Next Article in Journal
Liquid Biopsy as a Tool for Differentiation of Leiomyomas and Sarcomas of Corpus Uteri
Next Article in Special Issue
Prion Protein in Glioblastoma Multiforme
Previous Article in Journal
Hurdles to Cardioprotection in the Critically Ill
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies

by
Larisa Ryskalin
1,†,
Anderson Gaglione
2,†,
Fiona Limanaqi
1,
Francesca Biagioni
2,
Pietro Familiari
3,
Alessandro Frati
2,
Vincenzo Esposito
2,3 and
Francesco Fornai
1,2,*
1
Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy
2
I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli (IS), Italy
3
Department of Neuroscience, Mental Health and Sense Organs NESMOS, Sapienza University of Rome, 00185 Roma, Italy
*
Author to whom correspondence should be addressed.
These authors equally contributed to the present work.
Int. J. Mol. Sci. 2019, 20(15), 3824; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20153824
Submission received: 10 July 2019 / Revised: 26 July 2019 / Accepted: 3 August 2019 / Published: 5 August 2019
(This article belongs to the Special Issue Molecular Biology of Brain Tumors)

Abstract

:
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor featuring rapid cell proliferation, treatment resistance, and tumor relapse. This is largely due to the coexistence of heterogeneous tumor cell populations with different grades of differentiation, and in particular, to a small subset of tumor cells displaying stem cell-like properties. This is the case of glioma stem cells (GSCs), which possess a powerful self-renewal capacity, low differentiation, along with radio- and chemo-resistance. Molecular pathways that contribute to GBM stemness of GSCs include mTOR, Notch, Hedgehog, and Wnt/β-catenin. Remarkably, among the common biochemical effects that arise from alterations in these pathways, autophagy suppression may be key in promoting GSCs self-renewal, proliferation, and pluripotency maintenance. In fact, besides being a well-known downstream event of mTOR hyper-activation, autophagy downregulation is also bound to the effects of aberrantly activated Notch, Hedgehog, and Wnt/β-catenin pathways in GBM. As a major orchestrator of protein degradation and turnover, autophagy modulates proliferation and differentiation of normal neuronal stem cells (NSCs) as well as NSCs niche maintenance, while its failure may contribute to GSCs expansion and maintenance. Thus, in the present review we discuss the role of autophagy in GSCs metabolism and phenotype in relationship with dysregulations of a variety of NSCs controlling pathways, which may provide novel insights into GBM neurobiology.

1. Introduction

Gliomas are the most prevalent and lethal intracranial tumors in adults, accounting for approximately 36% of primary brain tumors [1]. Arising from glial cells, gliomas represent about 80% of malignant tumors of the central nervous system (CNS). Among these, astrocytomas including glioblastoma, constitute the largest subgroup.
Glioblastoma multiforme (GBM), a WHO grade IV malignant diffuse glioma, is the most frequent primary brain tumor in adults [2,3]. With a median overall survival of 14 months after diagnosis, this high-grade astrocytoma remains the most aggressive and lethal CNS tumor [4]. To date, conventional therapies provide only a slight improvement in the survival and quality of life of GBM patients [5]. Despite standard treatments with radiotherapy plus adjuvant chemotherapy, GBM frequently recurs with lower response rates to subsequent therapeutic approaches. Thus, advances in understanding GBM biology are urgently needed.
As suggested by the term “multiforme”, GBM is characterized by a marked intra-tumoral heterogeneity at both cellular and molecular levels. Among the multiple signaling pathways that have been implicated in GBM progression, the PTEN/PI3K/Akt/mTOR axis holds center stage being involved in tumor cell growth, proliferation, and metabolism [6]. The PTEN/PI3K/Akt/mTOR pathway is aberrantly activated in a variety of human cancers, including gliomas and GBM [7,8,9,10]. In detail, mutations in the tumor suppressor gene PTEN are reported in approximately 80% of GBM [11]. This leads to increased activation of the downstream effector molecule mTOR, which in turn sustains cell metabolism by promoting protein synthesis while suppressing autophagy, the major protein degradation pathway. A large body of clinical and experimental evidence indicates a key role of PI3K/Akt/mTOR hyper-activation in GBM biology [11,12,13,14,15,16,17]. An association between the PI3K/Akt/mTOR signaling and tumor malignancy is confirmed by studies documenting an upregulation of pAKT, pmTOR, and p-p70S6K in high-grade gliomas (grades III and IV) compared with low-grade gliomas (grades I and II) [18]. Again, the expression and phosphorylation of mTOR are associated with a worse prognosis in GBM [19]. In line with the evidence obtained in human GBM samples and cell lines, constitutive activation of mTOR in murine orthotopic xenograft models contributes to formation, growth, and progression of malignant glioma, thus recapitulating the main features of human GBM [12,20,21].
Among the various mechanisms through which PTEN/PI3K/Akt/mTOR hyper-activation sustains glioma cells metabolism, autophagy suppression plays a seminal role. As a proof of concept, the basal activity of autophagy is very low in astrocytomas including GBM from both patients and experimental models, while rescuing autophagy through mTOR inhibition is associated with a reduction of malignant glioma cells growth, proliferation, and invasion in vitro and in vivo [22,23,24,25,26,27,28,29,30,31,32].
Remarkably, mTOR hyper-activation and autophagy suppression are both implicated in a crucial standpoint of GBM pathophysiology, that is, maintaining the oncogenic properties of malignant glioma by promoting the growth and maintenance of glioma stem-like cells (GSCs). As it occurs in different hematopoietic and solid-tumors, GBM harbors a fraction of cancer stem cells known as GSCs, which are endowed with key features of normal neural stem cells (NSCs) of the adult brain, such as sustained self-renewal and proliferation [33]. GSCs are thought to be the driving force of GBM malignant phenotype since they are able to differentiate into phenotypically heterogeneous tumorigenic cancer cells and to establish and recapitulate a whole tumor upon intracranial transplantation [34]. Apart from contributing to GBM cellular heterogeneity, GSCs possess an increased therapeutic resistance, thus promoting tumor infiltration, treatment failure, and relapse [35,36,37,38,39].
mTOR and autophagy pathways are strongly related to the metabolism of both NSCs and GSCs [40,41,42,43]. The balance between mTOR activity and autophagy is seminal to modulate stem-cell niche homeostasis as well as quiescence, self-renewal, and differentiation of normal NSCs [44,45,46], while mTOR hyper-activation and autophagy impairment may occlude GSCs differentiation, thus sustaining the maintenance and expansion of the tumor stem cell niche [26,27,33,47,48,49]. Constitutive activation of the mTOR signaling and autophagy failure contribute to proliferation and pluripotency of GSCs [50,51,52,53,54]. On the other hand, mTOR inhibition and autophagy induction contribute to reducing stem cell-like properties, promoting differentiation, and restraining cell migration and invasion potential of GSCs [32,49,55,56]. In this scenario, dysregulations of mTOR and autophagy machinery do intermingle with a myriad of molecular pathways to sustain GSCs proliferation, GBM aggressiveness, and treatment resistance. This is the case of brain micro-environmental factors, which contribute to altering GSCs metabolism by acting on the mTOR pathway. Again, besides mTOR, autophagy is bound to several molecular pathways that regulate NSCs growth and differentiation and that are altered in GSCs and GBM. This is the case of Wnt/β-catenin, Notch, and Hedgehog pathways [57,58,59]. The present review aims to dissect those autophagy-related, mTOR-dependent and -independent biochemical pathways that characterize cancer stem cells in glioblastoma. Disclosing the mechanisms by which autophagy impairment may sustain GSCs self-renewal, proliferation, and resistance to therapies might provide novel insights into the neurobiology of GBM, and hopefully, contribute to the development of new therapeutic strategies.

2. Identification and Targeting of Cancer Stem Cells in Glioblastoma

The adult human brain possesses self-renewing and proliferative NSCs, which reside within restricted germinal regions, namely the subependymal ventricular zone (SVZ), the subgranular zone (SGZ) of the dentate gyrus of the hippocampus, and the subcortical white matter [60,61,62]. The SVZ of cornu temporalis of the lateral ventricle is one of the most active germinal regions within the adult human brain [63]. In fact, the SVZ continuously generates newborn differentiated neurons, thus being crucial in sustaining postnatal neurogenesis and maintaining the neurogenic niche of the mature brain. In the past 20 years, the identification of brain cancer stem cells (CSCs) as a therapeutic target has greatly improved the comprehension of the molecular pathways that are implicated in the pathophysiology of high-grade gliomas.
CSCs share core biological properties of normal NSCs, such as the potential of self-renewing and maintaining proliferation. In particular, GBM contains a subpopulation of CSCs with enhanced, long-term, self-renewal ability [33,34,64]. This is the case of GSCs residing in perivascular niches within the SVZ and the dentate gyrus of the hippocampus [65,66]. These cells potentially give rise to highly proliferative tumor cells, thus constituting a tumorigenic bulk within the healthy brain parenchyma (Figure 1). To date, it is still unclear whether GSCs originate from NSCs or undifferentiated neural/glial cells transform into CSCs; in any case, GSCs are considered to drive neoplastic transformation [67,68,69].
Fervent research has been carried out aimed at identifying specific subpopulations of GSCs that harbor tumor-initiating potential. Early in vivo studies demonstrated that a subpopulation of tumor cells expressing CD133 (Prominin-1) antigen was capable of tumor initiation when implanted into the adult NOD-SCID (non-obese diabetic, severe combined immunodeficient) mouse brains [34]. However, subsequent studies demonstrated that even CD133-negative glioma cells retain the ability to induce tumors in vivo [70,71]. Apart from the cell surface antigen CD133, which is classically associated with GSCs, these brain tumor cells populations express additional stem cell markers that are classically used to identify normal NSCs, such as Nestin, Bmi-1, and Musashi [72,73]. Other stem cells markers include CD15/SSEA-1, CD44, integrin α6, L1CAM, and A2B5 [74,75,76,77,78,79,80,81]. Moreover, several transcriptional factors are highly expressed in subgroups of GSCs, such as NANOG, SOX2, STAT3, OCT-4, and c-Myc [82,83,84,85]. These transcriptional factors regulate cancer stem cell properties, thereby contributing to CSCs self-renewal and pluripotency. Remarkably, their expression is under the control of mTOR via the activation of the PI3K/Akt pathway.
Nevertheless, none of these markers when considered alone is sufficient to confer stem cell-like properties to cancer cells. Again, due to the marked genetic and phenotypic heterogeneity, GSCs cannot be identified with a unique marker. Thus, the combined detection of different CSCs markers is needed to identify GSCs. For instance, Nestin, a type VI intermediate filament protein observed in NSCs, is frequently co-expressed along with other GSCs markers, such as CD133 or SOX2, and it is essential to confirm cell stemness [86,87].
Nonetheless, so far there is no specific biomarker being specific for the identification and isolation of CSCs in GBM. This stresses the need for a better understanding of the molecular mechanisms contributing to the invasive phenotype of GBM. Recent research points at mTOR signaling hyper-activation and subsequent autophagy suppression as key events implicated in GBM stem cell maintenance, tumor propagation, as well as treatment resistance, which will be dealt with in the following sections.

