for adult neurogenesis. Neuron 2000, 28, 713–726. [Google Scholar] [CrossRef]
  57. Tong, C.K.; Han, Y.-G.; Shah, J.K.; Obernier, K.; Guinto, C.D.; Alvarez-Buylla, A. Primary cilia are required in a unique subpopulation of neural progenitors. Proc. Natl. Acad. Sci. USA 2014, 111, 12438–12443. [Google Scholar] [CrossRef]
  58. Ihrie, R.A.; Álvarez-Buylla, A. Lake-front property: A unique germinal niche by the lateral ventricles of the adult brain. Neuron 2011, 70, 674–686. [Google Scholar] [CrossRef] [PubMed]
  59. Mirzadeh, Z.; Merkle, F.T.; Soriano-Navarro, M.; Garcia-Verdugo, J.M.; Alvarez-Buylla, A. Neural stem cells confer unique pinwheel architecture to the ventricular surface in neurogenic regions of the adult brain. Cell Stem Cell 2008, 3, 265–278. [Google Scholar] [CrossRef]
  60. Fuentealba, L.C.; Obernier, K.; Alvarez-Buylla, A. Adult neural stem cells bridge their niche. Cell Stem Cell 2012, 10, 698–708. [Google Scholar] [CrossRef]
  61. Kornack, D.R.; Rakic, P. The generation, migration, and differentiation of olfactory neurons in the adult primate brain. Proc. Natl. Acad. Sci. USA 2001, 98, 4752–4757. [Google Scholar] [CrossRef]
  62. Pencea, V.; Bingaman, K.D.; Freedman, L.J.; Luskin, M.B. Neurogenesis in the subventricular zone and rostral migratory stream of the neonatal and adult primate forebrain. Exp. Neurol. 2001, 172, 1–16. [Google Scholar] [CrossRef]
  63. Altman, J. Autoradiographic and histological studies of postnatal neurogenesis. IV. Cell proliferation and migration in the anterior forebrain, with special reference to persisting neurogenesis in the olfactory bulb. J. Comp. Neurol. 1969, 137, 433–457. [Google Scholar] [CrossRef]
  64. Lois, C.; Alvarez-Buylla, A. Long-distance neuronal migration in the adult mammalian brain. Science 1994, 264, 1145–1148. [Google Scholar] [CrossRef]
  65. Ayuso-Sacido, A.; Roy, N.S.; Schwartz, T.H.; Greenfield, J.P.; Boockvar, J.A. Long-term expansion of adult human brain subventricular zone precursors. Neurosurgery 2008, 62, 223–231. [Google Scholar] [CrossRef]
  66. Roy, N.S.; Benraiss, A.; Wang, S.; Fraser, R.A.; Goodman, R.; Couldwell, W.T.; Nedergaard, M.; Kawaguchi, A.; Okano, H.; Goldman, S.A. Promoter-targeted selection and isolation of neural progenitor cells from the adult human ventricular zone. J. Neurosci. Res. 2000, 59, 321–331. [Google Scholar] [CrossRef]
  67. Curtis, M.A.; Kam, M.; Nannmark, U.; Anderson, M.F.; Axell, M.Z.; Wikkelso, C.; Holtas, S.; van Roon-Mom, W.M.; Bjork-Eriksson, T.; Nordborg, C.; et al. Human neuroblasts migrate to the olfactory bulb via a lateral ventricular extension. Science 2007, 315, 1243–1249. [Google Scholar] [CrossRef] [PubMed]
  68. Sawamoto, K.; Hirota, Y.; Alfaro-Cervello, C.; Soriano-Navarro, M.; He, X.; Hayakawa-Yano, Y.; Yamada, M.; Hikishima, K.; Tabata, H.; Iwanami, A.; et al. Cellular composition and organization of the subventricular zone and rostral migratory stream in the adult and neonatal common marmoset brain. J. Comp. Neurol. 2011, 519, 690–713. [Google Scholar] [CrossRef]
  69. Haydon, P.G. Glia: Listening and talking to the synapse. Nat. Rev. Neurosci. 2001, 2, 185. [Google Scholar] [CrossRef]
  70. Pascual, O.; Haydon, P.G. Synaptic inhibition mediated by glia. Neuron 2003, 40, 873–875. [Google Scholar] [CrossRef]
  71. Kam, M.; Curtis, M.A.; McGlashan, S.R.; Connor, B.; Nannmark, U.; Faull, R.L. The cellular composition and morphological organization of the rostral migratory stream in the adult human brain. J. Chem. Neuroanat. 2009, 37, 196–205. [Google Scholar] [CrossRef]
  72. Bergmann, O.; Liebl, J.; Bernard, S.; Alkass, K.; Yeung, M.S.; Steier, P.; Kutschera, W.; Johnson, L.; Landen, M.; Druid, H.; et al. The age of olfactory bulb neurons in humans. Neuron 2012, 74, 634–639. [Google Scholar] [CrossRef]
  73. McKenzie, I.A.; Ohayon, D.; Li, H.; de Faria, J.P.; Emery, B.; Tohyama, K.; Richardson, W.D. Motor skill learning requires active central myelination. Science 2014, 346, 318–322. [Google Scholar] [CrossRef]
  74. Ernst, A.; Alkass, K.; Bernard, S.; Salehpour, M.; Perl, S.; Tisdale, J.; Possnert, G.; Druid, H.; Frisen, J. Neurogenesis in the striatum of the adult human brain. Cell 2014, 156, 1072–1083. [Google Scholar] [CrossRef]
  75. Serwanski, D.R.; Rasmussen, A.L.; Brunquell, C.B.; Perkins, S.S.; Nishiyama, A. Sequential contribution of parenchymal and neural stem cell-derived oligodendrocyte precursor cells toward remyelination. Neuroglia 2018, 1, 8. [Google Scholar] [CrossRef]
  76. Faiz, M.; Sachewsky, N.; Gascon, S.; Bang, K.W.; Morshead, C.M.; Nagy, A. Adult neural stem cells from the subventricular zone give rise to reactive astrocytes in the cortex after stroke. Cell Stem Cell 2015, 17, 624–634. [Google Scholar] [CrossRef]
  77. Xing, Y.L.; Roth, P.T.; Stratton, J.A.; Chuang, B.H.; Danne, J.; Ellis, S.L.; Ng, S.W.; Kilpatrick, T.J.; Merson, T.D. Adult neural precursor cells from the subventricular zone contribute significantly to oligodendrocyte regeneration and remyelination. J. Neurosci. 2014, 34, 14128–14146. [Google Scholar] [CrossRef]
  78. Klingener, M.; Chavali, M.; Singh, J.; McMillan, N.; Coomes, A.; Dempsey, P.J.; Chen, E.I.; Aguirre, A. N-cadherin promotes recruitment and migration of neural progenitor cells from the SVZ neural stem cell niche into demyelinated lesions. J. Neurosci. 2014, 34, 9590–9606. [Google Scholar] [CrossRef]
  79. Obernier, K.; Tong, C.K.; Alvarez-Buylla, A. Restricted nature of adult neural stem cells: Re-evaluation of their potential for brain repair. Front. Neurosci. 2014, 8, 162. [Google Scholar] [CrossRef]
  80. Parent, J.M. Injury-induced neurogenesis in the adult mammalian brain. Neurosci. A Rev. J. Bringing Neurobiol. Neurol. Psychiatry 2003, 9, 261–272. [Google Scholar] [CrossRef]
  81. Van den Berge, S.A.; van Strien, M.E.; Hol, E.M. Resident adult neural stem cells in Parkinson’s disease—The brain’s own repair system? Eur. J. Pharmacol. 2013, 719, 117–127. [Google Scholar] [CrossRef]
  82. Nait-Oumesmar, B.; Picard-Riera, N.; Kerninon, C.; Baron-Van Evercooren, A. The role of SVZ-derived neural precursors in demyelinating diseases: From animal models to multiple sclerosis. J. Neurol. Sci. 2008, 265, 26–31. [Google Scholar] [CrossRef]
  83. Fainstein, N.; Dan-Goor, N.; Ben-Hur, T. Resident brain neural precursor cells develop age-dependent loss of therapeutic functions in Alzheimer’s mice. Neurobiol. Aging 2018, 72, 40–52. [Google Scholar] [CrossRef]
  84. Chen, L.; Qiu, R.; Li, L.; He, D.; Lv, H.; Wu, X.; Gu, N. The role of exogenous neural stem cells transplantation in cerebral ischemic stroke. J. Biomed. Nanotechnol. 2014, 10, 3219–3230. [Google Scholar] [CrossRef]
  85. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef]
  86. Galderisi, U.; Cipollaro, M.; Giordano, A. Stem cells and brain cancer. Cell Death Differ. 2005, 13, 5. [Google Scholar] [CrossRef]
  87. Rodenhuis, S. Ras and human tumors. Semin. Cancer Biol. 1992, 3, 241–247. [Google Scholar]
  88. Alt, F.W.; Kellems, R.E.; Bertino, J.R.; Schimke, R.T. Selective multiplication of dihydrofolate reductase genes in methotrexate-resistant variants of cultured murine cells. J. Biol. Chem. 1978, 253, 1357–1370. [Google Scholar]
  89. Croce, C.M. Oncogenes and cancer. N. Engl. J. Med. 2008, 358, 502–511. [Google Scholar] [CrossRef]
  90. Martincorena, I.; Campbell, P.J. Somatic mutation in cancer and normal cells. Science 2015, 349, 1483–1489. [Google Scholar] [CrossRef]
  91. Hope, K.J.; Jin, L.; Dick, J.E. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat. Immunol. 2004, 5, 738–743. [Google Scholar] [CrossRef]
  92. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef]
  93. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef]
  94. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef]
  95. Uckun, F.M.; Sather, H.; Reaman, G.; Shuster, J.; Land, V.; Trigg, M.; Gunther, R.; Chelstrom, L.; Bleyer, A.; Gaynon, P.; et al. Leukemic cell growth in SCID mice as a predictor of relapse in high-risk B-lineage acute lymphoblastic leukemia. Blood 1995, 85, 873–878. [Google Scholar]
  96. Papaccio, F.; Paino, F.; Regad, T.; Papaccio, G.; Desiderio, V.; Tirino, V. Concise review: Cancer cells, cancer stem cells, and mesenchymal stem cells: Influence in cancer development. Stem Cells Transl. Med. 2017, 6, 2115–2125. [Google Scholar] [CrossRef]
  97. Marusyk, A.; Polyak, K. Tumor heterogeneity: Causes and consequences. Biochim. Biophys. Acta 2010, 1805, 105–117. [Google Scholar] [CrossRef]
  98. Weigelt, B.; Glas, A.M.; Wessels, L.F.; Witteveen, A.T.; Peterse, J.L.; van’t Veer, L.J. Gene expression profiles of primary breast tumors maintained in distant metastases. Proc. Natl. Acad. Sci. USA 2003, 100, 15901–15905. [Google Scholar] [CrossRef]
  99. Bardella, C.; Al-Shammari, A.R.; Soares, L.; Tomlinson, I.; O’Neill, E.; Szele, F.G. The role of inflammation in subventricular zone cancer. Prog. Neurobiol. 2018, 170, 37–52. [Google Scholar] [CrossRef]
  100. Wodarz, A.; Gonzalez, C. Connecting cancer to the asymmetric division of stem cells. Cell 2006, 124, 1121–1123. [Google Scholar] [CrossRef]
  101. Calabrese, C.; Poppleton, H.; Kocak, M.; Hogg, T.L.; Fuller, C.; Hamner, B.; Oh, E.Y.; Gaber, M.W.; Finklestein, D.; Allen, M.; et al. A perivascular niche for brain tumor stem cells. Cancer Cell 2007, 11, 69–82. [Google Scholar] [CrossRef]
  102. Clarke, M.F.; Fuller, M. Stem cells and cancer: Two faces of eve. Cell 2006, 124, 1111–1115. [Google Scholar] [CrossRef]
  103. Khalifa, J.; Tensaouti, F.; Lusque, A.; Plas, B.; Lotterie, J.A.; Benouaich-Amiel, A.; Uro-Coste, E.; Lubrano, V.; Cohen-Jonathan Moyal, E. Subventricular zones: New key targets for glioblastoma treatment. Radiat. Oncol. 2017, 12, 67. [Google Scholar] [CrossRef]
  104. Homem, C.C.; Repic, M.; Knoblich, J.A. Proliferation control in neural stem and progenitor cells. Nat. Rev. Neurosci. 2015, 16, 647–659. [Google Scholar] [CrossRef]
  105. Alagappan, D.; Lazzarino, D.A.; Felling, R.J.; Balan, M.; Kotenko, S.V.; Levison, S.W. Brain injury expands the numbers of neural stem cells and progenitors in the SVZ by enhancing their responsiveness to EGF. Asn Neuro 2009, 1, e00009. [Google Scholar] [CrossRef]
  106. Capilla-Gonzalez, V.; Lavell, E.; Quinones-Hinojosa, A.; Guerrero-Cazares, H. Regulation of subventricular zone-derived cells migration in the adult brain. Adv. Exp. Med. Biol. 2015, 853, 1–21. [Google Scholar] [CrossRef]