3. mTOR Function in Glioblastoma Cancer Stem Cells

The mammalian Target Of Rapamycin (mTOR) is a master regulator of cell growth, proliferation, and metabolism [88]. In particular, this kinase is a major effector of the PI3K/Akt pathway, which in turn is stimulated by several upstream environmental inputs, mainly nutrient availability and cellular energy levels. Upon ligands binding (i.e., growth factors, amino acids) to their respective transmembrane receptor, the upstream PI3K kinase phosphorylates Akt, which in turn activates the mTOR complex. Once activated, mTOR phosphorylates two major downstream effector molecules, namely p70S6K and 4E-BP1, which ultimately promote protein synthesis [88]. Whereas amino acids are conveyed through the PI3K/Akt/mTOR pathway to stimulate protein synthesis, they also inhibit autophagy, thus repressing protein degradation. In fact, insulin or nutrient-related signals activate mTOR, which in turn suppresses early steps in the biogenesis of autophagosomes by phosphorylating the ULK1 complex [89]. Conversely, nutrient depletion or administration of the lipophilic macrolide rapamycin inhibits mTOR activity, thereby stimulating the ULK1/ATG13/FIP200 complex formation, which is required to initiate autophagy [90]. Additionally, mTOR can indirectly control the autophagy pathway by inhibiting the transcription factor EB (TFEB), which regulates several lysosomal-related genes [91,92].
Over the last decade, aberrancies of mTOR signaling have been reported in several types of solid tumors, especially CNS tumors, where the constitutive hyper-activation of the PI3K/Akt/mTOR pathway represents one of the major contributors of tumor initiation and progression [16,93]. In particular, PI3K/Akt dysregulations may arise from different genetic alterations, encompassing mutations in upstream oncogenes and/or tumor suppressor genes, mutations in mTOR complex, or the mTOR gene itself [94,95].
In keeping with a role in cell proliferation, several studies demonstrated that mTOR is essential in CNS development and neural progenitor homeostasis. During brain development, mTOR dynamically regulates NSCs self-renewal and differentiation [63,96,97,98]. In fact, normal NSCs undergo self-renewing divisions to propagate the stem cell pool, but they also produce progenitor cells, which then differentiate into neurons, astrocytes, or oligodendrocytes. Proper activation of mTOR signaling is required for homeostatic regulation of normal NSCs niche. In baseline conditions, mTOR activity is finely tuned to maintain the delicate balance between proliferating and differentiating signals within the stem cell niche, where normal NSCs reside. There is a wealth of evidence relating dysregulated mTOR signaling with alterations in neural progenitor homeostasis [97,98,99,100,101,102]. For instance, mTOR hyper-activation in embryonic NSCs results in enhanced SVZ progeny generation and subsequent premature differentiation and impaired maturation [103]. At the same time, a persistent hyper-proliferation induced by mTOR hyper-activation may lead to the exhaustion of the NSCs pool. Such an effect can be reverted by the administration of the gold-standard mTOR inhibitor rapamycin [104]. Remarkably, the hyper-activation of mTOR signaling in transgenic mice results in a marked expansion of the SVZ stem cell compartment and subsequent glioma development [105]. Thus, it is not surprising that abnormal mTOR activity within NSCs is associated with severe CNS diseases ranging from brain tumors to neurodevelopmental disorders [106,107,108].
In keeping with GBM, mTOR has emerged as a critical cue in the maintenance of GSCs niche. Compared with normal NSCs, GSCs undergo uncontrolled proliferation and impaired differentiation, being a tumorigenic niche [78]. While finely tuned mTOR signaling is essential for normal CNS development, GSCs take advantage of an improper mTOR activity to fuel tumor growth and infiltration. This is also due to changes in the tumor niche micro-environment where GSCs receive proliferation signals that overcome growth-inhibiting ones. In fact, cell components within the brain tumor micro-environment, including endothelial cells, glia or neurons may promote GSCs proliferation by releasing signaling molecules such as mitogens, neurotrophic factors, and neurotransmitters.
It is remarkable that mTOR alterations are bound to several tumor-extrinsic mechanisms that support tumor growth and mediate GBM relapse and infiltration [109]. For instance, the activity-regulated secretion of the synaptic protein Neuroligin-3 (NLGN3) activates the PI3K/mTOR pathway to promote cell proliferation [110]. Similarly, neurotrophic factors released by astrocytes exert a proliferation-promoting effect on glioma cells [111]. In addition, it has been demonstrated that secreted factors released by endothelial cells reinforce GSCs stem-like phenotype through the mTOR pathway [51].
Hyper-activation of mTOR promotes self-renewal, proliferation, and pluripotency of GSCs, thus sustaining the oncogenic properties of malignant gliomas [50,52,53]. Conversely, the inhibition of mTOR with rapamycin counteracts these effects. For instance, the PI3K/Akt/mTOR pathway promotes the expression and activity of the transcription factor SOX2, which is required for the maintenance of GBM stem-like properties [112,113]. Conversely, rapamycin decreases GSCs self-renewal and proliferation through downregulation of SOX2 at both protein and mRNA levels [114]. Similarly, mTOR-dependent activation of the downstream effector HIF-1α enhances GSCs self-renewal and proliferation while maintaining their undifferentiated phenotype. In contrast, rapamycin abrogates these effects, which contributes to counteracting tumorigenesis [115]. Rapamycin also reduces GSCs sphere formation and the expression of GSCs-related markers, namely CD133 and Nestin [48]. This is in line with studies showing that targeting the PI3K/Akt/mTOR pathway represses stem-like cell properties in GBM cells by reducing the expression of other pluripotency-regulating transcription factors, such as NANOG and OCT-4 [116,117,118].
Similar results were documented by Mendiburu-Eliçabe et al. in two GBM patient-derived CSC lines, where rapamycin markedly reduces cell growth rate along with the stemness marker CD133 [119]. Moreover, Sunayama et al. [48] demonstrated that combined treatment with rapamycin and LY294002, a PI3K inhibitor, suppresses stemness while increasing the expression of the neuronal marker βIII-tubulin, which suggests a differentiating effect of dual PI3K/mTOR inhibition [48].
These data are in line with our previous studies demonstrating that rapamycin inhibits GBM cell growth in vitro and in vivo through gene expression changes which promote differentiation of GSCs towards a neuron-like phenotype [16,29,32]. Intriguingly, when administered in vivo to mice bearing GBM xenografts, rapamycin induces almost a total inhibition of tumor growth, which occurs in the absence of apoptotic or necrotic cell death [29]. Rapamycin counteracts GBM growth by suppressing the gene expression of the stemness marker Nestin, while stimulating those related to neuronal differentiation, encompassing both early and post-mitotic mitotic neuronal markers, such as βIII-tubulin, NeuroD, and NeuN [16,32]. Such an effect is associated with a reduced GSCs proliferation, tumorigenicity, and migration.
Recently, mTOR activation emerged as a crucial player in driving GSCs invasiveness, which represents another key factor contributing to GBM recurrence. In fact, hyper-activation of the Akt/mTOR pathway sustains GSCs migration and infiltration within the surrounding healthy brain parenchyma. As proof of concept, mTOR inhibitors suppress GSCs aggressiveness and invasive potential [117]. Moreover, mTOR inhibition downregulates both mRNA, protein levels, and the activity of the matrix metalloproteinases, MMP-9 and MMP-2, which promote tumor invasion through extracellular matrix degradation.
Overall, these findings suggest that mTOR inhibition coupled with radio- and/or chemo-therapy might hold great potential in hindering GBM progression.

4. Autophagy in Glioblastoma Cancer Stem Cells

It is widely recognized that autophagy is altered in GBM. This is not surprising since autophagy is essential in preserving stem cell homeostasis by finely tuning stem cell maintenance and differentiation [40,42]. On the other hand, altered autophagy may contribute to maintaining stem-like properties of GSCs, as well as diminished response to normal differentiation cues. Nonetheless, autophagy emerges as a double-edged sword in GBM development [120,121]. While in healthy cells autophagy acts as a tumor-suppressive mechanism by maintaining cell homeostasis, in cancer cells it may exert either a tumor-promoting or tumor-suppressing effect. Thus, it is still on debate whether autophagy induction or inhibition may represent the most promising approach for future GBM treatments. In addition, when attempting to analyze the autophagy status in GBM one has to face with multiple factors such as tumor stage, micro-environment, and GSCs heterogeneity.
Therefore, in the present section, we discuss the dual role of autophagy in GSCs generation, differentiation, migration/invasion, and treatment resistance, with a special emphasis on the emerging pro-differentiating effect of autophagy induction in GSCs. In fact, rather than extinguishing the GSCs population, current research has focused on forcing these cells to undergo differentiation through autophagy regulation.

4.1. Autophagy Promoting GSCs

A bulk of evidence points to the theory that autophagy inhibition may be beneficial when GBM cells are exposed to stressful stimuli, such as hypoxia, nutrient starvation, or even chemotherapy. In fact, a lack of oxygen and nutrients occur in GBM due to rapid tumor growth and insufficient nutrient supply from the lining vasculature, which may contribute to over-activating protective autophagy while desensitizing cells to chemotherapy. For instance, hypoxia increases the amount of CD133+ GSCs that are more resistant to BNIP3-dependent apoptosis compared to CD133 negative ones [122]. This may be due to BNIP3-dependent autophagy overactivation, which is associated with GSCs proliferation and chemoresistance [123,124]. Conversely, combined administration of autophagy inhibitors and chemotherapy drugs sensitizes GSCs to cytotoxicity [120,121,125].
Again, overactivation of BNIP3-dependent autophagy is also associated with an increased expression of MT1-MPP via JAK/STAT3 in GBM cell lines, which may contribute to the chemoresistant and invasive phenotype of GSCs [126].
Likewise, therapies targeting GSCs-promoting pathways such as Notch induce protective autophagy in glioma neurospheres, which is associated with chemoresistance since this latter is occluded by the combined treatment with autophagy inhibitors [127].
In this scenario, hypoxia-induced autophagy emerges as an adaptive mechanism that promotes cancer stem cell survival by ensuring nutrient and energy supply. In this context, tumor cells upregulate their levels of aerobic glycolysis while reducing mitochondrial energy supply. Thus, as the metabolic process rises, autophagy is recruited to support metabolic reconfiguration [128]. In fact, autophagy provides GCSs with necessary nutrients by recycling macromolecules and organelles. This same mechanism is thought to underlie autophagy-induced resistance to chemotherapeutic agents. In fact, autophagy can degrade dysfunctional organelles and reduce reactive oxygen species (ROS) accumulation, thus protecting the cell from pro-apoptotic stimuli while promoting genome stability. Such a cytoprotective mechanism may also result in the development of multidrug resistance (MDR).
From these studies, it emerges that autophagy inhibitors are most employed as a strategy to enhance chemotherapy-induced cytotoxicity. Nonetheless, recent studies suggest that drug-induced differentiation of GSCs may be a promising approach to eradicate GBM cancer stem cells. As we shall see in the next section, this is strongly bound to autophagy induction, which is key in controlling stem cells properties. As a proof of concept, activating autophagy inhibits GSCs proliferation, self-renewal, tumorigenesis, and reduces stemness, while restoring GSCs differentiation. This, in turn, may be beneficial in counteracting GBM progression.