  107. Osswald, M.; Jung, E.; Sahm, F.; Solecki, G.; Venkataramani, V.; Blaes, J.; Weil, S.; Horstmann, H.; Wiestler, B.; Syed, M.; et al. Brain tumour cells interconnect to a functional and resistant network. Nature 2015, 528, 93–98. [Google Scholar] [CrossRef]
  108. Weil, S.; Osswald, M.; Solecki, G.; Grosch, J.; Jung, E.; Lemke, D.; Ratliff, M.; Hänggi, D.; Wick, W.; Winkler, F. Tumor microtubes convey resistance to surgical lesions and chemotherapy in gliomas. Neuro Oncol. 2017, 19, 1316–1326. [Google Scholar] [CrossRef]
  109. Seidel, S.; Garvalov, B.K.; Wirta, V.; von Stechow, L.; Schanzer, A.; Meletis, K.; Wolter, M.; Sommerlad, D.; Henze, A.T.; Nister, M.; et al. A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain A J. Neurol. 2010, 133, 983–995. [Google Scholar] [CrossRef]
  110. Sharma, A.; Shiras, A. Cancer stem cell-vascular endothelial cell interactions in glioblastoma. Biochem. Biophys. Res. Commun. 2016, 473, 688–692. [Google Scholar] [CrossRef]
  111. Hira, V.V.; Ploegmakers, K.J.; Grevers, F.; Verbovsek, U.; Silvestre-Roig, C.; Aronica, E.; Tigchelaar, W.; Turnsek, T.L.; Molenaar, R.J.; Van Noorden, C.J. CD133+ and Nestin+ glioma stem-like cells reside around CD31+ arterioles in niches that express SDF-1alpha, CXCR4, osteopontin and cathepsin K. J. Histochem. Cytochem. 2015, 63, 481–493. [Google Scholar] [CrossRef]
  112. Filatova, A.; Seidel, S.; Bogurcu, N.; Graf, S.; Garvalov, B.K.; Acker, T. Acidosis acts through HSP90 in a PHD/VHL-independent manner to promote HIF function and stem cell maintenance in glioma. Cancer Res. 2016, 76, 5845–5856. [Google Scholar] [CrossRef]
  113. Li, Z.; Bao, S.; Wu, Q.; Wang, H.; Eyler, C.; Sathornsumetee, S.; Shi, Q.; Cao, Y.; Lathia, J.; McLendon, R.E.; et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 2009, 15, 501–513. [Google Scholar] [CrossRef]
  114. Aderetti, D.A.; Hira, V.V.V.; Molenaar, R.J.; van Noorden, C.J.F. The hypoxic peri-arteriolar glioma stem cell niche, an integrated concept of five types of niches in human glioblastoma. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 346–354. [Google Scholar] [CrossRef]
  115. Yang, Z.-J.; Ellis, T.; Markant, S.L.; Read, T.-A.; Kessler, J.D.; Bourboulas, M.; Schüller, U.; Machold, R.; Fishell, G.; Rowitch, D.H.; et al. Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell 2008, 14, 135–145. [Google Scholar] [CrossRef]
  116. Wang, Y.; Yang, J.; Zheng, H.; Tomasek, G.J.; Zhang, P.; McKeever, P.E.; Lee, E.Y.; Zhu, Y. Expression of mutant p53 proteins implicates a lineage relationship between neural stem cells and malignant astrocytic glioma in a murine model. Cancer Cell 2009, 15, 514–526. [Google Scholar] [CrossRef]
  117. Macas, J.; Ku, M.C.; Nern, C.; Xu, Y.; Buhler, H.; Remke, M.; Synowitz, M.; Franz, K.; Seifert, V.; Plate, K.H.; et al. Generation of neuronal progenitor cells in response to tumors in the human brain. Stem Cells 2014, 32, 244–257. [Google Scholar] [CrossRef]
  118. Abou-Kheir, W.G.; Hynes, P.G.; Martin, P.L.; Pierce, R.; Kelly, K. Characterizing the contribution of stem/progenitor cells to tumorigenesis in the Pten-/-TP53-/- prostate cancer model. Stem Cells 2010, 28, 2129–2140. [Google Scholar] [CrossRef]
  119. Liu, C.; Sage, J.C.; Miller, M.R.; Verhaak, R.G.; Hippenmeyer, S.; Vogel, H.; Foreman, O.; Bronson, R.T.; Nishiyama, A.; Luo, L.; et al. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell 2011, 146, 209–221. [Google Scholar] [CrossRef]
  120. Hide, T.; Takezaki, T.; Nakatani, Y.; Nakamura, H.; Kuratsu, J.-I.; Kondo, T. Combination of a Ptgs2 Inhibitor and an epidermal growth factor receptor-signaling inhibitor prevents tumorigenesis of oligodendrocyte lineage-derived glioma-initiating cells. Stem Cells 2011, 29, 590–599. [Google Scholar] [CrossRef]
  121. Ignatova, T.N.; Kukekov, V.G.; Laywell, E.D.; Suslov, O.N.; Vrionis, F.D.; Steindler, D.A. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia 2002, 39, 193–206. [Google Scholar] [CrossRef]
  122. Galli, R.; Binda, E.; Orfanelli, U.; Cipelletti, B.; Gritti, A.; De Vitis, S.; Fiocco, R.; Foroni, C.; Dimeco, F.; Vescovi, A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004, 64, 7011–7021. [Google Scholar] [CrossRef]
  123. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef]
  124. Singh, S.K.; Clarke, I.D.; Terasaki, M.; Bonn, V.E.; Hawkins, C.; Squire, J.; Dirks, P.B. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003, 63, 5821–5828. [Google Scholar]
  125. Hemmati, H.D.; Nakano, I.; Lazareff, J.A.; Masterman-Smith, M.; Geschwind, D.H.; Bronner-Fraser, M.; Kornblum, H.I. Cancerous stem cells can arise from pediatric brain tumors. Proc. Natl. Acad. Sci. USA 2003, 100, 15178–15183. [Google Scholar] [CrossRef]
  126. Kroonen, J.; Nassen, J.; Boulanger, Y.G.; Provenzano, F.; Capraro, V.; Bours, V.; Martin, D.; Deprez, M.; Robe, P.; Rogister, B. Human glioblastoma-initiating cells invade specifically the subventricular zones and olfactory bulbs of mice after striatal injection. Int. J. Cancer 2011, 129, 574–585. [Google Scholar] [CrossRef]
  127. Lee, J.; Kotliarova, S.; Kotliarov, Y.; Li, A.; Su, Q.; Donin, N.M.; Pastorino, S.; Purow, B.W.; Christopher, N.; Zhang, W.; et al. 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 Cell 2006, 9, 391–403. [Google Scholar] [CrossRef]
  128. Gupta, T.; Nair, V.; Jalali, R. Stem cell niche irradiation in glioblastoma: Providing a ray of hope? CNS Oncol. 2014, 3, 367–376. [Google Scholar] [CrossRef]
  129. Androutsellis-Theotokis, A.; Leker, R.R.; Soldner, F.; Hoeppner, D.J.; Ravin, R.; Poser, S.W.; Rueger, M.A.; Bae, S.K.; Kittappa, R.; McKay, R.D. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 2006, 442, 823–826. [Google Scholar] [CrossRef]
  130. Purow, B.W.; Haque, R.M.; Noel, M.W.; Su, Q.; Burdick, M.J.; Lee, J.; Sundaresan, T.; Pastorino, S.; Park, J.K.; Mikolaenko, I.; et al. Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res. 2005, 65, 2353–2363. [Google Scholar] [CrossRef]
  131. Neradil, J.; Veselska, R. Nestin as a marker of cancer stem cells. Cancer Sci. 2015, 106, 803–811. [Google Scholar] [CrossRef]
  132. Feng, W.; Liu, S.; Zhu, R.; Li, B.; Zhu, Z.; Yang, J.; Song, C. SOX10 induced Nestin expression regulates cancer stem cell properties of TNBC cells. Biochem. Biophys. Res. Commun. 2017, 485, 522–528. [Google Scholar] [CrossRef]
  133. Wu, B.; Sun, C.; Feng, F.; Ge, M.; Xia, L. Do relevant markers of cancer stem cells CD133 and nestin indicate a poor prognosis in glioma patients? A systematic review and meta-analysis. J. Exp. Clin. Cancer Res. 2015, 34, 44. [Google Scholar] [CrossRef]
  134. Shih, A.H.; Holland, E.C. Notch signaling enhances nestin expression in gliomas. Neoplasia 2006, 8, 1072–1082. [Google Scholar] [CrossRef]
  135. Dahlstrand, J.; Collins, V.P.; Lendahl, U. Expression of the class VI intermediate filament nestin in human central nervous system tumors. Cancer Res. 1992, 52, 5334–5341. [Google Scholar]
  136. Strojnik, T.; Rosland, G.V.; Sakariassen, P.O.; Kavalar, R.; Lah, T. Neural stem cell markers, nestin and musashi proteins, in the progression of human glioma: Correlation of nestin with prognosis of patient survival. Surg. Neurol. 2007, 68, 133–143. [Google Scholar] [CrossRef]
  137. Pallini, R.; Ricci-Vitiani, L.; Banna, G.L.; Signore, M.; Lombardi, D.; Todaro, M.; Stassi, G.; Martini, M.; Maira, G.; Larocca, L.M.; et al. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin. Cancer Res. 2008, 14, 8205–8212. [Google Scholar] [CrossRef]
  138. Toda, M.; Iizuka, Y.; Yu, W.; Imai, T.; Ikeda, E.; Yoshida, K.; Kawase, T.; Kawakami, Y.; Okano, H.; Uyemura, K. Expression of the neural RNA-binding protein Musashi1 in human gliomas. Glia 2001, 34, 1–7. [Google Scholar] [CrossRef]
  139. Bouvier, C.; Bartoli, C.; Aguirre-Cruz, L.; Virard, I.; Colin, C.; Fernandez, C.; Gouvernet, J.; Figarella-Branger, D. Shared oligodendrocyte lineage gene expression in gliomas and oligodendrocyte progenitor cells. J. Neurosurg. 2003, 99, 344–350. [Google Scholar] [CrossRef]
  140. Riemenschneider, M.J.; Koy, T.H.; Reifenberger, G. Expression of oligodendrocyte lineage genes in oligodendroglial and astrocytic gliomas. Acta Neuropathol. 2004, 107, 277–282. [Google Scholar] [CrossRef]
  141. Holland, E.C. A mouse model for glioma: Biology, pathology, and therapeutic opportunities. Toxicol. Pathol. 2000, 28, 171–177. [Google Scholar] [CrossRef]
  142. Maunoury, R.; Courdi, A.; Vedrenne, C.; Constans, J.P. Immunocytochemical localization of the GFAP in heterotransplanted human gliomas (author’s transl). Neuro Chir. 1978, 24, 221–226. [Google Scholar]
  143. Kimura, T.; Budka, H.; Soler-Federsppiel, S. An immunocytochemical comparison of the glia-associated proteins glial fibrillary acidic protein (GFAP) and S-100 protein (S100P) in human brain tumors. Clin. Neuropathol. 1986, 5, 21–27. [Google Scholar]
  144. Groszer, M.; Erickson, R.; Scripture-Adams, D.D.; Lesche, R.; Trumpp, A.; Zack, J.A.; Kornblum, H.I.; Liu, X.; Wu, H. Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo. Science 2001, 294, 2186–2189. [Google Scholar] [CrossRef]
  145. Vik-Mo, E.O.; Sandberg, C.; Joel, M.; Stangeland, B.; Watanabe, Y.; Mackay-Sim, A.; Moe, M.C.; Murrell, W.; Langmoen, I.A. A comparative study of the structural organization of spheres derived from the adult human subventricular zone and glioblastoma biopsies. Exp. Cell Res. 2011, 317, 1049–1059. [Google Scholar] [CrossRef]
  146. Bicker, F.; Vasic, V.; Horta, G.; Ortega, F.; Nolte, H.; Kavyanifar, A.; Keller, S.; Stankovic, N.D.; Harter, P.N.; Benedito, R.; et al. Neurovascular EGFL7 regulates adult neurogenesis in the subventricular zone and thereby affects olfactory perception. Nat. Commun. 2017, 8, 15922. [Google Scholar] [CrossRef]
  147. Fassl, A.; Tagscherer, K.E.; Richter, J.; Berriel Diaz, M.; Alcantara Llaguno, S.R.; Campos, B.; Kopitz, J.; Herold-Mende, C.; Herzig, S.; Schmidt, M.H.H.; et al. Notch1 signaling promotes survival of glioblastoma cells via EGFR-mediated induction of anti-apoptotic Mcl-1. Oncogene 2012, 31, 4698–4708. [Google Scholar] [CrossRef]
  148. Schmidt, M.H.H.; Bicker, F.; Nikolic, I.; Meister, J.; Babuke, T.; Picuric, S.; Muller-Esterl, W.; Plate, K.H.; Dikic, I. Epidermal growth factor-like domain 7 (EGFL7) modulates Notch signalling and affects neural stem cell renewal. Nat. Cell Biol. 2009, 11, 873–880. [Google Scholar] [CrossRef]