4.2. Autophagy Combating GSCs

Autophagy induction, mainly through inhibition of the mTOR pathway, exerts anti-proliferative and pro-differentiating effects on glioblastoma stem-like cells (Figure 2). For instance, the antifungal agent itraconazole suppresses GSCs proliferation through induction of autophagy [30]. The in vitro anti-proliferative effect of itraconazole is confirmed in vivo using a subcutaneous GBM xenograft mouse model, where itraconazole significantly decreases GBM growth. Conversely, autophagy blockage, by knocking down either ATG5 or BECN1 with small interfering RNA (siRNA), reverts the anti-proliferative effect of itraconazole. In detail, itraconazole acts as an autophagy inducer by downregulating the sterol carrier protein 2 (SCP2), which decreases cholesterol trafficking towards the plasma membrane to inhibit of the Akt/mTOR pathway [129].
Enhancing autophagy in GSCs produces a variety of effects well beyond the mere inhibition of cell proliferation (Figure 2). In fact, defective autophagy has been implicated in the maintenance of the oncogenic properties of GSCs, such as stem-like properties, self-renewal ability, and pluripotency. Conversely, autophagy induction through mTOR inhibition suppresses GSCs self-renewal and tumorigenicity in vitro and in vivo through promoting autophagy-dependent degradation and inhibition of Notch1 [43]. In fact, upregulation of Notch1 sustains glioma stem cell phenotype, while autophagy induction counteracts such an effect by suppressing Notch1 signaling [43]. Again, metformin-induced Akt/mTOR inhibition impairs GSCs sphere formation, an indirect index of self-renewal, via autophagy induction [130].
Again, autophagy induction suppresses GSCs aggressive phenotype through promoting GSCs differentiation towards a neuron-like phenotype [27,32,131,132]. A failure of autophagy machinery also contributes to GSCs chemo- and radio-resistance [133,134]. Remarkably, rapamycin-induced autophagy prevents GSCs chemo-resistance, while promoting cell differentiation [27]. Likewise, cannabidiol promotes GSCs differentiation through autophagy-dependent upregulation of Aml-1 transcription factors, which is associated with reduced GSCs chemoresistance, proliferation, and clonogenic potential [135]. This is in line with studies showing that inhibition of miR-17, which activates autophagy gene expression, suppresses tumor progression and improves chemo-and radio-therapy [136].
Autophagy is crucial for GSCs invasiveness. In fact, rapamycin-induced autophagy impairs GBM cell migration [32,55]. Stimulation of autophagy reverts GCSs invasiveness by reducing two transcriptional factors belonging to SNAI family, which control epithelial–mesenchymal transition (EMT). Conversely, when autophagy is occluded via silencing Atg5 and Atg7 GBM cell migration and invasion are enhanced [55].
Micro-environmental factors may influence GSCs phenotyope by affecting the autophagy machinery. For instance, CXCR4 ligands favor GSCs chemotactic migration through inhibition of autophagy. In fact, abnormal stimulation of CXCR4 triggers a marked reduction in autophagosomes biogenesis in GBM cancer cell lines while favoring the formation of adhesion complexes to the extracellular matrix [137]. Again, mitogen deprivation was shown to inhibit GSCs self-renewal and survival via the engagement of mTOR-dependent autophagy. On the other hand, blocking autophagy reproduces the effects of ex-vivo administered endothelial-secreted factors, that is promoting GSCs survival and stemness [51].
mTOR-dependent autophagy activation associates with the anti-proliferative effects that are induced by silencing CD164 (endolyn), a key factor implicated in GBM growth [138]. In detail, CD164 is a member of sialomucin family, which plays a role in proliferation, adhesion, and differentiation of hematopoietic stem cells [139] and different types of tumors, including gliomas [140]. When over-expressed, CD164 activates the PI3K/Akt/mTOR pathway to sustain GBM growth [140]. On the other hand, CD164 downregulation reduces glioma cell proliferation, migration, and tumor invasion via depression of the Akt/mTOR pathway and autophagy induction [138].
In recent years, natural compounds besides rapamycin, such as curcumin and resveratrol were used in GBM research owing to their anti-proliferative, anti-migratory, and anti-invasive effects on GSCs [27,116,130,141,142,143]. Most of the beneficial effects of these compounds are due to autophagy stimulation via PI3K/Akt/mTOR pathway inhibition. For instance, treatment with curcumin suppresses GSCs self-renewal and proliferation in vitro and in vivo while inducing GSCs differentiation through activation of the autophagy pathway [131]. In detail, curcumin-treated GSCs feature an up-regulation of the differentiation markers Tuj1, GFAP, Olig2, and βIII-tubulin, and a concomitant downregulation of the stem-like markers CD133 and Nestin. Such a phenotypic switch induced by curcumin is accompanied by a marked stimulation of autophagy in both GSCs cultures and xenograft tumors. This was evidenced by an increase in LC3 immunofluorescent puncta, LC3 immunoblotting, and ultrastructure of autophagosomes. Recent in vitro studies demonstrate that curcumin-induced autophagy suppresses GSCs migration and invasion [143].
Recently, resveratrol, a natural polyphenolic antioxidant found in grapes and red wine, has shown beneficial effects against malignant glioma cells [116]. Growing evidence suggests that the anti-tumor effects of resveratrol consist in a reduction of GSCs proliferation and self-renewal along with promoting GSCs differentiation [144]. Thus, resveratrol contributes to the depletion of GSCs niche and suppression of tumor growth. It is remarkable that these effects are all accompanied by enhanced autophagy, as confirmed by the upregulation of the autophagy proteins Atg5, Beclin-1, and LC3-II and the induction of autophagosome formation [144,145]. Again, similar to curcumin, resveratrol-induced autophagy can restrain the invasive behavior of malignant glioma cells by suppressing GSCs adhesion and migration [142].
Likewise, berberine, an isoquinoline alkaloid isolated from Berberis vulgaris L., enhances autophagy flux in GSCs cells through the inhibition of the AMPK/mTOR/ULK1 pathway. Remarkably, this effect associates with a reduction in the proliferative potential and invasive properties of GBM cells [146].
Again, nigericin, a polyether antibiotic derived from S. hygroscopicus that affects mitochondrial ion transport, was shown to suppresses the proliferation of GBM cells along with the inhibition of GSCs stem-like properties, which associates with marked induction of autophagy [147].

5. The Cross-Talk between Autophagy and Glioblastoma Stem Cells-Controlling Pathways

Apart from the PI3K/Akt/mTOR pathway, autophagy machinery interacts with many proteins and signaling pathways that are implicated in GBM stem-cell properties. These include Wnt/β-catenin, Hedgehog, Notch, Histone deacetylases (HDAC), STAT3, and the de-ubiquitinase ubiquitin carboxyl-terminal esterase L1 (UCHL1). Indeed, rather than acting independently in sustaining GSCs growth and proliferation, these pathways merge to produce a chain of epigenetic, transcriptional, metabolic, and post-translational events where autophagy plays a central role.

5.1. Wnt/β-Catenin, Notch, and Autophagy in GSCs

When Wnt/β-catenin and Notch pathways are aberrantly activated GSCs self-renewal, proliferation, and invasion occurs [148,149,150]. On the other hand, either single or dual inhibition of Wnt/β-catenin and Notch signaling promotes GSCs neuronal differentiation, inhibits their clonogenic potential, decreases radio-resistance and halts tumor growth [148,149,150]. Remarkably, these effects are reproduced by autophagy activators since downregulation of both Notch and Wnt/β-catenin in GBM cells relies on the very same autophagy pathway [43,151,152]. In fact, autophagy activation is seminal to degrade Notch1 and Dishevelled, an activator of Wnt/β-catenin. Autophagy also re-locates β-catenin within the cell by moving the nuclear protein towards the plasma membrane where it associates with N-cadherin to form epithelial-like cell-cell adhesion structures [152]. This is in line with an increase N-cadherins and induction of a molecular switch from a mesenchymal to an epithelial-like phenotype in GBM cellular models upon autophagy stimulation [55].

5.2. UCHL1 and Autophagy in GSCs

UCHL1 de-ubiquitinase is up-regulated in several cancers, including pediatric high-grade gliomas, where it contributes to promoting GSCs self-renewal, transformation, and invasion [153]. The activity of UCHL1 is linked to dysregulations of Akt, mTOR, and Wnt/β-catenin pathways [154,155,156,157] and, remarkably, autophagy suppression [158,159]. For instance, UCHL1 activates Wnt signaling through de-ubiquitination and stabilization of β-catenin [160]. Likewise, UCHL1 enhances mTORC2 stability, thus activating Akt signaling [157]. Aberrant activation of UCHL1 suppresses autophagy either by interacting with LC3 or by inducing PDGFB (platelet-derived growth factor B)-dependent mTOR phosphorylation [158,159]. Silencing UCHL1 in patient-derived glioma cells is associated with decreased GSCs self-renewal, proliferation, and invasion [153]. Remarkably these effects occur along with a 70% reduction in Wnt signaling, and again, PDGFB ranks among the top upstream regulators of the effects induced by UCHL1 silencing [153], suggesting that autophagy may be involved in the anti-proliferative effects of UCHL1 inhibition in GSCs.

5.3. SOX3, Hedgehog, and Autophagy in GSCs

SOX3 is remarkably increased in primary GBM, where it is suggested to promote the malignant behavior of GSCs by enhancing their self-renewal, proliferation, viability, migration, and invasion [161]. SOX3 up-regulation in GBM cells is accompanied by an enhanced activity of the Hedgehog signaling pathway and remarkably, by suppression of autophagy [161]. This is not surprising since a cross-talk exists between Hedgehog and autophagy pathway [162], and dysregulations of one pathway may affect the other to converge in GBM tumorigenesis and GSCs maintenance. For instance, mTOR hyper-activation enhances the expression Hedgehog pathway while amplifying its target genes to promote GSCs regeneration, proliferation, and invasion [163]. On the other hand, the combined inhibition of PI3K/Akt/mTOR and Hedgehog pathways is more effective in suppressing GBM growth, GSCs self-renewal, proliferation, and EMT compared with single pathway inhibition.

5.4. STAT3 and Autophagy in GSCs

Enhanced STAT3 phosphorylation, which is required for GSCs proliferation and maintenance of multi-potency [83], is associated with Notch hyper-activation [164] and autophagy down-regulation in GBM cells [134,145,165]. In fact, activation of JAK2/STAT3 signaling pathway by HMGB-1 (High mobility group box 1) leads to autophagy inhibition [166], while administration of autophagy activators in GBM models produces a concomitant STAT3 inhibition, associated with increased GSCs chemo-sensitization, decreased self-renewal and proliferation [134,145,165].

5.5. Epigenetic Enzymes and Autophagy in GSCs

Recently, aberrant expression and activity of HDACs have been implicated in GBM onset and progression [167]. HDAC inhibition induces GBM cell growth arrest in orthotopic xeno-transplanted mice, and it reduces neurosphere formation from patient-derived GSCs while inducing a neuronal-like phenotype as evident by Tuj-1 upregulation [167]. Remarkably, these effects are promoted by autophagy, while autophagy inhibition counteracts the pro-differentiating effect of GSCs, which is induced by HDAC inhibition [167].
A similar effect is obtained when autophagy is pharmacologically inhibited in GSCs that are treated with a G9a histone methyltransferase inhibitor (BIX01294) [168]. In detail, in glioma cell lines and GBM derived cell cultures, aberrant expression of a G9a histone methyltransferase is bound to low expression levels of both autophagy and differentiation-related genes. Thus, administration of BIX01294 promotes autophagy-dependent GSCs differentiation [168].
The present findings provide evidence about a key role of autophagy in GBM in the light of the interplay with a plethora of intracellular signaling pathways which sustain GSCs malignant phenotype (Figure 3).

6. Conclusions and Future Perspective

The evidence here discussed converges in that autophagy plays a crucial role in GSCs phenotype and GBM malignancy. However, much remains to be elucidated in terms of molecular mechanisms that mediate mTOR dependent and independent autophagy, which remains an important topic of investigation in glioma. Apart from mTOR, a variety of molecular pathways are implicated in GSCs neurobiology and GBM aggressiveness such as STAT3, Wnt/β-catenin and Notch signaling. All these are interconnected to mutually enhance autophagy impairment. Nonetheless, it is worth mentioning that controversial results on autophagy status in GBM still exist in the literature. In fact, some studies report that enhanced autophagy may be implicated in tumor progression. On the one hand, this has to face with the fact that a variety of factors such as tumor micro-environment and tumor stages may differently impact on the autophagy status and flux. On the other hand, misinterpretations often occur when assessing the autophagy status. This is best exemplified by an increase in LC3 levels, which does not necessarily reflect increased autophagy, since it may be due to an accumulation of autophagosomes that do not fuse with lysosomes. Thus, the assessment of autophagy status depends on a careful evaluation of several autophagy markers, such as LC3-I/II ratio, subcellular localization, Atg12–Atg15 accumulation, and p62 degradation along with an assessment of the autophagy flux [169].
In summary, considering the neurobiology of GSCs, the role of autophagy is even amplified compared with the well-established effects already evidenced on GBM as a whole. In fact, autophagy strongly impacts GSCs maintenance, proliferation and resistance to therapies.