  149. Nikolić, I.; Stanković, N.D.; Bicker, F.; Meister, J.; Braun, H.; Awwad, K.; Baumgart, J.; Simon, K.; Thal, S.C.; Patra, C.; et al. EGFL7 ligates αvβ3 integrin to enhance vessel formation. Blood 2013, 121, 3041–3050. [Google Scholar] [CrossRef]
  150. Dudvarski Stankovic, N.; Bicker, F.; Keller, S.; Jones, D.T.; Harter, P.N.; Kienzle, A.; Gillmann, C.; Arnold, P.; Golebiewska, A.; Keunen, O.; et al. EGFL7 enhances surface expression of integrin alpha5beta1 to promote angiogenesis in malignant brain tumors. EMBO Mol. Med. 2018, 10, e8420. [Google Scholar] [CrossRef]
  151. Lim, D.A.; Cha, S.; Mayo, M.C.; Chen, M.H.; Keles, E.; VandenBerg, S.; Berger, M.S. Relationship of glioblastoma multiforme to neural stem cell regions predicts invasive and multifocal tumor phenotype. Neuro Oncol. 2007, 9, 424–429. [Google Scholar] [CrossRef]
  152. Pope, W.B.; Sayre, J.; Perlina, A.; Villablanca, J.P.; Mischel, P.S.; Cloughesy, T.F. MR imaging correlates of survival in patients with high-grade gliomas. Am. J. Neuroradiol. 2005, 26, 2466–2474. [Google Scholar]
  153. Kappadakunnel, M.; Eskin, A.; Dong, J.; Nelson, S.F.; Mischel, P.S.; Liau, L.M.; Ngheimphu, P.; Lai, A.; Cloughesy, T.F.; Goldin, J.; et al. Stem cell associated gene expression in glioblastoma multiforme: Relationship to survival and the subventricular zone. J. Neuro Oncol. 2010, 96, 359–367. [Google Scholar] [CrossRef]
  154. Chaichana, K.L.; McGirt, M.J.; Frazier, J.; Attenello, F.; Guerrero-Cazares, H.; Quinones-Hinojosa, A. Relationship of glioblastoma multiforme to the lateral ventricles predicts survival following tumor resection. J. Neurooncol. 2008, 89, 219–224. [Google Scholar] [CrossRef]
  155. Mistry, A.M.; Dewan, M.C.; White-Dzuro, G.A.; Brinson, P.R.; Weaver, K.D.; Thompson, R.C.; Ihrie, R.A.; Chambless, L.B. Decreased survival in glioblastomas is specific to contact with the ventricular-subventricular zone, not subgranular zone or corpus callosum. J. Neurooncol. 2017, 132, 341–349. [Google Scholar] [CrossRef]
  156. Barami, K.; Sloan, A.E.; Rojiani, A.; Schell, M.J.; Staller, A.; Brem, S. Relationship of gliomas to the ventricular walls. J. Clin. Neurosci. 2009, 16, 195–201. [Google Scholar] [CrossRef]
  157. Gollapalli, K.; Ghantasala, S.; Kumar, S.; Srivastava, R.; Rapole, S.; Moiyadi, A.; Epari, S.; Srivastava, S. Subventricular zone involvement in Glioblastoma—A proteomic evaluation and clinicoradiological correlation. Sci. Rep. 2017, 7, 1449. [Google Scholar] [CrossRef]
  158. Zhu, Y.; Guignard, F.; Zhao, D.; Liu, L.; Burns, D.K.; Mason, R.P.; Messing, A.; Parada, L.F. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer Cell 2005, 8, 119–130. [Google Scholar] [CrossRef]
  159. Jiang, Y.; Marinescu, V.D.; Xie, Y.; Jarvius, M.; Maturi, N.P.; Haglund, C.; Olofsson, S.; Lindberg, N.; Olofsson, T.; Leijonmarck, C.; et al. Glioblastoma cell malignancy and drug sensitivity are affected by the cell of origin. Cell Rep. 2017, 18, 977–990. [Google Scholar] [CrossRef]
  160. Monnier, A.; Boniface, R.; Bouvet, R.; Etcheverry, A.; Aubry, M.; Avril, T.; Quillien, V.; Chevet, E.; Mosser, J. The expression of EMX2 lead to cell cycle arrest in glioblastoma cell line. BMC Cancer 2018, 18, 1213. [Google Scholar] [CrossRef]
  161. Llaguno, S.A.; Sun, D.; Pedraza, A.M.; Vera, E.; Wang, Z.; Burns, D.K.; Parada, L.F. Cell-of-origin susceptibility to glioblastoma formation declines with neural lineage restriction. Nat. Neurosci. 2019, 22, 545–555. [Google Scholar] [CrossRef]
  162. Rowitch, D.H.; Kriegstein, A.R. Developmental genetics of vertebrate glial—Cell specification. Nature 2010, 468, 214. [Google Scholar] [CrossRef]
  163. Jiang, Y.; Uhrbom, L. On the origin of glioma. Upsala J. Med. Sci. 2012, 117, 113–121. [Google Scholar] [CrossRef]
  164. Cavanagh, J.B. The proliferation of astrocytes around a needle wound in the rat brain. J. Anat. 1970, 106, 471–487. [Google Scholar]
  165. Jones, T.R.; Bigner, S.H.; Schold, S.C., Jr.; Eng, L.F.; Bigner, D.D. Anaplastic human gliomas grown in athymic mice. Morphology and glial fibrillary acidic protein expression. Am. J. Pathol. 1981, 105, 316–327. [Google Scholar]
  166. Sharif, A.; Legendre, P.; Prevot, V.; Allet, C.; Romao, L.; Studler, J.M.; Chneiweiss, H.; Junier, M.P. Transforming growth factor alpha promotes sequential conversion of mature astrocytes into neural progenitors and stem cells. Oncogene 2007, 26, 2695–2706. [Google Scholar] [CrossRef]
  167. Dufour, C.; Cadusseau, J.; Varlet, P.; Surena, A.L.; de Faria, G.P.; Dias-Morais, A.; Auger, N.; Leonard, N.; Daudigeos, E.; Dantas-Barbosa, C.; et al. Astrocytes reverted to a neural progenitor-like state with transforming growth factor alpha are sensitized to cancerous transformation. Stem Cells 2009, 27, 2373–2382. [Google Scholar] [CrossRef]
  168. Alves, J.A.; Barone, P.; Engelender, S.; Froes, M.M.; Menezes, J.R. Initial stages of radial glia astrocytic transformation in the early postnatal anterior subventricular zone. J. Neurobiol. 2002, 52, 251–265. [Google Scholar] [CrossRef]
  169. Lindberg, N.; Kastemar, M.; Olofsson, T.; Smits, A.; Uhrbom, L. Oligodendrocyte progenitor cells can act as cell of origin for experimental glioma. Oncogene 2009, 28, 2266–2275. [Google Scholar] [CrossRef]
  170. Shoshan, Y.; Nishiyama, A.; Chang, A.; Mork, S.; Barnett, G.H.; Cowell, J.K.; Trapp, B.D.; Staugaitis, S.M. Expression of oligodendrocyte progenitor cell antigens by gliomas: Implications for the histogenesis of brain tumors. Proc. Natl. Acad. Sci. USA 1999, 96, 10361–10366. [Google Scholar] [CrossRef]
  171. Parsons, D.W.; Jones, S.; Zhang, X.; Lin, J.C.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Siu, I.M.; Gallia, G.L.; et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008, 321, 1807–1812. [Google Scholar] [CrossRef]
  172. Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275. [Google Scholar] [CrossRef]
  173. Liu, G.; Yuan, X.; Zeng, Z.; Tunici, P.; Ng, H.; Abdulkadir, I.R.; Lu, L.; Irvin, D.; Black, K.L.; John, S.Y. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol. Cancer 2006, 5, 67. [Google Scholar] [CrossRef]
  174. Eramo, A.; Ricci-Vitiani, L.; Zeuner, A.; Pallini, R.; Lotti, F.; Sette, G.; Pilozzi, E.; Larocca, L.M.; Peschle, C.; De Maria, R. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ. 2006, 13, 1238. [Google Scholar] [CrossRef]
  175. Chen, J.; Li, Y.; Yu, T.-S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522. [Google Scholar] [CrossRef]
  176. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756. [Google Scholar] [CrossRef]
  177. Pardridge, W.M. Drug transport in brain via the cerebrospinal fluid. Fluids Barriers CNS 2011, 8, 7. [Google Scholar] [CrossRef]
  178. Glantz, M.; Kesari, S.; Recht, L.; Fleischhack, G.; Van Horn, A. Understanding the origins of gliomas and developing novel therapies: Cerebrospinal fluid and subventricular zone interplay. Semin. Oncol. 2009, 36, S17–S24. [Google Scholar] [CrossRef]
  179. Dragu, D.L.; Necula, L.G.; Bleotu, C.; Diaconu, C.C.; Chivu-Economescu, M. Therapies targeting cancer stem cells: Current trends and future challenges. World J. Stem Cells 2015, 7, 1185. [Google Scholar]
  180. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124. [Google Scholar] [CrossRef]
  181. Eramo, A.; Lotti, F.; Sette, G.; Pilozzi, E.; Biffoni, M.; Di Virgilio, A.; Conticello, C.; Ruco, L.; Peschle, C.; De Maria, R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008, 15, 504. [Google Scholar] [CrossRef]
  182. Ricci-Vitiani, L.; Lombardi, D.G.; Pilozzi, E.; Biffoni, M.; Todaro, M.; Peschle, C.; De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007, 445, 111. [Google Scholar] [CrossRef]
  183. Zeppernick, F.; Ahmadi, R.; Campos, B.; Dictus, C.; Helmke, B.M.; Becker, N.; Lichter, P.; Unterberg, A.; Radlwimmer, B.; Herold-Mende, C.C. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin. Cancer Res. 2008, 14, 123–129. [Google Scholar] [CrossRef]
  184. Beier, D.; Hau, P.; Proescholdt, M.; Lohmeier, A.; Wischhusen, J.; Oefner, P.J.; Aigner, L.; Brawanski, A.; Bogdahn, U.; Beier, C.P. CD133+ and CD133− glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007, 67, 4010–4015. [Google Scholar] [CrossRef]
  185. Bleau, A.-M.; Hambardzumyan, D.; Ozawa, T.; Fomchenko, E.I.; Huse, J.T.; Brennan, C.W.; Holland, E.C. PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell 2009, 4, 226–235. [Google Scholar] [CrossRef]
  186. Wang, J.; Wakeman, T.P.; Lathia, J.D.; Hjelmeland, A.B.; Wang, X.F.; White, R.R.; Rich, J.N.; Sullenger, B.A. Notch promotes radioresistance of glioma stem cells. Stem Cells 2010, 28, 17–28. [Google Scholar] [CrossRef]
  187. Rheinbay, E.; Suvà, M.L.; Gillespie, S.M.; Wakimoto, H.; Patel, A.P.; Shahid, M.; Oksuz, O.; Rabkin, S.D.; Martuza, R.L.; Rivera, M.N. An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 2013, 3, 1567–1579. [Google Scholar] [CrossRef]
  188. Stockhausen, M.-T.; Kristoffersen, K.; Poulsen, H.S. The functional role of Notch signaling in human gliomas. Neuro Oncol. 2009, 12, 199–211. [Google Scholar] [CrossRef]
  189. Saito, N.; Fu, J.; Zheng, S.; Yao, J.; Wang, S.; Liu, D.D.; Yuan, Y.; Sulman, E.P.; Lang, F.F.; Colman, H. A high notch pathway activation predicts response to γ secretase inhibitors in proneural subtype of glioma tumor-initiating cells. Stem Cells 2014, 32, 301–312. [Google Scholar] [CrossRef]
  190. Fan, X.; Khaki, L.; Zhu, T.S.; Soules, M.E.; Talsma, C.E.; Gul, N.; Koh, C.; Zhang, J.; Li, Y.M.; Maciaczyk, J. NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells 2010, 28, 5–16. [Google Scholar] [CrossRef]
  191. Semenza, G.L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 2003, 3, 721. [Google Scholar] [CrossRef]
  192. Bao, S.; Wu, Q.; Sathornsumetee, S.; Hao, Y.; Li, Z.; Hjelmeland, A.B.; Shi, Q.; McLendon, R.E.; Bigner, D.D.; Rich, J.N. Stem cell–like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006, 66, 7843–7848. [Google Scholar] [CrossRef]
  193. Unterkircher, T.; Cristofanon, S.; Vellanki, S.H.K.; Nonnenmacher, L.; Karpel-Massler, G.; Wirtz, C.; Debatin, K.-M.; Fulda, S. Bortezomib primes glioblastoma including glioblastoma stem cells for TRAIL by increasing tBid stability and mitochondrial apoptosis. Clin. Cancer Res. 2011, 7, 4019–4030. [Google Scholar] [CrossRef]
  194. Soeda, A.; Inagaki, A.; Oka, N.; Ikegame, Y.; Aoki, H.; Yoshimura, S.-I.; Nakashima, S.; Kunisada, T.; Iwama, T. Epidermal growth factor plays a crucial role in mitogenic regulation of human brain tumor stem cells. J. Biol. Chem. 2008, 283, 10958–10966. [Google Scholar] [CrossRef]