Author Contributions

Original draft preparation, writing, review and art work, L.R. and A.G.; review, editing, and art work, F.L. and F.B.; review, editing, P.F. conceptualization, A.F. and V.E.; supervision, F.F.

Funding

This work was funded by Ministero della Salute (Ricerca Corrente 2019).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Aml-1Acute myeloid leukemia 1
BNIP3BCL2/adenovirus E1B 19-kDa protein-interacting protein 3
CNSCentral nervous system
CSCsCancer stem cells
EMTEpithelial-mesenchymal transition
GSCsGlioma stem-like cells
HDACHistone Deacetylases
mTORmammalian Target of Rapamycin
MDRMultidrug resistance
MT1-MMPMembrane Type-1 Matrix Metalloproteinase
NLGN3Neuroligin-3
NSCsNeural stem cells
PDGFBPlatelet-derived growth factor B
PI3KPhosphoinositide 3-kinase
ROSReactive oxygen species (ROS)
SCP2Sterol carrier protein 2
SGZSugranular zone
SVZSubependymal ventricular zone
TFEBTranscription factor EB
UCHL1ubiquitin carboxyl-terminal esterase L1

References

  1. Agnihotri, S.; Burrell, K.E.; Wolf, A.; Jalali, S.; Hawkins, C.; Rutka, J.T.; Zadeh, G. Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies. Arch. Immunol. Ther. Exp. 2013, 61, 25–41. [Google Scholar] [CrossRef] [PubMed]
  2. Ostrom, Q.T.; Gittleman, H.; Farah, P.; Ondracek, A.; Chen, Y.; Wolinsky, Y.; Stroup, N.E.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro Oncol. 2013, 15, ii1–ii56. [Google Scholar] [CrossRef] [PubMed]
  3. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed]
  4. Wen, P.Y.; Kesari, S. Malignant gliomas in adults. N. Engl. J. Med. 2008, 359, 492–507. [Google Scholar] [CrossRef] [PubMed]
  5. Stupp, R.; Brada, M.; van den Bent, M.J.; Tonn, J.C.; Pentheroudakis, G.; ESMO Guidelines Working Group. High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2014, 25, iii93–iii101. [Google Scholar] [CrossRef] [PubMed]
  6. Zoncu, R.; Efeyan, A.; Sabatini, D.M. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 2011, 12, 21–35. [Google Scholar] [CrossRef] [PubMed]
  7. Choe, G.; Horvath, S.; Cloughesy, T.F.; Crosby, K.; Seligson, D.; Palotie, A.; Inge, L.; Smith, B.L.; Sawyers, C.L.; Mischel, P.S. Analysis of the phosphatidylinositol 3’-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res. 2003, 63, 2742–2746. [Google Scholar]
  8. Ermoian, R.P.; Kaprealian, T.; Lamborn, K.R.; Yang, X.; Jelluma, N.; Arvold, N.D.; Zeidman, R.; Berger, M.S.; Stokoe, D.; Haas-Kogan, D.A. Signal transduction molecules in gliomas of all grades. J. Neurooncol. 2009, 91, 19–26. [Google Scholar] [CrossRef]
  9. Yang, J.; Liao, D.; Wang, Z.; Liu, F.; Wu, G. Mammalian target of rapamycin signaling pathway contributes to glioma progression and patients’ prognosis. J. Surg. Res. 2011, 168, 97–102. [Google Scholar] [CrossRef]
  10. Korkolopoulou, P.; Levidou, G.; El-Habr, E.A.; Piperi, C.; Adamopoulos, C.; Samaras, V.; Boviatsis, E.; Thymara, I.; Trigka, E.A.; Sakellariou, S.; et al. Phosphorylated 4E-binding protein 1 (p-4E-BP1): A novel prognostic marker in human astrocytomas. Histopathology 2012, 61, 293–305. [Google Scholar] [CrossRef]
  11. Chakravarti, A.; Zhai, G.; Suzuki, Y.; Sarkesh, S.; Black, P.M.; Muzikansky, A.; Loeffler, J.S. The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. J. Clin. Oncol. 2004, 22, 1926–1933. [Google Scholar] [CrossRef]
  12. Hu, X.; Pandolfi, P.P.; Li, Y.; Koutcher, J.A.; Rosenblum, M.; Holland, E.C. mTOR promotes survival and astrocytic characteristics induced by Pten/AKT signaling in glioblastoma. Neoplasia 2005, 7, 356–368. [Google Scholar] [CrossRef]
  13. Akhavan, D.; Cloughesy, T.F.; Mischel, P.S. mTOR signaling in glioblastoma: Lessons learned from bench to bedside. NeuroOncology 2010, 12, 882–889. [Google Scholar] [CrossRef] [PubMed]
  14. Fan, Q.W.; Weiss, W.A. Inhibition of PI3K-Akt-mTOR signaling in glioblastoma by mTORC1/2 inhibitors. Methods Mol. Biol. 2012, 821, 349–359. [Google Scholar] [CrossRef]
  15. Li, X.; Wu, C.; Chen, N.; Gu, H.; Yen, A.; Cao, L.; Wang, E.; Wang, L. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget 2016, 7, 33440–33450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ryskalin, L.; Limanaqi, F.; Biagioni, F.; Frati, A.; Esposito, V.; Calierno, M.T.; Lenzi, P.; Fornai, F. The emerging role of m-TOR up-regulation in brain Astrocytoma. Histol. Histopathol. 2017, 32, 413–431. [Google Scholar]
  17. Mecca, C.; Giambanco, I.; Donato, R.; Arcuri, C. Targeting mTOR in Glioblastoma: Rationale and Preclinical/Clinical Evidence. Dis. Markers 2018, 2018, 9230479. [Google Scholar] [CrossRef]
  18. Li, X.Y.; Zhang, L.Q.; Zhang, X.G.; Li, X.; Ren, Y.B.; Ma, X.Y.; Li, X.G.; Wang, L.X. Association between AKT/mTOR signalling pathway and malignancy grade of human gliomas. J. Neurooncol. 2011, 103, 453–458. [Google Scholar] [CrossRef]
  19. Machado, L.E.; Alvarenga, A.W.; da Silva, F.F.; Roffé, M.; Begnami, M.D.; Torres, L.F.B.; da Cunha, I.W.; Martins, V.R.; Hajj, G.N.M. Overexpression of mTOR and p(240-244)S6 in IDH1 Wild-Type Human Glioblastomas Is Predictive of Low Survival. J. Histochem. Cytochem. 2018, 66, 403–414. [Google Scholar] [CrossRef]
  20. Neshat, M.S.; Mellinghoff, I.K.; Tran, C.; Stiles, B.; Thomas, G.; Petersen, R.; Frost, P.; Gibbons, J.J.; Wu, H.; Sawyers, C.L. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc. Natl. Acad. Sci. USA 2001, 98, 10314–10319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Radaelli, E.; Ceruti, R.; Patton, V.; Russo, M.; Degrassi, A.; Croci, V.; Caprera, F.; Stortini, G.; Scanziani, E.; Pesenti, E.; et al. Immunohistopathological and neuroimaging characterization of murine orthotopic xenograft models of glioblastoma multiforme recapitulating the most salient features of human disease. Histol. Histopathol. 2009, 24, 879–891. [Google Scholar]
  22. Iwamaru, A.; Kondo, Y.; Iwado, E.; Aoki, H.; Fujiwara, K.; Yokoyama, T.; Mills, G.B.; Kondo, S. Silencing mammalian target of rapamycin signaling by small interfering RNA enhances rapamycin-induced autophagy in malignant glioma cells. Oncogene 2007, 26, 1840–1851. [Google Scholar] [CrossRef]
  23. Jiang, H.; Gomez-Manzano, C.; Aoki, H.; Alonso, M.M.; Kondo, S.; McCormick, F.; Xu, J.; Kondo, Y.; Bekele, B.N.; Colman, H.; et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: Role of autophagic cell death. J. Natl. Cancer. Inst. 2007, 99, 1410–1414. [Google Scholar] [CrossRef]
  24. Jiang, H.; White, E.J.; Conrad, C.; Gomez-Manzano, C.; Fueyo, J. Autophagy pathways in glioblastoma. Methods Enzymol. 2009, 453, 273–286. [Google Scholar]
  25. Huang, X.; Bai, H.M.; Chen, L.; Li, B.; Lu, Y.C. Reduced expression of LC3B-II and Beclin 1 in glioblastoma multiforme indicates a down-regulated autophagic capacity that relates to the progression of astrocytic tumors. J. Clin. Neurosci. 2010, 17, 1515–1519. [Google Scholar] [CrossRef]
  26. Zhao, Y.; Huang, Q.; Yang, J.; Lou, M.; Wang, A.; Dong, J.; Qin, Z.; Zhang, T. Autophagy impairment inhibits differentiation of glioma stem/progenitor cells. Brain Res. 2010, 1313, 250–258. [Google Scholar] [CrossRef]
  27. Zhuang, W.; Li, B.; Long, L.; Chen, L.; Huang, Q.; Liang, Z. Induction of autophagy promotes differentiation of glioma-initiating cells and their radiosensitivity. Int. J. Cancer 2011, 129, 2720–2731. [Google Scholar] [CrossRef]
  28. Zhuang, W.; Li, B.; Long, L.; Chen, L.; Huang, Q.; Liang, Z.Q. Knockdown of the DNA-dependent protein kinase catalytic subunit radiosensitizes glioma-initiating cells by inducing autophagy. Brain Res. 2011, 1371, 7–15. [Google Scholar] [CrossRef]
  29. Arcella, F.; Biagioni, M.; Oliva, A.; Bucci, D.; Frati, A.; Esposito, V.; Cantore, G.; Giangaspero, F.; Fornai, F. Rapamycin inhibits the growth of glioblastoma. Brain Res. 2013, 1495, 37–51. [Google Scholar] [CrossRef]
  30. Liu, R.; Li, J.; Zhang, T.; Zou, L.; Chen, Y.; Wang, K.; Lei, Y.; Yuan, K.; Li, Y.; Lan, J.; et al. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: Involvement of abnormal cholesterol trafficking. Autophagy 2014, 10, 1241–1255. [Google Scholar] [CrossRef]
  31. Lenzi, P.; Lazzeri, G.; Biagioni, F.; Busceti, C.L.; Gambardella, S.; Salvetti, A.; Fornai, F. The Autophagoproteasome a Novel Cell Clearing Organelle in Baseline and Stimulated Conditions. Front. Neuroanat. 2016, 10, 78. [Google Scholar] [CrossRef] [Green Version]
  32. Ferrucci, M.; Biagioni, F.; Lenzi, P.; Gambardella, S.; Ferese, R.; Calierno, M.T.; Falleni, A.; Grimaldi, A.; Frati, A.; Esposito, V.; et al. Rapamycin promotes differentiation increasing βIII-tubulin, NeuN, and NeuroD while suppressing nestin expression in glioblastoma cells. Oncotarget 2017, 8, 29574–29599. [Google Scholar] [CrossRef]
  33. 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]
  34. 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]
  35. 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, 756–760. [Google Scholar] [CrossRef]
  36. Liu, Q.; Nguyen, D.H.; Dong, Q.; Shitaku, P.; Chung, K.; Liu, O.Y.; Tso, J.L.; Liu, J.Y.; Konkankit, V.; Cloughesy, T.F.; et al. Molecular properties of CD133+ glioblastoma stem cells derived from treatment-refractory recurrent brain tumors. J. Neurooncol. 2009, 94, 1–19. [Google Scholar] [CrossRef] [Green Version]
  37. Zhuang, W.; Qin, Z.; Liang, Z. The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim. Biophys. Sin. 2009, 41, 341–351. [Google Scholar] [CrossRef] [Green Version]
  38. Toda, M. Glioma stem cells and immunotherapy for the treatment of malignant gliomas. ISRN Oncol. 2013, 2013, 673793. [Google Scholar] [CrossRef]
  39. Inda, M.M.; Bonavia, R.; Seoane, J. Glioblastoma multiforme: A look inside its heterogeneous nature. Cancers 2014, 6, 226–239. [Google Scholar] [CrossRef]
  40. Vessoni, A.T.; Muotri, A.R.; Okamoto, O.K. Autophagy in stem cell maintenance and differentiation. Stem Cells Dev. 2012, 21, 513–520. [Google Scholar] [CrossRef]
  41. Wang, C.; Liang, C.C.; Bian, Z.C.; Zhu, Y.; Guan, J.L. FIP200 is required for maintenance and differentiation of postnatal neural stem cells. Nat. Neurosci. 2013, 16, 532–542. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, X.; He, Y.; Lu, F. Autophagy in Stem Cell Biology: A Perspective on Stem Cell Self-Renewal and Differentiation. Stem Cells Int. 2018, 2018, 9131397. [Google Scholar] [CrossRef]
  43. Tao, Z.; Li, T.; Ma, H.; Yang, Y.; Zhang, C.; Hai, L.; Liu, P.; Yuan, F.; Li, J.; Yi, L.; et al. Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death Dis. 2018, 9, 1063. [Google Scholar] [CrossRef]
  44. Lee, D.Y. Roles of mTOR Signaling in Brain Development. Exp. Neurobiol. 2015, 24, 177–185. [Google Scholar] [CrossRef] [Green Version]
  45. Casares-Crespo, L.; Calatayud-Baselga, I.; García-Corzo, L.; Mira, H. On the Role of Basal Autophagy in Adult Neural Stem Cells and Neurogenesis. Front. Cell. Neurosci. 2018, 12, 339. [Google Scholar] [CrossRef]
  46. LiCausi, F.; Hartman, N.W. Role of mTOR Complexes in Neurogenesis. Int. J. Mol. Sci. 2018, 19, 1544. [Google Scholar] [CrossRef]
  47. Vescovi, A.L.; Galli, R.; Reynolds, B.A. Brain tumor stem cells. Nat. Rev. Cancer 2006, 6, 425–436. [Google Scholar] [CrossRef]
  48. Sunayama, J.; Sato, A.; Matsuda, K.; Tachibana, K.; Suzuki, K.; Narita, Y.; Shibui, S.; Sakurada, K.; Kayama, T.; Tomiyama, A.; et al. Dual blocking of mTor and PI3K elicits a prodifferentiation effect on glioblastoma stem-like cells. Neuro Oncol. 2010, 12, 1205–1219. [Google Scholar] [CrossRef] [Green Version]
  49. Stepanenko, A.A.; Andreieva, S.V.; Korets, K.V.; Mykytenko, D.O.; Baklaushev, V.P.; Chekhonin, V.P.; Dmitrenko, V.V. mTOR inhibitor temsirolimus and MEK1/2 inhibitor U0126 promote chromosomal instability and cell type-dependent phenotype changes of glioblastoma cells. Gene 2016, 579, 58–68. [Google Scholar] [CrossRef]
  50. Ohgaki, H.; Kleihues, P. Genetic pathways to primary and secondary glioblastoma. Am. J. Pathol. 2007, 170, 1445–1453. [Google Scholar] [CrossRef]
  51. Galan-Moya, E.M.; LeGuelte, A.; Fernandes, E.L.; Thirant, C.; Dwyer, J.; Bidere, N.; Couraud, P.O.; Scott, M.G.; Junier, M.P.; Chneiweiss, H.; et al. Secreted factors from brain endothelial cells maintain glioblastoma stem-like cell expansion through the mTOR pathway. EMBO Rep. 2011, 12, 470–476. [Google Scholar] [CrossRef]
  52. Jhanwar-Uniyal, M.; Albert, L.; McKenna, E.; Karsy, M.; Rajdev, P.; Braun, A.; Murali, R. Deciphering the signaling pathways of cancer stem cells of glioblastoma multiforme: Role of Akt/mTOR and MAPK pathways. Adv. Enzyme Regul. 2011, 51, 164–170. [Google Scholar] [CrossRef]
  53. Jhanwar-Uniyal, M.; Jeevan, D.; Neil, J.; Shannon, C.; Albert, L.; Murali, R. Deconstructing mTOR complexes in regulation of glioblastoma multiforme and its stem cells. Adv. Biol. Regul. 2013, 53, 202–210. [Google Scholar] [CrossRef]
  54. Fu, J.; Liu, Z.G.; Liu, X.M.; Chen, F.R.; Shi, H.L.; Pangjesse, C.S.; Ng, H.K.; Chen, Z.P. Glioblastoma stem cells resistant to temozolomide-induced autophagy. Chin. Med. J. (Engl). 2009, 122, 1255–1259. [Google Scholar]
  55. Catalano, M.; D’Alessandro, G.; Lepore, F.; Corazzari, M.; Caldarola, S.; Valacca, C.; Faienza, F.; Esposito, V.; Limatola, C.; Cecconi, F.; et al. Autophagy induction impairs migration and invasion by reversing EMT in glioblastoma cells. Mol. Oncol. 2015, 9, 1612–1625. [Google Scholar] [CrossRef] [Green Version]
  56. Chandrika, G.; Natesh, K.; Ranade, D.; Chugh, A.; Shastry, P. Suppression of the invasive potential of glioblastoma cells by mTOR inhibitors involves modulation of NFκB and PKC-α signaling. Sci. Rep. 2016, 6, 22455. [Google Scholar] [CrossRef]
  57. Gong, A.; Huang, S. FoxM1 and Wnt/β-catenin signaling in glioma stem cells. Cancer Res. 2012, 72, 5658–5662. [Google Scholar] [CrossRef]
  58. Cochrane, C.R.; Szczepny, A.; Watkins, D.N.; Cain, J.E. Hedgehog Signaling in the Maintenance of Cancer Stem Cells. Cancers 2015, 7, 1554–1585. [Google Scholar] [CrossRef]
  59. Bazzoni, R.; Bentivegna, A. Role of Notch Signaling Pathway in Glioblastoma Pathogenesis. Cancers 2019, 11, 292. [Google Scholar] [CrossRef]
  60. 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]
  61. Nunes, M.C.; Roy, N.S.; Keyoung, H.M.; Goodman, R.R.; McKhann, G., 2nd; Jiang, L.; Kang, J.; Nedergaard, M.; Goldman, S.A. Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat. Med. 2003, 9, 439–447. [Google Scholar] [CrossRef]
  62. 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-García 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]
  63. Paliouras, G.N.; Hamilton, L.K.; Aumont, A.; Joppé, S.E.; Barnabé-Heider, F.; Fernandes, K.J. Mammalian target of rapamycin signaling is a key regulator of the transit-amplifying progenitor pool in the adult and aging forebrain. J. Neurosci. 2012, 32, 15012–15026. [Google Scholar] [CrossRef]
  64. 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]
  65. 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]
  66. Basak, O.; Taylor, V. Stem cells of the mammalian brain. Cell. Mol. Life Sci. 2009, 66, 1057–1072. [Google Scholar] [CrossRef]
  67. Visvader, J.E. Cells of origin in cancer. Nature 2011, 469, 314–322. [Google Scholar] [CrossRef]
  68. Zong, H.; Parada, L.F.; Baker, S.J. Cell of origin for malignant gliomas and its implication in therapeutic development. Cold Spring. Harb. Perspect Biol. 2015, 7, a020610. [Google Scholar] [CrossRef]
  69. Alcantara Llaguno, S.R.; Parada, L.F. Cell of origin of glioma: Biological and clinical implications. Br. J. Cancer 2016, 115, 1445–1450. [Google Scholar] [CrossRef]
  70. 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]
  71. Wang, J.; Sakariassen, P.Ø.; Tsinkalovsky, O.; Immervoll, H.; Bøe, S.O.; Svendsen, A.; Prestegarden, L.; Røsland, G.; Thorsen, F.; Stuhr, L.; et al. CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int. J. Cancer 2008, 122, 761–788. [Google Scholar] [CrossRef]
  72. Lendahl, U.; Zimmerman, L.B.; McKay, R.D. CNS stem cells express a new class of intermediate filament protein. Cell 1990, 60, 585–595. [Google Scholar] [CrossRef]
  73. 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] [Green Version]
  74. Ogden, A.T.; Waziri, A.E.; Lochhead, R.A.; Fusco, D.; Lopez, K.; Ellis, J.A.; Kang, J.; Assanah, M.; McKhann, G.M.; Sisti, M.B.; et al. Identification of A2B5+CD133- tumor-initiating cells in adult human gliomas. Neurosurgery 2008, 62, 505–514. [Google Scholar] [CrossRef]
  75. Son, M.J.; Woolard, K.; Nam, D.H.; Lee, J.; Fine, H.A. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell Stem Cell 2009, 4, 440–452. [Google Scholar] [CrossRef]
  76. Ward, R.J.; Lee, L.; Graham, K.; Satkunendran, T.; Yoshikawa, K.; Ling, E.; Harper, L.; Austin, R.; Nieuwenhuis, E.; Clarke, I.D.; et al. Multipotent CD15+ cancer stem cells in patched-1-deficient mouse medulloblastoma. Cancer Res. 2009, 69, 4682–4690. [Google Scholar] [CrossRef]
  77. Anido, J.; Saez-Borderias, A.; Gonzalez-Junca, A.; Rodon, L.; Folch, G.; Carmona, M.A.; Prieto-Sánchez, R.M.; Barba, I.; Martínez-Sáez, E.; Prudkin, L.; et al. TGF-beta receptor inhibitors target the CD44(high)/Id1(high) glioma-initiating cell population in human glioblastoma. Cancer Cell 2010, 18, 655–668. [Google Scholar] [CrossRef]
  78. Lathia, J.D.; Gallagher, J.; Heddleston, J.M.; Wang, J.; Eyler, C.E.; Macswords, J.; Wu, Q.; Vasanji, A.; McLendon, R.E.; Hjelmeland, A.B.; et al. Integrin α6 regulates glioblastoma stem cells. Cell Stem Cell 2010, 6, 421–432. [Google Scholar] [CrossRef]
  79. Tchoghandjian, A.; Baeza, N.; Colin, C.; Cayre, M.; Metellus, P.; Beclin, C.; Ouafik, L.; Figarella-Branger, D. A2B5 cells from human glioblastoma have cancer stem cell properties. Brain Pathol. 2010, 20, 211–221. [Google Scholar] [CrossRef]
  80. Cheng, L.; Wu, Q.L.; Huang, Z.; Guryanova, O.A.; Huang, Q.A.; Shou, W.N.; Rich, J.N.; Bao, S. L1CAM regulates DNA damage checkpoint response of glioblastoma stem cells through NBS1. EMBO J. 2011, 30, 800–813. [Google Scholar] [CrossRef]
  81. Bhat, K.P.L.; Balasubramaniyan, V.; Vaillant, B.; Ezhilarasan, R.; Hummelink, K.; Hollingsworth, F.; Wani, K.; Heathcock, L.; James, J.D.; Goodman, L.D.; et al. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell 2013, 24, 331–346. [Google Scholar] [CrossRef]
  82. Wang, J.; Wang, H.; Li, Z.; Wu, Q.; Lathia, J.D.; McLendon, R.E.; Hjelmeland, A.B.; Rich, J.N. c-Myc is required for maintenance of glioma cancer stem cells. PLoS ONE 2008, 3, e3769. [Google Scholar] [CrossRef]
  83. Sherry, M.M.; Reeves, A.; Wu, J.K.; Cochran, B.H. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 2009, 27, 2383–2392. [Google Scholar] [CrossRef]
  84. Zbinden, M.; Duquet, A.; Lorente-Trigos, A.; Ngwabyt, S.N.; Borges, I.; Ruiz i Altaba, A. NANOG regulates glioma stem cells and is essential in vivo acting in a cross-functional network with GLI1 and p53. EMBO J. 2010, 29, 2659–2674. [Google Scholar] [CrossRef] [Green Version]
  85. Talsma, C.E.; Flack, C.G.; Zhu, T.; He, X.; Soules, M.; Heth, J.A.; Muraszko, K.; Fan, X. Oct4 regulates GBM neurosphere growth and its expression is associated with poor survival in GBM patients. Neuro Oncol. 2011, 13, 145–153. [Google Scholar]
  86. 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]
  87. Iacopino, F.; Angelucci, C.; Piacentini, R.; Biamonte, F.; Mangiola, A.; Maira, G.; Grassi, C.; Sica, G. Isolation of cancer stem cells from three human glioblastoma cell lines: Characterization of two selected clones. PLoS ONE 2014, 9, e105166. [Google Scholar] [CrossRef]
  88. Laplante, M.; Sabatini, D.M. mTOR signaling in growth control and disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef]
  89. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell. Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [Green Version]
  90. Meijer, A.J.; Codogno, P. Signalling and autophagy regulation in health, aging and disease. Mol. Asp. Med. 2006, 27, 411–425. [Google Scholar] [CrossRef]
  91. Roczniak-Ferguson, A.; Petit, C.S.; Froehlich, F.; Qian, S.; Ky, J.; Angarola, B.; Walther, T.C.; Ferguson, S.M. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal. 2012, 5, ra42. [Google Scholar] [CrossRef]
  92. Vega-Rubin-de-Celis, S.; Peña-Llopis, S.; Konda, M.; Brugarolas, J. Multistep regulation of TFEB by MTORC1. Autophagy 2017, 13, 464–472. [Google Scholar] [CrossRef] [Green Version]
  93. Pachow, D.; Wick, W.; Gutmann, D.H.; Mawrin, C. The mTOR signaling pathway as a treatment target for intracranial neoplasms. Neuro Oncol. 2015, 17, 189–199. [Google Scholar] [CrossRef]
  94. Tsang, C.K.; Qi, H.; Liu, L.F.; Zheng, X.F. Targeting mammalian target of rapamycin (mTOR) for health and diseases. Drug Discov. Today 2007, 12, 112–124. [Google Scholar] [CrossRef]
  95. Tian, T.; Li, X.; Zhang, J. Mtor signaling in cancer and mtor inhibitors in solid tumor targeting therapy. Int. J. Mol. Sci. 2019, 20, 755. [Google Scholar] [CrossRef]
  96. Sato, A.; Sunayama, J.; Matsuda, K.; Tachibana, K.; Sakurada, K.; Tomiyama, A.; Kayama, T.; Kitanaka, C. Regulation of neural stem/progenitor cell maintenance by PI3K and mTOR. Neurosci. Lett. 2010, 470, 115–120. [Google Scholar] [CrossRef]
  97. Hartman, N.W.; Lin, T.V.; Zhang, L.; Paquelet, G.E.; Feliciano, D.M.; Bordey, A. mTORC1 targets the translational repressor 4E-BP2, but not S6 kinase 1/2, to regulate neural stem cell self-renewal in vivo. Cell Rep. 2013, 5, 433–444. [Google Scholar] [CrossRef]
  98. Ka, M.; Condorelli, G.; Woodgett, J.R.; Kim, W.Y. mTOR regulates brain morphogenesis by mediating GSK3 signaling. Development 2014, 141, 4076–4086. [Google Scholar] [CrossRef] [Green Version]
  99. Cloetta, D.; Thomanetz, V.; Baranek, C.; Lustenberger, R.M.; Lin, S.; Oliveri, F.; Atanasoski, S.; Ruegg, M.A. Inactivation of mTORC1 in the developing brain causes microcephaly and affects gliogenesis. J. Neurosci. 2013, 33, 7799–7810. [Google Scholar] [CrossRef]
  100. Lafourcade, C.A.; Lin, T.V.; Feliciano, D.M.; Zhang, L.; Hsieh, L.S.; Bordey, A. Rheb activation in subventricular zone progenitors leads to heterotopia, ectopic neuronal differentiation, and rapamycin-sensitive olfactory micronodules and dendrite hypertrophy of newborn neurons. J. Neurosci. 2013, 33, 2419–2431. [Google Scholar] [CrossRef]
  101. Kassai, H.; Sugaya, Y.; Noda, S.; Nakao, K.; Maeda, T.; Kano, M.; Aiba, A. Selective activation of mTORC1 signaling recapitulates microcephaly, tuberous sclerosis, and neurodegenerative diseases. Cell Rep. 2014, 7, 1626–1639. [Google Scholar] [CrossRef]
  102. Magini, A.; Polchi, A.; Di Meo, D.; Mariucci, G.; Sagini, K.; De Marco, F.; Cassano, T.; Giovagnoli, S.; Dolcetta, D.; Emiliani, C. TFEB activation restores migration ability to Tsc1-deficient adult neural stem/progenitor cells. Hum. Mol. Genet. 2017, 26, 3303–3312. [Google Scholar] [CrossRef] [Green Version]
  103. Magri, L.; Cambiaghi, M.; Cominelli, M.; Alfaro-Cervello, C.; Cursi, M.; Pala, M.; Bulfone, A.; García-Verdugo, J.M.; Leocani, L.; Minicucci, F.; et al. Sustained activation of mTOR pathway in embryonic neural stem cells leads to development of tuberous sclerosis complex-associated lesions. Cell Stem Cell 2011, 9, 447–462. [Google Scholar] [CrossRef]
  104. Nieto-González, J.L.; Gómez-Sánchez, L.; Mavillard, F.; Linares-Clemente, P.; Rivero, M.C.; Valenzuela-Villatoro, M.; Muñoz-Bravo, J.L.; Pardal, R.; Fernández-Chacón, R. Loss of postnatal quiescence of neural stem cells through mTOR activation upon genetic removal of cysteine string protein-α. Proc. Natl. Acad. Sci. USA 2019, 116, 8000–8009. [Google Scholar] [CrossRef]
  105. Bashir, T.; Cloninger, C.; Artinian, N.; Anderson, L.; Bernath, A.; Holmes, B.; Benavides-Serrato, A.; Sabha, N.; Nishimura, R.N.; Guha, A.; et al. Conditional astroglial Rictor overexpression induces malignant glioma in mice. PLoS ONE 2012, 7, e47741. [Google Scholar] [CrossRef]
  106. Takei, N.; Nawa, H. mTOR signaling and its roles in normal and abnormal brain development. Front. Mol. Neurosci. 2014, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  107. Tee, A.R.; Sampson, J.R.; Pal, D.K.; Bateman, J.M. The role of mTOR signalling in neurogenesis, insights from tuberous sclerosis complex. Semin. Cell. Dev. Biol. 2016, 52, 12–20. [Google Scholar] [CrossRef]
  108. Ryskalin, L.; Lazzeri, G.; Flaibani, M.; Biagioni, F.; Gambardella, S.; Frati, A.; Fornai, F. mTOR-Dependent Cell Proliferation in the Brain. Biomed Res. Int. 2017, 2017, 7082696. [Google Scholar] [CrossRef]
  109. Zhao, H.F.; Wang, J.; Shao, W.; Wu, C.P.; Chen, Z.P.; To, S.T.; Li, W.P. Recent advances in the use of PI3K inhibitors for glioblastoma multiforme: Current preclinical and clinical development. Mol. Cancer 2017, 16, 100. [Google Scholar] [CrossRef]
  110. Venkatesh, H.S.; Johung, T.B.; Caretti, V.; Noll, A.; Tang, Y.; Nagaraja, S.; Gibson, E.M.; Mount, C.W.; Polepalli, J.; Mitra, S.S.; et al. Neuronal Activity Promotes Glioma Growth through Neuroligin-3 Secretion. Cell 2015, 161, 803–816. [Google Scholar] [CrossRef] [Green Version]
  111. Pietras, A.; Katz, A.M.; Ekström, E.J.; Wee, B.; Halliday, J.J.; Pitter, K.L.; Werbeck, J.L.; Amankulor, N.M.; Huse, J.T.; Holland, E.C. Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell 2014, 14, 357–369. [Google Scholar] [CrossRef]
  112. Gangemi, R.M.; Griffero, F.; Marubbi, D.; Perera, M.; Capra, M.C.; Malatesta, P.; Ravetti, G.L.; Zona, G.L.; Daga, A.; Corte, G. SOX2 silencing in glioblastoma tumor-initiating cells causes stop of proliferation and loss of tumorigenicity. Stem Cells 2009, 27, 40–48. [Google Scholar] [CrossRef]
  113. Suvà, M.L.; Rheinbay, E.; Gillespie, S.M.; Patel, A.P.; Wakimoto, H.; Rabkin, S.D.; Riggi, N.; Chi, A.S.; Cahill, D.P.; Nahed, B.V.; et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell 2014, 157, 580–594. [Google Scholar] [CrossRef]
  114. Garros-Regulez, L.; Garcia, I.; Carrasco-Garcia, E.; Lantero, A.; Aldaz, P.; Moreno-Cugnon, L.; Arrizabalaga, O.; Undabeitia, J.; Torres-Bayona, S.; Villanua, J.; et al. Targeting SOX2 as a Therapeutic Strategy in Glioblastoma. Front. Oncol. 2016, 6, 222. [Google Scholar] [CrossRef]
  115. Soeda, A.; Park, M.; Lee, D.; Mintz, A.; Androutsellis-Theotokis, A.; McKay, R.D.; Engh, J.; Iwama, T.; Kunisada, T.; Kassam, A.B.; et al. Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1alpha. Oncogene 2009, 28, 3949–3959. [Google Scholar] [CrossRef]
  116. Sato, A.; Okada, M.; Shibuya, K.; Watanabe, E.; Seino, S.; Suzuki, K.; Narita, Y.; Shibui, S.; Kayama, T.; Kitanaka, C. Resveratrol promotes proteasome-dependent degradation of Nanog via p53 activation and induces differentiation of glioma stem cells. Stem Cell Res. 2013, 11, 601–610. [Google Scholar] [CrossRef] [Green Version]
  117. Chandrika, G.; Natesh, K.; Ranade, D.; Chugh, A.; Shastry, P. Mammalian target of rapamycin inhibitors, temsirolimus and torin 1, attenuate stemness-associated properties and expression of mesenchymal markers promoted by phorbol-myristate-acetate and oncostatin-M in glioblastoma cells. Tumour Biol. 2017, 39, 1010428317695921. [Google Scholar] [CrossRef]
  118. Nanta, R.; Shrivastava, A.; Sharma, J.; Shankar, S.; Srivastava, R.K. Inhibition of sonic hedgehog and PI3K/Akt/mTOR pathways cooperate in suppressing survival, self-renewal and tumorigenic potential of glioblastoma-initiating cells. Mol. Cell. Biochem. 2019, 454, 11–23. [Google Scholar] [CrossRef]
  119. Mendiburu-Eliçabe, M.; Gil-Ranedo, J.; Izquierdo, M. Efficacy of rapamycin against glioblastoma cancer stem cells. Clin. Transl. Oncol. 2014, 16, 495–502. [Google Scholar] [CrossRef]
  120. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef] [Green Version]
  121. Taylor, M.A.; Das, B.C.; Ray, S.K. Targeting autophagy for combating chemoresistance and radioresistance in glioblastoma. Apoptosis 2018, 23, 563–575. [Google Scholar] [CrossRef]
  122. Kahlert, U.D.; Maciaczyk, D.; Dai, F.; Claus, R.; Firat, E.; Doostkam, S.; Bogiel, T.; Carro, M.S.; Döbrössy, M.; Herold-Mende, C.; et al. Resistance to hypoxia-induced, BNIP3-mediated cell death contributes to an increase in a CD133-positive cell population in human glioblastomas in vitro. J. Neuropathol. Exp. Neurol. 2012, 71, 1086–1099. [Google Scholar] [CrossRef]
  123. Hu, Y.L.; DeLay, M.; Jahangiri, A.; Molinaro, A.M.; Rose, S.D.; Carbonell, W.S.; Aghi, M.K. Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res. 2012, 72, 1773–1783. [Google Scholar] [CrossRef]
  124. Jawhari, S.; Ratinaud, M.H.; Verdier, M. Glioblastoma, hypoxia and autophagy: A survival-prone ‘ménage-à-trois’. Cell Death Dis. 2016, 7, e2434. [Google Scholar] [CrossRef]
  125. Golden, E.B.; Cho, H.Y.; Jahanian, A.; Hofman, F.M.; Louie, S.G.; Schönthal, A.H.; Chen, T.C. Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg. Focus 2014, 37, E12. [Google Scholar] [CrossRef] [Green Version]
  126. Pratt, J.; Annabi, B. Induction of autophagy biomarker BNIP3 requires a JAK2/STAT3 and MT1-MMP signaling interplay in Concanavalin-A-activated U87 glioblastoma cells. Cell Signal. 2014, 26, 917–924. [Google Scholar] [CrossRef]
  127. Natsumeda, M.; Maitani, K.; Liu, Y.; Miyahara, H.; Kaur, H.; Chu, Q.; Zhang, H.; Kahlert, U.D.; Eberhart, C.G. Targeting Notch Signaling and Autophagy Increases Cytotoxicity in Glioblastoma Neurospheres. Brain Pathol. 2016, 26, 713–723. [Google Scholar] [CrossRef]
  128. Bischof, J.; Westhoff, M.A.; Wagner, J.E.; Halatsch, M.E.; Trentmann, S.; Knippschild, U.; Wirtz, C.R.; Burster, T. Cancer stem cells: The potential role of autophagy, proteolysis, and cathepsins in glioblastoma stem cells. Tumour Biol. 2017, 39, 1010428317692227. [Google Scholar] [CrossRef]
  129. Xu, J.; Dang, Y.; Ren, Y.R.; Liu, J.O. Cholesterol trafficking is required for mTOR activation in endothelial cells. Proc. Natl. Acad. Sci. USA 2010, 107, 4764–4769. [Google Scholar] [CrossRef] [Green Version]
  130. Würth, R.; Pattarozzi, A.; Gatti, M.; Bajetto, A.; Corsaro, A.; Parodi, A.; Sirito, R.; Massollo, M.; Marini, C.; Zona, G.; et al. Metformin selectively affects human glioblastoma tumor-initiating cell viability: A role for metformin-induced inhibition of Akt. Cell Cycle 2013, 12, 145–156. [Google Scholar] [CrossRef]
  131. Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef]
  132. Friedman, M.D.; Jeevan, D.S.; Tobias, M.; Murali, R.; Jhanwar-Uniyal, M. Targeting cancer stem cells in glioblastoma multiforme using mTOR inhibitors and the differentiating agent all-trans retinoic acid. Oncol. Rep. 2013, 30, 1645–1650. [Google Scholar] [CrossRef] [Green Version]
  133. Wang, W.J.; Long, L.M.; Yang, N.; Zhang, Q.Q.; Ji, W.J.; Zhao, J.H.; Qin, Z.H.; Wang, Z.; Chen, G.; Liang, Z.Q. NVP-BEZ235, a novel dual PI3K/mTOR inhibitor, enhances the radiosensitivity of human glioma stem cells in vitro. Acta Pharmacol. Sin. 2013, 34, 681–690. [Google Scholar] [CrossRef] [Green Version]
  134. Li, H.; Chen, L.; Li, J.J.; Zhou, Q.; Huang, A.; Liu, W.W.; Wang, K.; Gao, L.; Qi, S.T.; Lu, Y.T. miR-519a enhances chemosensitivity and promotes autophagy in glioblastoma by targeting STAT3/Bcl2 signaling pathway. J. Hematol. Oncol. 2018, 11, 70. [Google Scholar] [CrossRef]
  135. Nabissi, M.; Morelli, M.B.; Amantini, C.; Liberati, S.; Santoni, M.; Ricci-Vitiani, L.; Pallini, R.; Santoni, G. Cannabidiol stimulates Aml-1a-dependent glial differentiation and inhibits glioma stem-like cells proliferation by inducing autophagy in a TRPV2-dependent manner. Int. J. Cancer 2015, 137, 1855–1869. [Google Scholar] [CrossRef] [Green Version]
  136. Comincini, S.; Allavena, G.; Palumbo, S.; Morini, M.; Durando, F.; Angeletti, F.; Pirtoli, L.; Miracco, C. microRNA-17 regulates the expression of ATG7 and modulates the autophagy process, improving the sensitivity to temozolomide and low-dose ionizing radiation treatments in human glioblastoma cells. Cancer Biol. Ther. 2013, 14, 574–586. [Google Scholar] [CrossRef]
  137. Coly, P.M.; Perzo, N.; Le Joncour, V.; Lecointre, C.; Schouft, M.T.; Desrues, L.; Tonon, M.C.; Wurtz, O.; Gandolfo, P.; Castel, H.; et al. Chemotactic G protein-coupled receptors control cell migration by repressing autophagosome biogenesis. Autophagy 2016, 12, 2344–2362. [Google Scholar] [CrossRef] [Green Version]
  138. Wang, C.C.; Hueng, D.Y.; Huang, A.F.; Chen, W.L.; Huang, S.M.; Yi-Hsin Chan, J. CD164 regulates proliferation, progression, and invasion of human glioblastoma cells. Oncotarget 2019, 10, 2041–2054. [Google Scholar] [CrossRef]
  139. Watt, S.M.; Chan, J.Y. CD164–a novel sialomucin on CD34+ cells. Leuk. Lymphoma 2000, 37, 1–25. [Google Scholar] [CrossRef]
  140. Tu, M.; Cai, L.; Zheng, W.; Su, Z.; Chen, Y.; Qi, S. CD164 regulates proliferation and apoptosis by targeting PTEN in human glioma. Mol. Med. Rep. 2017, 15, 1713–1721. [Google Scholar] [CrossRef]
  141. Cheng, Y.C.; Hueng, D.Y.; Huang, H.Y.; Chen, J.Y.; Chen, Y. Magnolol and honokiol exert a synergistic anti-tumor effect through autophagy and apoptosis in human glioblastomas. Oncotarget 2016, 7, 29116–29130. [Google Scholar] [CrossRef]
  142. Jiao, Y.; Li, H.; Liu, Y.; Guo, A.; Xu, X.; Qu, X.; Wang, S.; Zhao, J.; Li, Y.; Cao, Y. Resveratrol Inhibits the Invasion of Glioblastoma-Initiating Cells via Down-Regulation of the PI3K/Akt/NF-κB Signaling Pathway. Nutrients 2015, 7, 4383–4402. [Google Scholar] [CrossRef]
  143. Zhang, H.; Zhu, Y.; Sun, X.; He, X.; Wang, M.; Wang, Z.; Wang, Q.; Zhu, R.; Wang, S. Curcumin-Loaded Layered Double Hydroxide Nanoparticles-Induced Autophagy for Reducing Glioma Cell Migration and Invasion. J. Biomed Nanotechnol. 2016, 12, 2051–2062. [Google Scholar] [CrossRef]
  144. Filippi-Chiela, E.C.; Villodre, E.S.; Zamin, L.L.; Lenz, G. Autophagy interplay with apoptosis and cell cycle regulation in the growth inhibiting effect of resveratrol in glioma cells. PLoS ONE 2011, 6, e20849. [Google Scholar] [CrossRef]
  145. Xue, S.; Xiao-Hong, S.; Lin, S.; Jie, B.; Li-Li, W.; Jia-Yao, G.; Shun, S.; Pei-Nan, L.; Mo-Li, W.; Qian, W.; et al. Lumbar puncture-administered resveratrol inhibits STAT3 activation, enhancing autophagy and apoptosis in orthotopic rat glioblastomas. Oncotarget 2016, 7, 75790–75799. [Google Scholar] [CrossRef]
  146. Wang, J.; Qi, Q.; Feng, Z.; Zhang, X.; Huang, B.; Chen, A.; Prestegarden, L.; Li, X.; Wang, J. Berberine induces autophagy in glioblastoma by targeting the AMPK/mTOR/ULK1-pathway. Oncotarget 2016, 7, 66944–66958. [Google Scholar] [CrossRef] [Green Version]
  147. Hegazy, A.M.; Yamada, D.; Kobayashi, M.; Kohno, S.; Ueno, M.; Ali, M.A.; Ohta, K.; Tadokoro, Y.; Ino, Y.; Todo, T.; et al. Therapeutic strategy for targeting aggressive malignant gliomas by disrupting their energy balance. J. Biol. Chem. 2016, 291, 21496–21509. [Google Scholar] [CrossRef]
  148. Fan, X.; Khaki, L.; Zhu, T.S.; Soules, M.E.; Talsma, C.E.; Gul, N.; Koh, C.; Zhang, J.; Li, Y.M.; Maciaczyk, J.; et al. 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]
  149. 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]
  150. Rajakulendran, N.; Rowland, K.J.; Selvadurai, H.J.; Ahmadi, M.; Park, N.I.; Naumenko, S.; Dolma, S.; Ward, R.J.; So, M.; Lee, L.; et al. Wnt and Notch signaling govern self-renewal and differentiation in a subset of human glioblastoma stem cells. Genes Dev. 2019, 33, 498–510. [Google Scholar] [CrossRef]
  151. Wu, X.; Fleming, A.; Ricketts, T.; Pavel, M.; Virgin, H.; Menzies, F.M.; Rubinsztein, D.C. Autophagy regulates Notch degradation and modulates stem cell development and neurogenesis. Nat. Commun. 2016, 7, 10533. [Google Scholar] [CrossRef] [Green Version]
  152. Colella, B.; Faienza, F.; Carinci, M.; D’Alessandro, G.; Catalano, M.; Santoro, A.; Cecconi, F.; Limatola, C.; Di Bartolomeo, S. Autophagy induction impairs Wnt/β-catenin signalling through β-catenin relocalisation in glioblastoma cells. Cell Signal. 2019, 53, 357–364. [Google Scholar] [CrossRef]
  153. Sanchez-Diaz, P.C.; Chang, J.C.; Moses, E.S.; Dao, T.; Chen, Y.; Hung, J.Y. Ubiquitin carboxyl-terminal esterase L1 (UCHL1) is associated with stem-like cancer cell functions in pediatric high-grade glioma. PLoS ONE 2017, 12, e0176879. [Google Scholar] [CrossRef]
  154. Hussain, S.; Foreman, O.; Perkins, S.L.; Witzig, T.E.; Miles, R.R.; van Deursen, J.; Galardy, P.J. The de-ubiquitinase UCH-L1 is an oncogene that drives the development of lymphoma in vivo by deregulating PHLPP1 and Akt signaling. Leukemia 2010, 24, 1641–1655. [Google Scholar] [CrossRef]
  155. Hurst-Kennedy, J.; Chin, L.S.; Li, L. Ubiquitin C-terminal hydrolase l1 in tumorigenesis. Biochem. Res. Int. 2012, 2012, 123706. [Google Scholar] [CrossRef]
  156. Xiang, T.; Li, L.; Yin, X.; Yuan, C.; Tan, C.; Su, X.; Xiong, L.; Putti, T.C.; Oberst, M.; Kelly, K.; et al. The ubiquitin peptidase UCHL1 induces G0/G1 cell cycle arrest and apoptosis through stabilizing p53 and is frequently silenced in breast cancer. PLoS ONE 2012, 7, e29783. [Google Scholar] [CrossRef]
  157. Hussain, S.; Feldman, A.L.; Das, C.; Ziesmer, S.C.; Ansell, S.M.; Galardy, P.J. Ubiquitin hydrolase UCH-L1 destabilizes mTOR complex 1 by antagonizing DDB1-CUL4-mediated ubiquitination of raptor. Mol. Cell Biol. 2013, 33, 1188–1197. [Google Scholar] [CrossRef]
  158. Yan, C.; Huo, H.; Yang, C.; Zhang, T.; Chu, Y.; Liu, Y. Ubiquitin C-Terminal Hydrolase L1 regulates autophagy by inhibiting autophagosome formation through its deubiquitinating enzyme activity. Biochem. Biophys. Res. Commun. 2018, 497, 726–733. [Google Scholar] [CrossRef]
  159. Zhang, X.; Guo, L.; Niu, T.; Shao, L.; Li, H.; Wu, W.; Wang, W.; Lv, L.; Qin, Q.; Wang, F.; et al. Ubiquitin carboxyl terminal hydrolyase L1-suppressed autophagic degradation of p21WAF1/Cip1 as a novel feedback mechanism in the control of cardiac fibroblast proliferation. PLoS ONE 2014, 9, e94658. [Google Scholar] [CrossRef]
  160. Zhong, J.; Zhao, M.; Ma, Y.; Luo, Q.; Liu, J.; Wang, J.; Yuan, X.; Sang, J.; Huang, C. UCHL1 acts as a colorectal cancer oncogene via activation of the β-catenin/TCF pathway through its deubiquitinating activity. Int. J. Mol. Med. 2012, 30, 430–436. [Google Scholar] [CrossRef]
  161. Marjanovic Vicentic, J.; Drakulic, D.; Garcia, I.; Vukovic, V.; Aldaz, P.; Puskas, N.; Nikolic, I.; Tasic, G.; Raicevic, S.; Garros-Regulez, L.; et al. SOX3 can promote the malignant behavior of glioblastoma cells. Cell Oncol. (Dordr). 2019, 42, 41–54. [Google Scholar] [CrossRef]
  162. Zeng, X.; Ju, D. Hedgehog Signaling Pathway and Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 2279. [Google Scholar] [CrossRef]
  163. Maiti, S.; Mondal, S.; Satyavarapu, E.M.; Mandal, C. mTORC2 regulates hedgehog pathway activity by promoting stability to Gli2 protein and its nuclear translocation. Cell Death Dis. 2017, 8, e2926. [Google Scholar] [CrossRef]
  164. Cheng, W.; Zhang, C.; Ren, X.; Jiang, Y.; Han, S.; Liu, Y.; Cai, J.; Li, M.; Wang, K.; Liu, Y.; et al. Bioinformatic analyses reveal a distinct Notch activation induced by STAT3 phosphorylation in the mesenchymal subtype of glioblastoma. J. Neurosurg. 2017, 126, 249–259. [Google Scholar] [CrossRef]
  165. Liu, T.; Li, A.; Xu, Y.; Xin, Y. Momelotinib sensitizes glioblastoma cells to temozolomide by enhancement of autophagy via JAK2/STAT3 inhibition. Oncol. Rep. 2019, 41, 1883–1892. [Google Scholar] [CrossRef] [Green Version]
  166. Wang, G.; Zhang, J.; Dui, D.; Ren, H.; Liu, J. High mobility group box 1 induces the activation of the Janus kinase 2 and signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway in pancreatic acinar cells in rats, while AG490 and rapamycin inhibit their activation. Bosn. J. Basic Med. Sci. 2016, 16, 307–312. [Google Scholar] [CrossRef] [Green Version]
  167. Marampon, F.; Leoni, F.; Mancini, A.; Pietrantoni, I.; Codenotti, S.; Letizia, F.; Megiorni, F.; Porro, G.; Galbiati, E.; Pozzi, P.; et al. Histone deacetylase inhibitor ITF2357 (givinostat) reverts transformed phenotype and counteracts stemness in in vitro and in vivo models of human glioblastoma. J. Cancer Res. Clin. Oncol. 2019, 145, 393–409. [Google Scholar] [CrossRef]
  168. Ciechomska, I.A.; Przanowski, P.; Jackl, J.; Wojtas, B.; Kaminska, B. BIX01294, an inhibitor of histone methyltransferase, induces autophagy-dependent differentiation of glioma stem-like cells. Sci. Rep. 2016, 6, 38723. [Google Scholar] [CrossRef] [Green Version]
  169. Ferrucci, M.; Biagioni, F.; Ryskalin, L.; Limanaqi, F.; Gambardella, S.; Frati, A.; Fornai, F. Ambiguous Effects of Autophagy Activation Following Hypoperfusion/Ischemia. Int. J. Mol. Sci. 2018, 19, 2756. [Google Scholar] [CrossRef]
Figure 1. Identifying cancer stem cells in glioblastoma. Neural stem cells (NSCs) reside within the subependymal ventricular zone (SVZ), which represents the classic neurogenic niche of the adult brain. Within the SVZ, NSCs can undergo self-renewal or they can differentiate into neurons, astrocytes, and oligodendrocytes. The development of glioblastoma multiforme (GBM) depends on a small population of tumor cells known as glioma stem cells (GSCs). GSCs share several core properties of NSCs, such as stemness and sustained proliferation. GSCs that harbor tumor-initiating potential can be identified through specific markers such as CD133, Nestin, SOX2, NANOG, STAT3, Musashi, Bmi-1.
Figure 1. Identifying cancer stem cells in glioblastoma. Neural stem cells (NSCs) reside within the subependymal ventricular zone (SVZ), which represents the classic neurogenic niche of the adult brain. Within the SVZ, NSCs can undergo self-renewal or they can differentiate into neurons, astrocytes, and oligodendrocytes. The development of glioblastoma multiforme (GBM) depends on a small population of tumor cells known as glioma stem cells (GSCs). GSCs share several core properties of NSCs, such as stemness and sustained proliferation. GSCs that harbor tumor-initiating potential can be identified through specific markers such as CD133, Nestin, SOX2, NANOG, STAT3, Musashi, Bmi-1.
Ijms 20 03824 g001
Figure 2. Autophagy in Glioma Stem Cells (GSCs). During development, baseline autophagy guarantees neuronal differentiation and homeostasis within the neural stem cells (NSCs) niche. Impaired autophagy seems to be crucial for GSCs tumor initiation. In fact, when autophagy is impaired (central while circle), GSCs undergo uncontrolled self-renewal and rapid proliferation in the absence of differentiation. GSCs also infiltrate within the healthy brain parenchyma, thus displaying enhanced invasion and resistance to therapy, the two hallmarks of GBM aggressiveness.
Figure 2. Autophagy in Glioma Stem Cells (GSCs). During development, baseline autophagy guarantees neuronal differentiation and homeostasis within the neural stem cells (NSCs) niche. Impaired autophagy seems to be crucial for GSCs tumor initiation. In fact, when autophagy is impaired (central while circle), GSCs undergo uncontrolled self-renewal and rapid proliferation in the absence of differentiation. GSCs also infiltrate within the healthy brain parenchyma, thus displaying enhanced invasion and resistance to therapy, the two hallmarks of GBM aggressiveness.
Ijms 20 03824 g002
Figure 3. Molecular pathways at the crossroad between autophagy and glioma stem cells proliferation. An interdependency exists between autophagy and the foremost molecular pathways that contribute to GBM features by increasing stemness. For instance, hyperactive mTOR due to either nutrient/growth factor abundance, PTEN mutations, or UCKH1-related PDGFB-dependent phosphorylation of mTORC1, leads to autophagy suppression and mTOR-related transcriptional changes in the nucleus. UCHL1 also enhances mTORC2 stability thus potentiating activation of Akt signaling and mTOR hyperactivation. Again Hedgehog (HDGH) upregulation contributes to activating mTOR, suppressing autophagy and translocating HDGH within the nucleus. Likewise, JAK/STAT signaling leads to upregulation of STAT3, which impairs autophagy through HMGB. STAT3 may also contribute to Notch upregulation, which produces persistent transcriptional changes in the nucleus since its degradation is occluded when autophagy is impaired. Similarly, sustained Wnt/β-catenin activation occurs since the autophagy-dependent degradation of Dishevelled (DSL) is impaired. Altogether these events converge in transcriptional changes, which enhance GSCs self-renewal, proliferation, multi-potency, and migration. Rescuing autophagy through mTOR inhibitors such as rapamycin, curcumin, and resveratrol reverts these biochemical events while promoting GSCs differentiation.
Figure 3. Molecular pathways at the crossroad between autophagy and glioma stem cells proliferation. An interdependency exists between autophagy and the foremost molecular pathways that contribute to GBM features by increasing stemness. For instance, hyperactive mTOR due to either nutrient/growth factor abundance, PTEN mutations, or UCKH1-related PDGFB-dependent phosphorylation of mTORC1, leads to autophagy suppression and mTOR-related transcriptional changes in the nucleus. UCHL1 also enhances mTORC2 stability thus potentiating activation of Akt signaling and mTOR hyperactivation. Again Hedgehog (HDGH) upregulation contributes to activating mTOR, suppressing autophagy and translocating HDGH within the nucleus. Likewise, JAK/STAT signaling leads to upregulation of STAT3, which impairs autophagy through HMGB. STAT3 may also contribute to Notch upregulation, which produces persistent transcriptional changes in the nucleus since its degradation is occluded when autophagy is impaired. Similarly, sustained Wnt/β-catenin activation occurs since the autophagy-dependent degradation of Dishevelled (DSL) is impaired. Altogether these events converge in transcriptional changes, which enhance GSCs self-renewal, proliferation, multi-potency, and migration. Rescuing autophagy through mTOR inhibitors such as rapamycin, curcumin, and resveratrol reverts these biochemical events while promoting GSCs differentiation.
Ijms 20 03824 g003

Share and Cite

MDPI and ACS Style

Ryskalin, L.; Gaglione, A.; Limanaqi, F.; Biagioni, F.; Familiari, P.; Frati, A.; Esposito, V.; Fornai, F. The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies. Int. J. Mol. Sci. 2019, 20, 3824. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20153824

AMA Style

Ryskalin L, Gaglione A, Limanaqi F, Biagioni F, Familiari P, Frati A, Esposito V, Fornai F. The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies. International Journal of Molecular Sciences. 2019; 20(15):3824. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20153824

Chicago/Turabian Style

Ryskalin, Larisa, Anderson Gaglione, Fiona Limanaqi, Francesca Biagioni, Pietro Familiari, Alessandro Frati, Vincenzo Esposito, and Francesco Fornai. 2019. "The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies" International Journal of Molecular Sciences 20, no. 15: 3824. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20153824

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