  195. Nazio, F.; Bordi, M.; Cianfanelli, V.; Locatelli, F.; Cecconi, F. Autophagy and cancer stem cells: Molecular mechanisms and therapeutic applications. Cell Death Differ. 2019, 26, 690–702. [Google Scholar] [CrossRef]
  196. Sui, X.; Chen, R.; Wang, Z.; Huang, Z.; Kong, N.; Zhang, M.; Han, W.; Lou, F.; Yang, J.; Zhang, Q.; et al. Autophagy and chemotherapy resistance: A promising therapeutic target for cancer treatment. Cell Death Dis. 2013, 4, e838. [Google Scholar] [CrossRef]
  197. Sun, R.; Shen, S.; Zhang, Y.-J.; Xu, C.-F.; Cao, Z.-T.; Wen, L.-P.; Wang, J. Nanoparticle-facilitated autophagy inhibition promotes the efficacy of chemotherapeutics against breast cancer stem cells. Biomaterials 2016, 103, 44–55. [Google Scholar] [CrossRef]
  198. Encouse, B.G.; Hee-Yeon, C.; Ardeshir, J.; Florence, M.H.; Stan, G.L.; Axel, H.S.; Thomas, C.C. Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg. Focus 2014, 37, e12. [Google Scholar] [CrossRef]
  199. Huang, H.; Song, J.; Liu, Z.; Pan, L.; Xu, G. Autophagy activation promotes bevacizumab resistance in glioblastoma by suppressing Akt/mTOR signaling pathway. Oncol. Lett. 2018, 15, 1487–1494. [Google Scholar] [CrossRef]
  200. Buccarelli, M.; Marconi, M.; Pacioni, S.; De Pascalis, I.; D’Alessandris, Q.G.; Martini, M.; Ascione, B.; Malorni, W.; Larocca, L.M.; Pallini, R.; et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018, 9, 841. [Google Scholar] [CrossRef]
  201. Massard, C.; Deutsch, E.; Soria, J. Tumour stem cell-targeted treatment: Elimination or differentiation. Ann. Oncol. 2006, 17, 1620–1624. [Google Scholar] [CrossRef]
  202. Piccirillo, S.; Reynolds, B.A.; Zanetti, N.; Lamorte, G.; Binda, E.; Broggi, G.; Brem, H.; Olivi, A.; Dimeco, F.; Vescovi, A.L. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 2006, 444, 761. [Google Scholar] [CrossRef]
  203. Bezecny, P. Histone deacetylase inhibitors in glioblastoma: Pre-clinical and clinical experience. Med. Oncol. 2014, 31, 985. [Google Scholar] [CrossRef]
  204. Chang, C.-J.; Hsu, C.-C.; Yung, M.-C.; Chen, K.-Y.; Tzao, C.; Wu, W.-F.; Chou, H.-Y.; Lee, Y.-Y.; Lu, K.-H.; Chiou, S.-H. Enhanced radiosensitivity and radiation-induced apoptosis in glioma CD133-positive cells by knockdown of SirT1 expression. Biochem. Biophys. Res. Commun. 2009, 380, 236–242. [Google Scholar] [CrossRef]
  205. Phuphanich, S.; Wheeler, C.J.; Rudnick, J.D.; Mazer, M.; Wang, H.; Nuno, M.A.; Richardson, J.E.; Fan, X.; Ji, J.; Chu, R.M. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol. Immunother. 2013, 62, 125–135. [Google Scholar] [CrossRef]
  206. Elicin, O.; Inac, E.; Uzel, E.K.; Karacam, S.; Uzel, O.E. Relationship between survival and increased radiation dose to subventricular zone in glioblastoma is controversial. J. Neuro-Oncol. 2014, 118, 413–419. [Google Scholar] [CrossRef]
  207. Chua, M.; Kusumawidjaja, G.; Gan, P.; Tan, D.H.Y.; Chua, E.T.; Tham, C.K.; Wong, F.Y. Dose-escalated intensity modulated radiotherapy (IMRT) and increased radiation doses to subventricular zones (SVZ) in treatment outcomes of patients with glioblastoma multiforme (GBM). J. Clin. Oncol. 2014, 32, e13031. [Google Scholar] [CrossRef]
  208. Malik, M.; Akram, K.S.; Joseph, D.; Valiyaveettil, D.; Ahmed, S.F. Prospective study of irradiation of potential stem cell niches in glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, S111. [Google Scholar] [CrossRef]
  209. Nourallah, B.; Digpal, R.; Jena, R.; Watts, C. Irradiating the subventricular zone in glioblastoma patients: Is there a case for a clinical trial? Clin. Oncol. 2017, 29, 26–33. [Google Scholar] [CrossRef]
  210. Adeberg, S.; Harrabi, S.B.; Bougatf, N.; Bernhardt, D.; Mohr, A.; Rieber, J.; Koelsche, C.; Rieken, S.; Debus, J. Do increased doses to stem-cell niches during radiation therapy improve glioblastoma survival? Stem Cells Int. 2016, 2016, 8793462. [Google Scholar] [CrossRef]
  211. Gupta, T.; Nair, V.; Paul, S.N.; Kannan, S.; Moiyadi, A.; Epari, S.; Jalali, R. Can irradiation of potential cancer stem-cell niche in the subventricular zone influence survival in patients with newly diagnosed glioblastoma? J. Neurooncol. 2012, 109, 195–203. [Google Scholar] [CrossRef]
  212. Evers, P.; Lee, P.P.; DeMarco, J.; Agazaryan, N.; Sayre, J.W.; Selch, M.; Pajonk, F. Irradiation of the potential cancer stem cell niches in the adult brain improves progression-free survival of patients with malignant glioma. BMC Cancer 2010, 10, 384. [Google Scholar] [CrossRef]
  213. Lee, P.; Eppinga, W.; Lagerwaard, F.; Cloughesy, T.; Slotman, B.; Nghiemphu, P.L.; Wang, P.C.; Kupelian, P.; Agazaryan, N.; Demarco, J.; et al. Evaluation of high ipsilateral subventricular zone radiation therapy dose in glioblastoma: A pooled analysis. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 609–615. [Google Scholar] [CrossRef]
  214. Chen, L.; Guerrero-Cazares, H.; Ye, X.; Ford, E.; McNutt, T.; Kleinberg, L.; Lim, M.; Chaichana, K.; Quinones-Hinojosa, A.; Redmond, K. Increased subventricular zone radiation dose correlates with survival in glioblastoma patients after gross total resection. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 616–622. [Google Scholar] [CrossRef]
  215. Mathew, B.S.; Kaliyath, S.B.; Krishnan, J.; Bhasi, S. Impact of subventricular zone irradiation on outcome of patients with glioblastoma. J. Cancer Res. Ther. 2018, 14, 1202–1206. [Google Scholar] [CrossRef]
  216. Murchison, S.C.; Wiksyk, B.; Gossman, S.; Jensen, B.; Sayers, D.; Lesperance, M.; Truong, P.T.; Alexander, A. Subventricular zone radiation dose and outcome for glioblastoma treated between 2006 and 2012. Cureus 2018, 10, e3618. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the subventricular zone (SVZ) in a coronal view of the human brain and the adult human neurogenic niche in the sub-ependymal zone showing the cellular composition. The neurogenic niche in the SVZ can be divided into four layers (layer I–IV). The ependymal layer (layer I) separates the SVZ form the lumen of the ventricle by a thin single layer (monolayer) of ependymal cells (grey), which feature several apical microvilli and basal expansions. The ependyma is followed by the hypocellular layer (layer II), mainly containing astrocytic and ependymal processes. The astrocytic ribbon (layer III) primary contains astrocyte-like neural stem cells (NSCs) (red) and neuroblasts (orange), closely followed by the transitional zone (layer IV) which contains myelinated axons (black) and oligodendrocytes (violet). Thereafter the parenchyma begins, which mostly consists of neurons (green) and glia.
Figure 1. Schematic representation of the subventricular zone (SVZ) in a coronal view of the human brain and the adult human neurogenic niche in the sub-ependymal zone showing the cellular composition. The neurogenic niche in the SVZ can be divided into four layers (layer I–IV). The ependymal layer (layer I) separates the SVZ form the lumen of the ventricle by a thin single layer (monolayer) of ependymal cells (grey), which feature several apical microvilli and basal expansions. The ependyma is followed by the hypocellular layer (layer II), mainly containing astrocytic and ependymal processes. The astrocytic ribbon (layer III) primary contains astrocyte-like neural stem cells (NSCs) (red) and neuroblasts (orange), closely followed by the transitional zone (layer IV) which contains myelinated axons (black) and oligodendrocytes (violet). Thereafter the parenchyma begins, which mostly consists of neurons (green) and glia.
Cancers 11 00448 g001
Figure 2. Schematic representation of the origin of brain tumor propagating cells (BTPCs) based on the cancer stem cell theory. In the SVZ of the adult brain, NSCs can be found in the sub-ependymal zone. There are two properties that NSCs need to achieve to become BTPCs. First, the cells need to become tumorigenic, including several mutations, resulting in mutated NSCs. Second these mutated NSCs have to be able to migrate over a long distance into other brain regions, where they form a tumor, which consists of BTPCs and more differentiated cancer cells which originate from the BTPCs themselves.
Figure 2. Schematic representation of the origin of brain tumor propagating cells (BTPCs) based on the cancer stem cell theory. In the SVZ of the adult brain, NSCs can be found in the sub-ependymal zone. There are two properties that NSCs need to achieve to become BTPCs. First, the cells need to become tumorigenic, including several mutations, resulting in mutated NSCs. Second these mutated NSCs have to be able to migrate over a long distance into other brain regions, where they form a tumor, which consists of BTPCs and more differentiated cancer cells which originate from the BTPCs themselves.
Cancers 11 00448 g002
Figure 3. There are three therapeutic strategies to target brain tumor propagating cells (BTPCs). (I) Most common approaches include targeting specific cell surface markers, aberrantly up- or down-regulated signaling pathways or the BTPCs’ microenvironment. (II) The induction of apoptosis or stem cell differentiation as well as the inhibition of autophagy, which aims to reduce the number of BTPCs. (III) Application of vaccines or epigenetic drugs, which are still in development.
Figure 3. There are three therapeutic strategies to target brain tumor propagating cells (BTPCs). (I) Most common approaches include targeting specific cell surface markers, aberrantly up- or down-regulated signaling pathways or the BTPCs’ microenvironment. (II) The induction of apoptosis or stem cell differentiation as well as the inhibition of autophagy, which aims to reduce the number of BTPCs. (III) Application of vaccines or epigenetic drugs, which are still in development.
Cancers 11 00448 g003

Share and Cite

MDPI and ACS Style

Altmann, C.; Keller, S.; Schmidt, M.H.H. The Role of SVZ Stem Cells in Glioblastoma. Cancers 2019, 11, 448. https://doi.org/10.3390/cancers11040448

AMA Style

Altmann C, Keller S, Schmidt MHH. The Role of SVZ Stem Cells in Glioblastoma. Cancers. 2019; 11(4):448. https://doi.org/10.3390/cancers11040448

Chicago/Turabian Style

Altmann, Christine, Stefanie Keller, and Mirko H. H. Schmidt. 2019. "The Role of SVZ Stem Cells in Glioblastoma" Cancers 11, no. 4: 448. https://doi.org/10.3390/cancers11040448

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop