Next Article in Journal
The Role of microRNAs in Inflammatory Bowel Disease
Previous Article in Journal
The Alpha/Beta-Hydrolase Fold Superfamily in Brassica napus: Expression Profiles and Functional Implications of Clade-3 BnABH Proteins in Response to Abiotic Stress
Previous Article in Special Issue
Insulin Receptor Substrate-2 Regulates the Secretion of Growth Factors in Response to Amino Acid Deprivation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis

by
Abigail J. Harland
and
Claire M. Perks
*
Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(10), 4749; https://doi.org/10.3390/ijms26104749
Submission received: 2 April 2025 / Revised: 9 May 2025 / Accepted: 12 May 2025 / Published: 15 May 2025
(This article belongs to the Special Issue The Role of the IGF Axis in Disease, 4th Edition)

Abstract

:
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation—most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent of a neurodevelopmental hierarchy, in which neural stem and progenitor markers are widely expressed by tumour stem-like cells. However, NSC fates and the cues that drive them are poorly understood. Studying the crosstalk within NSC niches may better inform our understanding of glioblastoma initiation and development. Insulin-like growth factor binding protein 2 (IGFBP-2) has a well-established prognostic role in glioblastoma, and cell-based mechanistic studies show the independent activation of downstream oncogenic pathways. However, IGFBP-2 is more commonly recognised as a modulator of insulin-like growth factors (IGFs) for receptor tyrosine kinase signal propagation or attenuation. In the adult human brain, both IGFBP-2 and IGF-II expression are retained in the choroid plexus (ChP) and secreted into the cerebral spinal fluid (CSF). Moreover, secretion by closely associated cells and NSCs themselves position IGFBP-2 and IGF-II as interesting factors within the NSC niche. In this review, we will highlight the experimental findings that show IGFBP-2 and IGF-II influence NSC behaviour. Moreover, we will link this to glioblastoma biology and demonstrate the requirement for further analysis of these factors in glioma stem cells (GSCs).

1. Introduction

Glioblastoma is a fatal brain cancer with limited therapeutic options and a survival time of 15–18 months [1,2]. The most common diagnostic age range is 55–85 years, with an incidence of approximately 3% in children [3]. Earlier intervention in disease pathology is hoped to lead to novel treatment approaches and better patient outcomes [4]. Of note, the overexpression of insulin-like growth factor binding protein 2 (IGFBP-2) in glioblastoma tumour tissue has been repeatedly reported and is associated with shorter patient survival [5,6,7,8,9]. In efforts to rationalise this prognostic effect, in vitro glioblastoma studies suggest that IGFBP-2 frequently functions independently of insulin-like growth factor (IGF) signalling [10,11,12,13]. Instead, functional motifs within the structure are thought to promote the survival, expansion and migration of immortalised glioblastoma cell lines [14]. However, glioblastoma is a poorly differentiated tumour. Progression is frequently compared to the processes that take place during pre- and postnatal neurodevelopment. Thus, there is good evidence to suggest that glioblastoma is a stem cell-driven cancer, propagated from a non-malignant neural stem cell origin [15]. Therefore, further to previously delineated observations of IGFBP2 overexpression in tumour tissue and immortalised cell lines, emerging evidence suggests additional roles for the maintenance of glioblastoma cells with stem cell properties—commonly referred to as glioma stem cells (GSCs) [16]. However, IGF-II must not be ignored. It is becoming increasingly clear that IGF2 expression surrounding neurogenic niches in the adult human brain (the subventricular zone (SVZ) or the subgranular zone (SGZ)) can influence cell fate [17]. IGF2 is an imprinted gene and therefore the paternal allele is expressed by most tissues, as well as the SGZ [18]. However, biallelic expression is present in the SVZ neurogenic niche [18]. The delineation of these differences has been related to contrasting functionalities, such that IGF-II can act in a paracrine manner as a mitogen in the SVZ, or in an autocrine manner in the SGZ as a survival factor [17]. Indeed, these findings could have implications for delineating the mechanisms that take place during glioblastoma development.
This review will focus on the functions of IGFBP-2 in the maintenance of neural stem cells (NSCs) and their malignant counterparts (GSCs). Due to the overexpression of IGFBP2 within tumour tissue, many studies focus on the functions of IGFBP-2 in glioblastoma. However, whilst the study of IGFBP-2 is more robust, IGF-II secretion into the cerebral spinal fluid (CSF) similarly positions it near key neurogenic regions. Therefore, despite a lack of study in GSCs, evidence from NSC experiments highlights IGF-II as a biologically plausible yet underexamined contributor to the glioblastoma microenvironment.

2. IGF and Insulin Axis Components

IGF and insulin signalling coordinate several biological processes with overarching outcomes including the modulation of metabolism and mitogenesis. The IGF/insulin signalling axis comprises ligands: insulin, IGF-I, IGF-II with variants (high-molecular-weight IGF-II), two structurally homologous transmembrane receptor tyrosine kinases (the IGF-I receptor (IGF-1R) and the insulin receptor (IR)), along with hybrid heterodimers (IR-A and IR-B) and hybrid tetramers (IGF-1R/IR, (Figure 1). IGF-2R, also known as the mannose-6-phosphate (M6P) receptor, is a type I transmembrane protein, lacks kinase activity and is structurally distinct from IR and IGF-1R [19]. IGF-2R binds to several ligands including those containing mannose-6-phospate to mediate lysosomal enzyme trafficking and degradation, as well as IGF-II, resulting in signal attenuation and growth modulation [20,21]. The IGF system additionally includes IGFs distributed between the cell surface (IGF-II/the mannose-6-phosphate (M6P) receptor) and secreted into the extracellular environment. Six IGF-binding proteins (IGFBP-1 to 6) rapidly bind IGFs, increasing their half-life and relative bioavailability [22,23,24,25]. Notably, and in the context of the current review, the binding affinity between IGFBP-2 and IGF-II is 10–20-fold greater than that of IGF-I [26].
For an extensive review of IGF and insulin signalling in physiology and disease, please refer to the review by LeRoith et al. (2021) [27].
The early identification and description of IGFBPs was as IGF carrier proteins without independent biological activity [31]. By 1991, each of the six IGFBPs had been identified and discriminated via differing molecular weights. Sources included human serum, CSF, rat serum and conditioned media (CM) from the culture of human cells [32,33,34,35,36,37,38,39]. IGFBP-1, 2, 4 and 6 bind IGFs in a binary fashion, forming complexes of 30–40 kDa and increasing their half-life from 10 to 30–90 min [24]. Greater half-life extension to 12–20 h is achieved by ternary complexing of IGFs with IGFBP-3/5 and the acid-liable subunit [24,40,41,42]. The binding affinity of IGFBPs to IGF-I and IGF-II is greater than between the ligands and receptors, thus modulating ligand–receptor binding and activation [43]. IGFBPs provide a further layer of sophistication to the IGF axis, due to spatial and temporal tissue expression, as well as IGF binding affinity. For IGF release, IGFBPs undergo fragmentation via extracellular proteases with different specificities such as pregnancy-associated plasma protein-A (PAPP-A) or matrix metalloproteinase 9 (MMP-9), which target sites within the central linker domain of IGFBP-2, 4 and 5 [44,45,46,47]. Further subtle modulation of IGF activity can be achieved through high-affinity extracellular matrix (ECM)-IGFBP binding which can either reduce or potentiate IGF signalling [48].

3. IGFBP-2, IGF-II and the Adult Human Brain

In the 1990s, immunohistochemistry (IHC) and in situ hybridisation (ISH) studies sought to delineate the roles of IGFs throughout embryogenesis, with many reporting distinct distribution patterns [49,50]. Of particular interest was the developing central nervous system (CNS), which is tightly regulated by growth factors including IGFs [51,52]. For example, widespread embryonic expression of IGF1 can be detected in neurones and glial cells, with region-specific expressions including the olfactory bulb [50,53,54,55,56,57,58]. Postnatally, peak IGF1 expression coincides with rapid neurogenesis, neuronal proliferation and myelination in the second week after birth, with the highest detectable levels in the olfactory bulb and hippocampus [54,59,60,61]. IGF2 on the other hand is expressed at its highest levels prenatally with widespread messenger ribonucleic acid (mRNA) expression including the choroid plexus (ChP), leptomeninges, hypothalamus and floor of the third ventricle [53,62]. In adults, IGF-II is retained at higher levels than IGF-I, with biallelic expression in the meninges, ChP and vascular endothelial cells [18,63,64,65]. IGF2 can also be monoallelically expressed by SGZ NSCs [17,18]. The differences between biallelic expression in the SVZ niche and monoallelic expression in the SGZ appear to be of functional importance, with mitogenic paracrine outcomes in the SVZ, and cell survival autocrine outcomes in the SGZ [18]. Moreover, a deficiency in IGF2 in vivo appears to impair SVZ NSC proliferation [18]. In addition to mature IGF-II, higher-molecular-weight variants which are stable but have undergone incomplete processing of the prohormone have been detected in adult human serum [66]. It is worth noting that high-molecular-weight variants predominate in several malignancies and are considered the principal circulating form in tumour-associated hypoglycaemia, as reviewed by Scalia et al. (2023) [67]. In 1985, Haselbacher et al. identified several molecular weight forms of IGF-II including 13 kDa, 26 kDa and 38 kDa in addition to mature 7.5 kDa IGF-II across 24 regions of the human brain, and high-molecular-weight IGF-II had previously been detected in human spinal fluid [68,69]. Despite the 9 kDa variant in CSF exhibiting biological activity via radioimmunoassay, the effects on the human brain are not yet fully understood [68].
The regulatory pathways underlying the control of IGF component levels in the brain for normal homeostasis are relatively understudied. Current knowledge of adult IGF induction is mainly derived from brain injury and pathology [70,71]. Spatial and temporal expression and abundance of IGFBPs have also been closely studied to determine the neuroanatomical distribution and synchronous effects on IGF-mediated CNS signalling [71,72,73,74]. In the adult human brain, IGFBP-2 is the most abundant IGFBP and is secreted into the CSF from ChP [75,76]. Moreover, functional IGFBP-2 produced in the brain is distinct to that of circulating plasma following the establishment of the CSF–blood barrier [76,77]. IGF-II expression is also retained in the ChP, and free IGFs can be actively transported across the blood–brain barrier (BBB) into adulthood [75,78].

3.1. Neural Stem Cells Are Retained in the Adult Human Brain

Cells of the CNS that maintain neurogenesis from embryogenesis into adulthood in mammals were first reported in the 1960s using thymidine-H3 autoradiographic labelling in rat and cat brains [79,80]. Adult NSCs are believed to arise from embryonic radial glial cells (RGCs) via transient foetal brain structures such as the ganglionic eminences [81,82,83,84]. Bromodeoxyuridine labelling of SGZ and SVZ human brain tissue postmortem by Eriksson et al. (1998) showed that progenitor cells resided in both these regions and were capable of neurogenesis [85]. Further analysis of NSCs from each region found that NSCs in the SVZ (or type B cells) were reminiscent of astroglia, expressing markers of self-renewal (sex determining region Y-box 2 (SOX2) and nestin), markers associated with the astrocyte lineage (glial fibrillary acidic protein (GFAP), glutamate aspartate transporter, tailless and vascular cell adhesion molecule 1 [86,87,88]. NSCs of the SVZ have been shown to give rise to neurones and oligodendrocytes [86,89]. On the other hand, NSCs identified in the SGZ (radial astrocytes/type I cells), defined by SOX2, Nestin and GFAP expression, could self-renew and mainly function as precursors for postnatal neurogenesis [90,91,92,93] (Figure 2A).
Much of the evidence for NSC development in the mammalian brain has been derived from rodent studies [94]. Type I cells in the SGZ have the capacity to generate neurones and dentate-granule cells, contributing to spatial learning and short-term memory [95,96]. However, in the SVZ, NSCs form type C multipotent progenitors, which in turn can form neuroblasts (type A cells) that can migrate to the olfactory bulb along the rostral migratory stream to generate interneurons [86,94]. In humans, the SVZ is a 3–5 mm thick region that also contains NSCs [97]. However, in contrast to rodents, there is no evidence for the rostral migratory stream [98]. Moreover, olfactory-bulb-localised interneuron production has been detected in human infants up to the age of 18 months [99] (Figure 2B).
Figure 2. Neural stem cell lineage tree and location in human and mice. (A) Neuroepithelial cells (NEPCs) arise during neural tube formation and can undergo symmetric division, supporting development and retaining their population. The differentiation of NEPCs and adoption of astroglial characteristics give rise to early and late radial glial cells (RGCs). RGCs undergo asymmetric division, giving rise to neural stem cels (NSCs) (type B cells in the subventricular zone (SVZ) or type I cells in the subgranular zone (SGZ)) which persist postnatally/into adulthood and have the potential to form differentiated progenies. NSC differentiation gives rise to multipotent progenitor cells in the SVZ (type C) and SGZ (type II cells). Downstream differentiation into neural progenitor cells (NPCs)/type C cells, oligodendrocyte progenitor cells (OPCs) and astrocyte progenitor cells (APCs) has the capacity to form progenies of neurones, oligodendrocytes, astrocytes and ependymal cells throughout embryonic brain development. OPCs can stay undifferentiated and continue to proliferate in the adult brain. NSCs can also directly differentiate into mature cell types. NSCs in the SVZ generate type C cells which can form type A cells. Type A cells migrate into the olfactory bulb for neurogenesis, whereas SGZ NSCs generate neurones locally in the dentate gyrus [86]. Adapted from [100]. (B) Schematic showing the location of adult NSCs in the SVZ and SGZs of human and mice brains. Adapted from [101,102]. Created with BioRender.com.
Figure 2. Neural stem cell lineage tree and location in human and mice. (A) Neuroepithelial cells (NEPCs) arise during neural tube formation and can undergo symmetric division, supporting development and retaining their population. The differentiation of NEPCs and adoption of astroglial characteristics give rise to early and late radial glial cells (RGCs). RGCs undergo asymmetric division, giving rise to neural stem cels (NSCs) (type B cells in the subventricular zone (SVZ) or type I cells in the subgranular zone (SGZ)) which persist postnatally/into adulthood and have the potential to form differentiated progenies. NSC differentiation gives rise to multipotent progenitor cells in the SVZ (type C) and SGZ (type II cells). Downstream differentiation into neural progenitor cells (NPCs)/type C cells, oligodendrocyte progenitor cells (OPCs) and astrocyte progenitor cells (APCs) has the capacity to form progenies of neurones, oligodendrocytes, astrocytes and ependymal cells throughout embryonic brain development. OPCs can stay undifferentiated and continue to proliferate in the adult brain. NSCs can also directly differentiate into mature cell types. NSCs in the SVZ generate type C cells which can form type A cells. Type A cells migrate into the olfactory bulb for neurogenesis, whereas SGZ NSCs generate neurones locally in the dentate gyrus [86]. Adapted from [100]. (B) Schematic showing the location of adult NSCs in the SVZ and SGZs of human and mice brains. Adapted from [101,102]. Created with BioRender.com.
Ijms 26 04749 g002

3.2. The Neural Stem Cell Secretome: IGFBP-2 and IGF-II

The NSC niche is a term used to describe the specialised microenvironment that supports cell behaviours: self-renewal, proliferation and differentiation. For example, cells that may directly influence NSCs through cellular contact, autocrine/paracrine communication include NSCs themselves, astrocytes, ependymal cells, type C cells, ChP epithelial cells, vascular endothelial cells and microglia [18,103,104].
NSC secreted factors or extracellular vesicles could result in autocrine and/or paracrine regulation of other NSCs or cell types in the brain with diverse biological effects depending on the molecular features of the recipient cell [105,106,107]. As reviewed by Dause et al. (2022), the secretome of NSCs varies across neurodevelopment [105]. For example, secreted molecules such as sonic hedgehog (Shh) are required for the proliferation and differentiation of RGCs in the ventricular zone after neural tube formation [108]. However, NSCs in adults require Shh signalling for self-renewal and maintenance [109]. Thus, molecules such as these are spatially and temporally regulated for tightly controlled localised signalling required at different developmental stages.
Historically, trophic factors, chemokines and cytokines have been measured using pre-selected antibody arrays or liquid chromatography–tandem mass spectrometry (LC-MS/MS) of CM produced by cells grown in serum-free conditions [110,111]. In a study by Cervenka et al. (2021), proteomic analysis of the CM collected from NSCs (derived from human embryonic stem cells (ESCs)) was compared with those subjected to growth factor withdrawal for different durations [110]. The group found that subject to differentiation, NSC secreted protein profiles undergo significant global changes. Notably, following 21 days of epidermal growth factor (EGF) and fibroblast growth factor (FGF2) withdrawal, IGFBP-2 showed a −2.6-fold change relative to day 0, supporting the notion that IGFBP-2 abundance is decreased upon neuronal differentiation [110]. IGF-II was not detected in this study. Previously, the inclusion of IGFBP-2 in pre-selected antibody arrays has led to detection in CM from NPCs (derived from human ESCs) [111]. In contrast, other analyses of neurosphere-derived CM using mass spectrometry have not detected IGFBP-2 or IGF-II. Instead, molecules such as apolipoprotein E (APOE), DSD-1 proteoglycan and chondroitin sulfate proteoglycan were detected, required for stimulating neurosphere formation and proliferation in vitro [112].
Improved approaches have recently emerged, integrating both in vivo and in vitro assessment to provide a more comprehensive picture of secretomes in both contexts [113]. Lee et al. (2012) created secretory molecule expression profiles from isolated cells: SVZ NSCs, type C cells, ependymal cells, astrocytes and vascular cells [103]. Notably, in this study, previously identified APOE was highly expressed by all niche cells analysed. However, IGFBP2 showed specific expression in ChP cells. Surprisingly, given its expression in the ChP, IGF2 was not reported in any of these cell types in this study [103]. Other approaches using protein arrays and LC-MS/MS have been used to characterise novel factors and facilitate comparison between transcript/protein abundance. Using these approaches to analyse mouse-derived hippocampal NSCs, Denninger (2020) revealed many proteins that have previously remained undetected [114]. Notably, three IGF binding proteins (IGFBP-2, IGFBP-3 and IGFBP-5) showed high signal intensity in antibody arrays and unbiased LC-MS/MS. Further enzyme-linked immunosorbent assays of NPSC CM and lysates showed particularly high concentrations of IGFBP-2 (10-fold greater than vascular endothelial growth factor (VEGF) which is a key protein for NPSC maintenance) [115,116]. Surprisingly, despite the high expression of IGF2 previously detected in hippocampal NSCs, IGF-II was not detected. Despite these improved approaches, technical limitations remain in studying the NSC niche. Notably, many of the bioinformatics tools employed are still limited by profile size and may still not capture many of the essential factors required for NSC identity [103].

3.3. The SVZ Niche, IGFBP-2 and IGF-II

In the adult human brain, one of the major sites for the retention of IGF-II and IGFBP-2 abundance is the ChP [76]. The ChP is a tissue present in the third, fourth and lateral ventricles of the brain from which CSF is secreted [117,118]. This structure is highly vascularised and constitutes the blood–CSF barrier to restrict solute flow via epithelial tight junctions [117] (Figure 3). CSF provides a rich source of mitogens and growth factors to the brain with a unique biochemistry to that of plasma [119]. Due to the close spatial proximity of the SVZ to the ChP, adult SVZ NSC processes can contact the CSF via apical primary cilia [120,121,122] (Figure 3). Indeed, it has been shown that pinwheel structures are set up in which ependymal cells surround apical cilia of type B NSCs [121]. Therefore, the CSF must be considered as an extremely important component of the NSC niche. Notably, CSF blockage or composition changes inhibit the proliferation of neural precursors, inhibit neuronal migration and result in abnormal cortex development [123,124,125].
Many early studies sought to delineate the complexity of the CSF proteome in rats and humans [120,128,129,130,131]. However, the first to show that the fate decisions of neural progenitors could be mediated by CSF-derived IGFs was published in 2011 by Lehtinen et al. [120]. The authors described intricate mechanisms by which protein associated with lin 7 (Pals1) and phosphatase and tensin homolog (PTEN) spatially regulate the localisation of the IGF-1R to the SVZ NSC apical complex in mice, thus governing the transduction of signals from the CSF [120]. Other examples of essential cell polarity proteins/mediators of extrinsic IGF-I/insulin signalling are α-E-catenin in skin keratinocytes and β-catenin in oligodendrocyte precursor cells (OPCs). Furthermore, IGF-II was identified as the predominant CSF-derived IGF, with 10.7-fold higher mRNA expression in adult rat ChP tissue than the cortex, potentiating NSC IGF-1Rβ, Akt, mitogen-activated protein kinase (MAPK) phosphorylation and neurosphere maintenance in vitro. Notably, CSF derived from IGF-II null mice was insufficient to stimulate the proliferation of cortical progenitors, resulting in an overall reduction in brain size (24%) [120]. Further CSF screening revealed the presence of well-established proteins involved in morphological patterning and neurogenesis including Wnts, bone morphogenic proteins (BMPs), transforming growth factor (TGF)-β factors and retinoic acid [120,132,133,134,135].
In 2015, Lun et al. investigated regional differences between choroid plexi and derived CSF from the lateral and fourth ventricles [136]. Ribonucleic acid (RNA)-sequencing analysis of bulk ChP tissue demonstrated vast region specificity with 684 differentially expressed genes across the two sites. At E18.5, IGF2 was upregulated in the fourth ventricle ChP and IGFBP2 was expressed in both [120,136,137]. Furthermore, IGF-II and IGFBP-2 were in the top 25 abundantly secreted proteins from ChP epithelial tissue [136]. An investigation of adult primate and human ChP samples also showed a high abundance of Shh and IGF-II [136]. Finally, in 2016, independent transcriptomic analysis of ChP tissue and the corresponding secretome in adult mice further identified growth factors, hormones and ECM remodelling proteins including FGF2, VEGF-A, IGF-I, -II, IGFBP-2,-3, SERPINE1, tissue inhibitor matrix metalloproteinase 1 (TIMP1) and MMP-8 [122]. The composition of factors was dynamically regulated with age [122]. Unsurprisingly, given essential prenatal neurogenic effects, IGF-I was one of the most enriched factors in the ChP secretome of 2-month-old mice [122]. However, a 14% decline in IGF-I has been shown to occur per decade [138,139]. Furthermore, the levels of IGF-II also reduce with age [140]. The group concluded that intricate combinations of secreted factors change over time and may reflect dynamic NSC/progenitor fates [122].
Further studies to analyse the effects of IGF-II on NSC fates have corroborated other findings in which IGF-II facilitates NSC expansion and self-renewal [17,120,122,139]. In NSCs isolated from the hippocampus dentate gyrus, IGF2 appears to be highly expressed and can influence neurogenesis in an autocrine manner [17]. In contrast, extremely low measurements of IGF2 in SVZ-derived NSCs confirm that it is not endogenously expressed in these cells [17]. These results corroborate the fact that IGF-II appears to function in a paracrine manner in the SVZ niche—secreted from the ChP epithelium and available to NSCs via the CSF [17,120,139]. IGF-II is also secreted from the leptomeninges and vasculature of the brain [17,18]. In contrast to studies in hippocampal NSCs—in which IGF-II was mainly shown to activate IGF-1R to regulate proliferation, by knocking out IGF-1R, Ziegler et al. (2012) showed that IGF-II can promote stemness of neural stem/progenitor cells (NSPCs) through the activation of IR-A—distinct to the actions of IGF-I [17,139]. A comparison of outcomes from IGF-I and IGF-II treatment revealed that NPSCs treated with IGF-II had increased expansion and increased mRNA expression of OCT4, SOX1 and fatty acid binding protein 7 (FABP7) [139]. By comparison, it is the interaction between IGF-II and other imprinted genes that has more recently been identified as a major influence for differentiation and lineage specification [141]. Lozano-Urena et al. (2023) showed that adult NSCs treated with recombinant IGF-II showed reduced stemness, and under differentiation conditions (mitogen withdrawal and addition of 2% foetal bovine serum (FBS)) promoted terminal differentiation [141]. Mechanistic analysis revealed that IGF-II activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway mediates expression of the maternal Cdkn1c allele, inducing terminal differentiation under these conditions. Therefore, the retention and maintenance of the NSC pool may be reliant upon the interaction between IGF-II and Cdkn1c [141].
Despite a significant decrease by the third week after birth, IGFBP-2 levels remain the highest of all IGFBPs in the human CNS, particularly in the ChP and meninges [75]. However, thus far there are just two publications that study the effects of recombinant IGFBP-2 on NSCs. In the study by Wang et al. (2017), the treatment of a mouse NSC line with recombinant IGFBP-2 enhanced in vitro proliferation [142]. Moreover, the group claimed that IGFBP-2 treatment inhibited differentiation to the neuronal lineage but enhanced astrocytic differentiation [142]. Despite these findings, the culture of NSCs in this study included the use of 10% FBS—a differentiation enhancer which is typically omitted from stem cell culture systems [142]. In 2023, studies by Shahin et al. showed that IGFBP-2 can be detected in the CM of adult mouse-derived SVZ NSCs using LC-MS/MS. In this study, they proposed that the effects of self-secreted IGFBP-2 on SVZ NSCs are redox-dependent [143]. NSC proliferation and differentiation have previously been shown in response to reactive oxygen species (ROS); however, it appears that redox-sensitive proteins may mediate specific effects [144,145]. Therefore, the presence and amount of ROS present in the microenvironment may be vital for influencing the balance between self-renewal and differentiation. The increased abundance of ROS was shown to result in the oxidation of cysteine 43 in the IGFBP-2 structure, increasing its bioavailability and enhancement of self-renewal—measured via increased neurosphere formation [143]. They additionally uncovered that the secretory pathway of IGFBP-2 may be redox-dependent, such that addition of hydrogen peroxide to NSC cultures in vitro resulted in greater detectable levels in the CM [143]. Therefore, the amount of ROS present in the microenvironment may be vital for the ability for IGFBP-2 to influence the balance between self-renewal and differentiation, and as such, studying the influence of IGFBP-2 under hypoxic conditions would be interesting. Lastly, analysis of IGFBP-2 binding to IGF-I and IGF-II revealed no redox dependent changes, and therefore the role of IGFBP-2 in regulating NSC maintenance and fates in this study was found to be independent of these ligands.

4. Glioblastoma: Identifying the Cell of Origin

The clinical presentation of glioblastoma often takes place at advanced stages of tumour development due to non-specific symptoms such as headaches, fatigue, nausea and vomiting [146,147]. Therefore, the time to diagnosis from symptom onset is <6 months in 85% of patients [148]. Previously, secondary glioblastoma (isocitrate dehydrogenase (IDH) mutant) was used to describe a tumour that had progressed from a lower-grade neoplasm [149,150]. However, the reclassification of glioblastoma in 2021 by the World Health Organisation (WHO) as a separate entity (only including IDH wildtype tumours) has highlighted the de novo nature of the tumour, and streamlined approaches to identify the tumour-initiating cell [2,151]. It is hoped that identifying the mechanisms that regulate tumour initiation, propagation and treatment evasion could lead to approaches to prevent and treat glioblastoma.
Leukaemia cells capable of tumour generation and mouse lethality were first observed in 1937 followed by the identification of leukaemia cells with enhanced self-renewal, believed to arise from malignant transformation of a primitive hemopoietic stem cell [152,153]. Subsequent revelations that cancer stem cells (CSCs) could be identified and isolated from breast, prostate, colorectal, pancreatic and brain cancer took place in the early 2000s [154,155,156,157,158,159,160,161]. In glioblastoma, these cells are now widely referred to as GSCs.
Despite this, the cellular origin of glioblastoma is still regarded as an open question. Rapid evolution and dynamics driving vast heterogeneity can quickly mask the original genetic features of the tumour. Therefore, there are several candidate untransformed cell types with the potential to propagate a tumour following oncogenic transformation, including NSCs, OPCs and astrocytes [162,163]. It is worth noting that studies such as those by Bachoo et al. (2002) showed that NSC and astrocytic transformation via deletion of the Ink4a/Arf locus (that encodes Retinoblastoma (RB) and p53), and epidermal growth factor receptor (EGFR) overexpression yielded tumours phenotypically similar to high-grade gliomas in vivo [162]. However, it remains to be determined whether the genetic manipulation of mature cells in vitro is reminiscent of the events that take place during glioblastoma development [164]. The delineation of early disease markers for detection or disease prevention may rely on identification of the origin cell.

Evidence for Neural Stem Cells as the Glioblastoma Cell of Origin

Transcriptional and genetic evidence suggests that malignant alterations of NSCs could lead to the growth of gliomas. It is increasingly apparent that subsets of cells isolated from glioblastoma are genetically and transcriptionally regulated by cues associated with immature and progenitor cell populations [16,160,165]. Genome-wide CRISPR-Cas9 screening of 10 patient-derived GSCs and two NSCs by Macleod et al. (2019) revealed that despite the diverse molecular alterations of glioblastoma, a core gene set governs both GSC and NSC maintenance and growth including SOX genes, oligodendrocyte transcription factor 2 (OLIG2) and SALL1 [166,167].
Several studies have shown that combinations of TSG mutations/allele deletions (tumour protein p53 (TP53), Neurofibromatosis type 1 (NF1), PTEN and RB1) in the NSCs of mouse models lead to the development of high-grade glioma [168,169,170,171]. Follow-up studies by Llaguno et al. (2019) confirmed that NF1, TP53 and PTEN allele deletions in early-stage progenitors were most likely to lead to glioblastoma development [172]. By inducing the same gene allele deletions in neuroblasts and terminally differentiated neurones reminiscent of adult neurogenesis, they found that early-stage cells (NPCs) were more likely to undergo malignant transformation than neuroblasts and differentiated neurones, with OPCs least resistant overall [172]. Indeed, it has been suggested that OLIG2-positive OPCs, which make up the largest group of dividing cells in the adult brain, are a major progenitor for glioma and medulloblastoma formation [163,171,172,173,174,175,176]. Moreover, OPCs are the major cell type produced by SVZ-NSCs, and markers (OLIG1/2, platelet-derived growth factor receptor (PDGFRα) and NG2) have been repeatedly recorded histologically in human gliomas and those induced in mouse models [173,177,178,179]. Moreover, by carrying out lineage tracing from an NSC with TP53 and NF1 deletions, Liu et al. (2011) showed that despite retaining neural and glial lineage formation capacity, only hyperproliferative-derived OPCs resulted in tumour formation [163].
In 2018, Lee et al. published a study showing that 56.3% of NSCs isolated from the SVZs of 26 patients with temporal lobe-located IDH-wildtype glioblastoma exhibited low-level matching driver mutations to the tumour (Figure 4) [180]. Notably, TERT promoter mutations were detected at much higher rates in NSCs from patients with IDH-wildtype glioblastoma than controls. Furthermore, the induction of clinically relevant driver mutations—TP53, PTEN and EGFR—in SVZ cells led to the development of brain tumours that were analogous to high-grade proliferative gliomas in 9/10 mouse models [2,180]. Deep whole exome sequencing of SVZ samples away from the tumour mass is the most convincing evidence to date that the SVZ-NSC population in the adult human brain is a source for glioblastoma development [180]. Furthermore, Mendelian randomisation studies suggest that TERT promoter mutations may be an early indicator of the ability of NSCs to avoid replicative senescence, instead prolonging self-renewal capacity and the likelihood of further mutational acquisition [181,182]. It is worth noting that SGZ-NSCs appear to be less susceptible to malignant transformation—reviewed by Fontan-Lozano et al. (2020) [183].
Glioblastomas are closely associated with the SVZ in 50–60% of cases and manifest a more aggressive disease [97,184,185,186]. Moreover, these tumours are associated with a shorter survival post-resection [97,185,187,188,189,190]. Brain swelling and enhanced CSF brain volume confer a reduced OS of glioblastoma patients [191]. SVZ-directed radiotherapy post-resection results in significant increases in overall survival (OS) and progression-free survival (PFS) [192]. Furthermore, a retrospective review of glioblastoma tumours from the initial diagnostic MRI and resulting clinical outcome data, often referred to a “Lim classification”, has prompted subdivision into type (I–IV) tumours based upon the degree of SVZ involvement [185,189]. Those patients with SVZ-associated disease display more rapid tumour recurrence and a decreased OS and PFS [185,189,193]. In response to these observations, a call for intensive research into “SVZ-associated glioblastoma” and translation into clinical decision making was made in 2016 by Smith et al. [97].
The repeated isolation of well-established CSF-derived morphogens and growth factors including FGF2, EGF, Shh, retinoic acid and Wnt5a in combination with less-well characterised components such as IGFBP-2 and IGF-II underscores the importance the ChP secreted niche [194,195,196,197,198]. It is not unreasonable to suggest that alterations to the CSF milieu as a result of brain malignancy or injury may be a major source of inflammatory or tumorigenic signals. Indeed, in 2022, it was shown that gliomas in close proximity to the lateral ventricle cause ependymal barrier disruption and increased tumour–CSF contact [186] (Figure 5).

5. IGFBP-2, IGF-II and Glioblastoma

5.1. IGFBP-2 and Glioblastoma

Early microarray analysis of glioblastoma by Rickman et al. (2001) revealed 360 unique genes when compared with pilocytic astrocytoma including IGFBP2, murine double minute 2 (MDM2), CD44 and cyclin-dependent kinase 4 (CDK4) [204]. In the early 2000s, widespread gene expression profiling to assist histological tumour evaluation potentiated studies evaluating gene predictors of glioma malignancy and patient survival [205,206,207,208]. In 2007, the combined absence of IGFBP-2 and IQ motif containing GTPase-activating protein 1 (IQGAP1) protein expression in glioblastoma patient tissue was shown to correlate with longer survival (>3 years) in studies by McDonald et al. (2007) [209]. Furthermore, in 2010, Colman et al. reviewed transcriptional findings from four independently published glioblastoma datasets [210]. This, in combination with analysis of glioblastoma formalin-fixed paraffin-embedded tissue samples identified a nine-gene signature including IGFBP-2, chitinase-3-like protein 1 (CHI3L1)/YKL-40, galectin 3 (LGALS3) and OLIG2 that was predictive of worse clinical outcomes [210]. Unsurprisingly, poorly differentiated tumours expressing nestin and CD133 were also associated with the nine-gene signature and worse responses to current treatment regimens [210]. Further molecular profiling of 1122 diffuse glioma samples by Ceccarelli and colleagues in 2016 revealed IGFBP-2 protein elevation in glioblastoma with EGFR, P-EGFR and P-Akt, when compared with lower-grade gliomas (2016 CNS WHO grades II and III) [211]. A prognostic relationship between high-grade glioblastoma, IGFBP2 expression and the incidence of TERT promoter mutation was further described by Yuan et al. (2019) [9]. The most prevalent TERT promoter mutation recorded in this study was C228T—previously suggested as an early event in the transformation of NSCs [9,180].
Within the last 5 years, efforts to stratify and optimise glioblastoma patients for predicted responses to therapy by Yin, Prasad, Yu and colleagues independently identified IGFBP-2 in gene signatures (with under 20 genes) associated with worse outcomes [212,213,214]. The identification of such gene signatures with predictive and prognostic utility is hoped to drive the development of novel therapeutic options for patient subsets or enable the gene signatures to act as therapeutic targets themselves. Furthermore, an assessment of IGFBP expression levels across 19 different cancer types deposited by The Cancer Genome Atlas in 2023 found that IGFBP2 mRNA levels were significantly associated with the prognosis of low-grade glioma (LGG) and glioblastoma (p-values: 8.3 × 10−33 and 1.2 × 10−3, respectively) (hazard ratios (95% confidence interval): 1.55 (1.44,1.68) and 1.24 (1.08, 1.40), respectively) [215].
Many mechanistic studies of IGFBP-2 thus far have been carried out in immortalised glioblastoma cell lines such as U251, U87 and T98G due to their ease of use and relatively low cost to maintain. However, the addition of animal serum into culture media and in vitro passaging facilitate the acquisition of genetic and epigenetic alterations over time, unlikely to maintain the gene expression profiles present in the glioblastoma from which they were derived [165]. Moreover, it is important to note that despite studies showing the low expression of IGFs from these glioblastoma cells themselves, the inclusion of animal serum introduces IGFs and other growth factors, confounding the ability to determine whether the effects of IGFBP-2 in these studies are IGF-independent [216].
In 2014, Han et al. showed that the exogenous delivery of IGFBP-2 to U87, SU3 and U251 cells promoted invasion and proliferation, whilst endogenous upregulation was specific to increased invasive potential [14]. The proliferative and invasive effects upon recombinant human IGFBP-2 dosing were attributed to integrin β1, downstream extracellular signal-related kinase (ERK) phosphorylation and nuclear translocation of IGFBP-2 [14]. The study additionally showed that exogenous IGFBP-2 integrinβ1/ERK pathway signalling resulted in abrogation of temozolomide (TMZ) activity, which was not replicated upon endogenous overexpression/knockdown. In another study, exogenous activation of integrin α5β1, downstream focal adhesion kinase (FAK)/ERK and JUN NH2 terminal kinase (JNK) phosphorylation, and subsequent transcriptional upregulation of VE cadherin (CD144) and MMP2 in U87, and primary glioblastoma cells was shown by Liu et al. [11]. In agreement with studies by Han et al. (2014), IGFBP2 knockdown in high-IGFBP2-expressing U251 cells decreased IGFBP-2 secretion levels and cellular migratory capacity, but did not affect proliferation [11,14]. Similarly, the interaction between IGFBP-2 and integrin α5β1 in SNB19 glioblastoma cells overexpressing IGFBP-2 was shown to activate JNK with downstream cell migration enhancement [12]. Ingenuity pathway analysis of the Repository for Molecular Brain Neoplasia Data (Rembrandt) by Holmes et al. also showed that cellular migration and invasion pathways including integrin linked kinase (ILK) and nuclear factor kappa-light-chain enhancer of activated B cells (NF-ΚB) are associated with IGFBP-2 [10]. In vitro knockdown studies showed that ILK and NF-ΚB activation was downstream of IGFBP-2-integrin α5β1 binding and resulted in the transcription of invasion-related genes [10]. Additionally, in 2016, following delineation in breast cancer, Patil et al. showed that the association between the arginine–glycine–aspartate (RGD) domain of IGFBP-2 and integrins could lead to the stabilisation of nuclear β-catenin, contributing to tumour growth [217,218]. An RGD containing the C-terminal but not N-terminal fragment of IGFBP-2 led to FAK activation and inactivation of glycogen synthase kinase (GSK3β)—required for β-catenin phosphorylation and degradation via the proteosome [217]. Upstream IGFBP-2 activation of Wnt/β-catenin signalling has more recently been shown to induce EMT in hepatocellular carcinoma [219]. Nuclear accumulation of β-catenin in glioblastoma cells led to the upregulation of transcriptional targets including Oct-4, MMP-2, Nanog and c-Myc [217].
Despite a wealth of research that indicates that IGFBP-2 exhibits oncogenic properties within glioblastoma, the specific effect of IGFBP-2 on the stem cell compartment (NSCs and GSCs) is sparse. Quantitative polymerase chain reaction (qPCR) analysis of GSCs, normal brain tissue and differentiated glioblastoma cell lines by Hsieh et al. (2010) showed that IGFBP2 was overexpressed in GSCs independently of IGFBP5, IGF1 and IGF1R [220]. Moreover, parallel secretome profiling between serum-cultured GSCs and their stem-like equivalents was recently investigated by Robilliard et al. (2022) [221]. The authors suggest how imperative different cellular states may be in modulating the surrounding immune microenvironment. High expression of immune modulatory genes including IGFBP2 was detected in serum-cultured cells/GSCs via NanoString analysis [221] Moreover, 5–50-fold higher levels of secreted proteins were detected from serum-derived cultures. However, simultaneous proteome profiling of cells cultured in both serum and serum-free conditions detected an elevated IGFBP-2 abundance in GSC cultures. Since IGFBP2 and VEGFA mRNA were significantly associated with elevated macrophages in the dormant state (M0) and decreased anti-inflammatory state (M2), the authors suggest that elevated IGFBP-2 secretion from GSCs could be an important factor for shaping the glioblastoma immune microenvironment. Finally, it was proposed that IGFBP-2 may serve to maintain the stemness of GSCs via autocrine signalling [221].
Experimental knockdown of IGFBP2 in GSCs by Hsieh et al. (2010) decreased the expression of SOX2, NES, BM1 and CD133, indicating a role of IGFBP-2 as part of the GSC transcriptional network [220]. The link between SOX2 abundance and IGFBP-2 was further elucidated in studies by Berezovsky et al. (2014) [222]. The group showed that patient-derived GSCs exposed to serum retained the potential to differentiate via SOX2. However, this was abolished when SOX2 was silenced [222]. Moreover, SOX2 knockdown mediated differential expressions of genes involved with embryogenic stem cell renewal, cytokine signalling and malignancy including a 6-fold downregulation of IGFBP2 [222]. In medulloblastoma—a childhood brain tumour—IGFBP-2 is instrumental for the SOX2-driven Shh subtype. In this study, IGFBP-2 was shown to mediate primary patient cell proliferation and migration through a signal transducer and activator of transcription 3 (STAT3)-mediated pathway [223]. In addition, using a genetic mouse model of medulloblastoma, Vanner et al. (2014) showed that Shh subgroup cells could maintain quiescent states during anti-mitotic chemotherapy through SOX2 positivity [224]. Therefore, there may be a functional relationship between IGFBP-2 and SOX2 that has not yet been explored in specific subtypes of CSCs.

5.2. IGF-II and Glioblastoma

In vitro studies with immortalised glioblastoma cell lines including U87, U251, U373 and T98G show low expression of the IGFRs, and in many cases lack cellular IGF-I or -II [225,226,227]. As follows, several studies in these cell lines found that IGF-I or -II treatment caused little mitogenic effect in vitro and in vivo [225,226]. ISH or IHC of astrocytoma and glioblastoma tissue has detected the IGF1R, IGF1 and IGF2, although overall IGF2 seems much less prevalent (5/88 positive glioblastomas, Soroceanu et al. (2007); 29/50 primary and secondary glioblastomas, Suvasini et al. (2011)) [228,229,230,231,232]. Early studies comparing normal brain and mixed glioma specimens largely indicated elevated tumour IGF1 and IGF2 expression and protein abundance; however, these reports suffered from ambiguity due to small sample sizes and differing detection methods [228,229,233,234]. Larger-scale comparison of low/high-grade astrocytomas, glioblastomas and normal human brain tissue showed no significant differences in IGF expression [232,235,236]. Moreover, 78% of 218 glioblastoma samples studied by Maris et al. (2015) showed no positivity for IGF-II when analysed by IHC [237]. Interestingly, however, of the glioblastomas with positivity, IGF-II has been mostly identified in peri necrotic areas, suggestive of a hypoxic relationship in these tumours [237,238].
It is worth noting that a small subset of glioblastomas with elevated IGF2 expression may be prognostically relevant. Further investigation of this subgroup by Soroceanu et al. (2007) demonstrated that 16/121 glioblastomas in their study displayed enhanced IGF2 expression and were associated with a lack of EGFR modifications, loss of PTEN, elevated levels of the IGF-1R, PI3K/P-Akt proliferation and poorer patient survival [231,235]. Indeed, in relation to the findings by Mu et al. (2014), it is possible that IGF-II upregulation is activated in a subset of glioblastomas by insulin-like growth factor 2 mRNA-binding protein 2 (IMP2) [239]. This study showed that IMP2 can regulate IGF-II activity, to promote downstream PI3K/Akt signalling [239].
As referred to previously, there have been no experimental approaches to study the effects of IGF-II on the patient-derived GSC population. This, however, would be interesting since IGF-II activity has been shown to substitute for EGF and can cooperate with FGF to maintain stem cell potency and survival [240]. Furthermore, IGF-II appears to support the growth of neurospheres derived from glioma cell lines in vitro via the activation of IGF-1R and downstream PI3K-Akt signalling [231].

6. Discussion

Glioblastoma is a poorly differentiated tumour, strongly aligned with an NSC origin [180,241]. Studies to investigate the cell of origin are essential for understanding the biological basis of and clinical implications for glioblastoma [242]. GSC populations create a moving target, associated with treatment evasion and the inhibition of long-term remission [243]. Standard treatment regimens including radiotherapy and TMZ chemotherapy require the presence of actively cycling cells. Many of the regulatory cues required for the quiescent/activated states of NSCs are largely understudied but could be essential for cellular ‘priming’ to cycling and a targetable state [244,245]. Therefore, the unique biochemistry of the brain, glial/neural cell biology and localised secretion could all be essential in the establishment of the heterogeneity for which glioblastoma is so well known.
Several studies have reported IGFBP-2 overexpression in glioma of increasing grade and aggression [246,247]. Many of these studies also present a strong prognostic role for IGFBP-2 [9,210]. In vitro studies including immortalised glioblastoma cell lines also reveal higher expression than normal brain samples under serum-cultured conditions, and several functions have been elucidated in other neoplasms [216]. Despite the inverse relationship between tumour IGFBP-2 abundance and survival, the mechanistic basis and functional consequences are poorly understood.
IGFBP-2 is highly expressed in the prenatal CNS [76,248]. It has been recorded at key regions of differentiation priming, morphogenesis and growth [249]. In particular, the notochord, which has the capacity to differentiate into the floor plate for further development of the nervous system of rats, coincides with high IGFBP-2 expression [250,251]. A high abundance of IGFBP-2 in proliferative NSCs in the mouse neural tube at E11.5 shows high correlation with and apical polarity with nestin and SOX2 [252]. The elevation of neuron-specific βIII-tubulin (TUJ1) and GFAP at E17.5 coincides with decreased IGFBP-2 [252]. Intricate expression patterns in mid-gestational rat embryos by Wood et al. (1992) showed that transitioning neuroblasts increased the neuronal intermediate filament protein alpha-internexin and lost IGFBP-2 [249,253]. IGFBP2 knockdown studies also highlighted an increased capacity for differentiation of mouse NSCs [252]. The abundance in self-renewing populations is indicative of a role for IGFBP-2 in their maintenance prior to lineage commitment and differentiation induction. Furthermore, established roles in brain development have been supplemented by emerging functions for higher-order brain functioning into adulthood [254,255,256].
Although IGF-II does not seem to be overexpressed in glioblastoma cells, the study of 88 glioblastomas by Soroceanu et al. (2007) suggest that there could be a subset of patients with high IGF2 expression [231]. Moreover, as mentioned previously, the measurement of high-molecular-weight variants of IGF-II requires further study. Notably, the secretion of high-molecular-weight IGF-II has been detected in the serum of cancer patients, with a direct link to non-islet tumour cell hypoglycaemia—reviewed by Van Doorn et al. (2020) [257]. Whether these variants are similarly enriched in glioblastoma remains unconfirmed. Similarly, given their preserved receptor activation capacity, but potentially altered affinity for IGF-binding proteins, including IGFBP-2, it is plausible that high-molecular-weight IGF-II may differentially modulate IGF bioavailability and receptor activation in the glioblastoma microenvironment [258,259]. Indeed, the detection of a 9 kDa variant in human CSF reported by Haselbacher et al. (1982) warrants further investigation in this context [68].
Drawing from studies in NSPCs, the interaction between IGF-II and IR-A appears to be important for cell clonogenicity and stemness [139]. However, despite evidence for the overexpression of IR-A in aggressive cancers, there is a lack of study on glioblastoma [260,261,262]. Again, despite a lack of research in patient-derived GSCs, there is evidence to suggest that IR-A may enhance cancer stemness in hepatocellular carcinoma (HCC) [263]. Of note, the use of IGF-II neutralising antibodies holds promise in slowing the growth of mouse HCC xenografts in mice [264].
In the adult human brain, IGFBP-2 is locally produced and too large to cross the BBB into systemic circulation and vice versa [76]. Postnatal retention of IGFBP2 expression by ChP epithelia and hippocampus localises IGFBP-2 to areas of adult NSCs [114,122]. Fluorescent labelling of mouse SVZ and SGZ regions postnatally indicated that IGFBP2 was restricted to cells with nestin positivity [252]. Similarly, IGF2 is expressed by ChP and can be detected at higher levels in the CSF of glioblastoma patients [120]. With a molecular weight of 6765 Daltons, IGF-II can cross the BBB and could also be delivered to the tumour via the vasculature [78]. Modern secretome analysis is hoped to expand our understanding of cellular crosstalk and signal transduction in disease [265]. With many of the available methods requiring pre-selected antibody arrays, the development of unbiased methods such as LC-MS/MS may provide more comprehensive characterisation of secreted factors [114].
The modulation of IGFs is a well-established role for IGFBP-2 with downstream mitogenic and systemic effects including metabolism [266]. Therefore, the extracellular role of IGFBP-2 and IGF-II in close proximity to SVZ or SGZ NSCs could be described by canonical IGF signalling. However, this does not explain the overexpression and prognostic effect of IGFBP-2 in glioblastoma studies. Therefore, independent functioning also seems likely, relying on the additional functional motifs present within the IGFBP-2 structure [77]. On the other hand, the role of IGF-II still requires elucidation, and its expression may be prognostically relevant in a subset of patients. Lastly, the proximity of SVZ NSCs to the CSF raises the possibility that IGFBP-2, IGF-II and other understudied factors here could be involved in CNS disease [122].

7. Conclusions

To conclude, the study of NSCs, their states and how these are regulated may be imperative to our understanding of glioblastoma initiation. The expressions of IGFBP2 and IGF2 are retained in the adult human brain, most notably in the ChP and derived CSF—highlighting them both as interesting constituents of the adult SVZ NSC niche [122,267]. Moreover, IGFBP-2 has been detected in the CM of NSCs in vitro [110,114]. Studying how these factors regulate NSCs and their progenitors may provide clues as to how they modulate GSCs. Indeed, evidence is suggestive of the role of IGFBP-2 in NSC and GSC maintenance [220,252]. Although not well defined, the expression of IGF2 in the adult ChP and CSF, combined with signalling though IR-A, supports a rationale for further investigation in the context of GSC maintenance. Further study should focus on IGFBP-2 and IGF-II as secreted factors and their functions, both dependent and independent of each other.

Author Contributions

Conceptualisation, A.J.H. writing—original draft preparation, A.J.H.; writing—review and editing, A.J.H. and C.M.P.; supervision, C.M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Southmead Hospital Charity, North Bristol Trust Registered Charity No: 1055900.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
APCAstrocyte progenitor cell
APOEApolipoprotein E
BBBBlood–brain barrier
BMPBone morphogenic protein
CDK4Cyclin-dependent kinase 4
CHI3L1Chitinase-3-like protein 1
ChPChoroid plexus
CMConditioned media
CNSCentral nervous system
CSCCancer stem cell
CSFCerebral spinal fluid
ECMExtracellular matrix
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
ERKExtracellular signal-related kinase
ESCEmbryonic stem cell
FABP7Fatty acid binding protein 7
FAKFocal adhesion kinase
FBSFoetal bovine serum
FGFFibroblast growth factor
GFAPGlial fibrillary acidic protein
GSCGlioma stem cell
IDHIsocitrate dehydrogenase
IGFInsulin-like growth factor
IGF-1RInsulin-like growth factor I receptor
IGF-2RInsulin-like growth factor II receptor
IGFBPInsulin-like growth factor binding protein
IHCImmunohistochemistry
ILKIntegrin linked kinase
IQGAP1IQ motif containing GTPase-activating protein 1
IRInsulin receptor
IRSInsulin receptor substrate
ISHIn situ hybridisation
JNKJUN NH2 terminal kinase
LC-MS/MSLiquid chromatography with tandem mass spectrometry
LGALS3Galectin 3
M6PMannose-6-phosphate
MAPKMitogen-activated protein kinase
MDM2Mouse double minute 2 homolog
MEKMitogen-activated protein kinase
MMPMatrix metalloproteinase
mRNAMessenger ribonucleic acid
NF1Neurofibromatosis type 1
NPCNeural precursor cell
NSCNeural stem cell
NSPCNeural stem/progenitor cell
OLIG2Oligodendrocyte transcription factor 2
OPCOligodendrocyte precursor cell
OSOverall survival
PDGFRPlatelet-derived growth factor receptor
PFSProgression free survival
PI3KPhosphatidylinositol-3-kinase
PTENPhosphatase and tensin homolog
qPCRQuantitative polymerase chain reaction
RBRetinoblastoma protein
RembrandtRepository for Molecular Brain Neoplasia Data
RGCRadial glial cell
RGDArginine–glycine–aspartate
RNARibonucleic acid
ROSReactive oxygen species
ShhSonic hedgehog
SOX2Sex determining region Y-box 2
STAT3Signal transducer and activator of transcription 3
SVZSubventricular zone
SGZSubgranular zone
TERTTelomerase reverse transcriptase
TGFTransforming growth factor
TIMP1Tissue inhibitor matrix metalloproteinase 1
TMZTemozolomide
TP53Tumour protein p53
VEGFVascular endothelial growth factor
WHOWorld Health Organisation

References

  1. Brodbelt, A.; Greenberg, D.; Winters, T.; Williams, M.; Vernon, S.; Collins, V.P. Glioblastoma in England: 2007–2011. Eur. J. Cancer 2015, 51, 533–542. [Google Scholar] [CrossRef] [PubMed]
  2. David, N.; Louis, A.P. Pieter Wesseling, Daniel J Brat, Ian A Cree, Dominique Figarella-Branger, Cynthia Hawkins, H K Ng, Stefan M Pfister, Guido Reifenberger, Riccardo Soffietti, Andreas von Deimling, David W Ellison. In WHO Classification of Central Nervous System Tumour, 5th ed.; WHO: Geneva, Switzerland, 2021. [Google Scholar]
  3. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2012–2016. Neuro-Oncol. 2019, 21 (Suppl. S5), v1–v100. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  4. Qi, D.; Li, J.; Quarles, C.C.; Fonkem, E.; Wu, E. Assessment and prediction of glioblastoma therapy response: Challenges and opportunities. Brain 2023, 146, 1281–1298. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Gerber, N.K.; Goenka, A.; Turcan, S.; Reyngold, M.; Makarov, V.; Kannan, K.; Beal, K.; Omuro, A.; Yamada, Y.; Gutin, P.; et al. Transcriptional diversity of long-term glioblastoma survivors. Neuro-Oncol. 2014, 16, 1186–1195. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  6. Kalya, M.; Kel, A.; Wlochowitz, D.; Wingender, E.; Beißbarth, T. IGFBP2 Is a Potential Master Regulator Driving the Dysregulated Gene Network Responsible for Short Survival in Glioblastoma Multiforme. Front. Genet. 2021, 12, 670240. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  7. Li, F.; Li, Y.; Zhang, K.; Li, Y.; He, P.; Liu, Y.; Yuan, H.; Lu, H.; Liu, J.; Che, S.; et al. FBLN4 as candidate gene associated with long-term and short-term survival with primary glioblastoma. Onco Targets Ther. 2017, 10, 387–395. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  8. Lindström, M.S. Expanding the scope of candidate prognostic marker IGFBP2 in glioblastoma. Biosci. Rep. 2019, 39, BSR20190770. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Yuan, Q.; Cai, H.-Q.; Zhong, Y.; Zhang, M.-J.; Cheng, Z.-J.; Hao, J.-J.; Wang, M.-R.; Wan, J.-H. Overexpression of IGFBP2 mRNA predicts poor survival in patients with glioblastoma. Biosci. Rep. 2019, 39, BSR20190045. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Holmes, K.M.; Annala, M.; Chua, C.Y.X.; Dunlap, S.M.; Liu, Y.; Hugen, N.; Moore, L.M.; Cogdell, D.; Hu, L.; Nykter, M.; et al. Insulin-like growth factor-binding protein 2-driven glioma progression is prevented by blocking a clinically significant integrin, integrin-linked kinase, and NF-κB network. Proc. Natl. Acad. Sci. USA 2012, 109, 3475–3480. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  11. Liu, Y.; Li, F.; Yang, Y.T.; Xu, X.D.; Chen, J.S.; Chen, T.L.; Chen, H.J.; Zhu, Y.B.; Lin, J.Y.; Li, Y.; et al. IGFBP2 promotes vasculogenic mimicry formation via regulating CD144 and MMP2 expression in glioma. Oncogene 2019, 38, 1815–1831. [Google Scholar] [CrossRef] [PubMed]
  12. Mendes, K.N.; Wang, G.K.; Fuller, G.N.; Zhang, W. JNK mediates insulin-like growth factor binding protein 2/integrin alpha5-dependent glioma cell migration. Int. J. Oncol. 2010, 37, 143–153. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, G.K.; Hu, L.; Fuller, G.N.; Zhang, W. An interaction between insulin-like growth factor-binding protein 2 (IGFBP2) and integrin alpha5 is essential for IGFBP2-induced cell mobility. J. Biol. Chem. 2006, 281, 14085–14091. [Google Scholar] [CrossRef] [PubMed]
  14. Han, S.; Li, Z.; Master, L.M.; Master, Z.W.; Wu, A. Exogenous IGFBP-2 promotes proliferation, invasion, and chemoresistance to temozolomide in glioma cells via the integrin β1-ERK pathway. Br. J. Cancer 2014, 111, 1400–1409. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. Loras, A.; Gonzalez-Bonet, L.G.; Gutierrez-Arroyo, J.L.; Martinez-Cadenas, C.; Marques-Torrejon, M.A. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life 2023, 13, 905. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Pollard, S.M.; Yoshikawa, K.; Clarke, I.D.; Danovi, D.; Stricker, S.; Russell, R.; Bayani, J.; Head, R.; Lee, M.; Bernstein, M.; et al. Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell 2009, 4, 568–580. [Google Scholar] [CrossRef] [PubMed]
  17. Bracko, O.; Singer, T.; Aigner, S.; Knobloch, M.; Winner, B.; Ray, J.; Clemenson, G.D.; Suh, H.; Couillard-Despres, S.; Aigner, L.; et al. Gene expression profiling of neural stem cells and their neuronal progeny reveals IGF2 as a regulator of adult hippocampal neurogenesis. J. Neurosci. 2012, 32, 3376–3387. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Ferrón, S.R.; Radford, E.J.; Domingo-Muelas, A.; Kleine, I.; Ramme, A.; Gray, D.; Sandovici, I.; Constancia, M.; Ward, A.; Menheniott, T.R.; et al. Differential genomic imprinting regulates paracrine and autocrine roles of IGF2 in mouse adult neurogenesis. Nat. Commun. 2015, 6, 8265. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  19. Massagué, J.; Czech, M.P. The subunit structures of two distinct receptors for insulin-like growth factors I and II and their relationship to the insulin receptor. J. Biol. Chem. 1982, 257, 5038–5045. [Google Scholar] [CrossRef] [PubMed]
  20. Brown, J.; Jones, E.Y.; Forbes, B.E. Interactions of IGF-II with the IGF2R/cation-independent mannose-6-phosphate receptor mechanism and biological outcomes. Vitam. Horm. 2009, 80, 699–719. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, Z.Q.; Fung, M.R.; Barlow, D.P.; Wagner, E.F. Regulation of embryonic growth and lysosomal targeting by the imprinted Igf2/Mpr gene. Nature 1994, 372, 464–467. [Google Scholar] [CrossRef] [PubMed]
  22. Allard, J.B.; Duan, C. IGF-Binding Proteins: Why Do They Exist and Why Are There So Many? Front. Endocrinol. 2018, 9, 117. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Elgin, R.G.; Busby, W.H.; Clemmons, D.R. An insulin-like growth factor (IGF) binding protein enhances the biologic response to IGF-I. Proc. Natl. Acad. Sci. USA 1987, 84, 3254–3258. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Guler, H.P.; Zapf, J.; Schmid, C.; Froesch, E.R. Insulin-like growth factors I and II in healthy man. Estimations of half-lives and production rates. Acta Endocrinol. 1989, 121, 753–758. [Google Scholar] [CrossRef] [PubMed]
  25. Massoner, P.; Ladurner-Rennau, M.; Eder, I.E.; Klocker, H. Insulin-like growth factors and insulin control a multifunctional signalling network of significant importance in cancer. Br. J. Cancer 2010, 103, 1479–1484. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Rosenfeld, R.G.; Pham, H.; Conover, C.A.; Hintz, R.L.; Baxter, R.C. Structural and immunological comparison of insulin-like growth factor binding proteins of cerebrospinal and amniotic fluids. J. Clin. Endocrinol. Metab. 1989, 68, 638–646. [Google Scholar] [CrossRef] [PubMed]
  27. LeRoith, D.; Holly, J.M.P.; Forbes, B.E. Insulin-like growth factors: Ligands, binding proteins, and receptors. Mol. Metab. 2021, 52, 101245. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Allan, G.J.; Flint, D.J.; Patel, K. Insulin-like growth factor axis during embryonic development. Reproduction 2001, 122, 31–39. [Google Scholar] [CrossRef] [PubMed]
  29. Hawkes, C.; Kar, S. The insulin-like growth factor-II/mannose-6-phosphate receptor: Structure, distribution and function in the central nervous system. Brain Res. Brain Res. Rev. 2004, 44, 117–140. [Google Scholar] [CrossRef] [PubMed]
  30. Lee, J.S.; Tocheny, C.E.; Shaw, L.M. The Insulin-like Growth Factor Signaling Pathway in Breast Cancer: An Elusive Therapeutic Target. Life 2022, 12, 1992. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  31. Hintz, R.L.; Liu, F. Demonstration of specific plasma protein binding sites for somatomedin. J. Clin. Endocrinol. Metab. 1977, 45, 988–995. [Google Scholar] [CrossRef] [PubMed]
  32. Andress, D.L.; Birnbaum, R.S. A novel human insulin-like growth factor binding protein secreted by osteoblast-like cells. Biochem. Biophys. Res. Commun. 1991, 176, 213–218. [Google Scholar] [CrossRef] [PubMed]
  33. Baxter, R.C.; Martin, J.L.; Tyler, M.I.; Howden, M.E. Growth hormone-dependent insulin-like growth factor (IGF) binding protein from human plasma differs from other human IGF binding proteins. Biochem. Biophys. Res. Commun. 1986, 139, 1256–1261. [Google Scholar] [CrossRef] [PubMed]
  34. Mohan, S.; Bautista, C.M.; Wergedal, J.; Baylink, D.J. Isolation of an inhibitory insulin-like growth factor (IGF) binding protein from bone cell-conditioned medium: A potential local regulator of IGF action. Proc. Natl. Acad. Sci. USA 1989, 86, 8338–8342. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Moses, A.C.; Nissley, S.P.; Passamani, J.; White, R.M. Further characterization of growth hormone-dependent somatomedin-binding proteins in rat serum and demonstration of somatomedin-binding proteins produced by rat liver cells in culture. Endocrinology 1979, 104, 536–546. [Google Scholar] [CrossRef] [PubMed]
  36. Mottola, C.; MacDonald, R.G.; Brackett, J.L.; Mole, J.E.; Anderson, J.K.; Czech, M.P. Purification and amino-terminal sequence of an insulin-like growth factor-binding protein secreted by rat liver BRL-3A cells. J. Biol. Chem. 1986, 261, 11180–11188. [Google Scholar] [CrossRef] [PubMed]
  37. Shimasaki, S.; Gao, L.; Shimonaka, M.; Ling, N. Isolation and molecular cloning of insulin-like growth factor-binding protein-6. Mol. Endocrinol. 1991, 5, 938–948. [Google Scholar] [CrossRef] [PubMed]
  38. Shimasaki, S.; Shimonaka, M.; Zhang, H.P.; Ling, N. Identification of five different insulin-like growth factor binding proteins (IGFBPs) from adult rat serum and molecular cloning of a novel IGFBP-5 in rat and human. J. Biol. Chem. 1991, 266, 10646–10653. [Google Scholar] [CrossRef] [PubMed]
  39. Shimonaka, M.; Schroeder, R.; Shimasaki, S.; Ling, N. Identification of a novel binding protein for insulin-like growth factors in adult rat serum. Biochem. Biophys. Res. Commun. 1989, 165, 189–195. [Google Scholar] [CrossRef] [PubMed]
  40. Baxter, R.C. Characterization of the acid-labile subunit of the growth hormone-dependent insulin-like growth factor binding protein complex. J. Clin. Endocrinol. Metab. 1988, 67, 265–272. [Google Scholar] [CrossRef] [PubMed]
  41. Firth, S.M.; Clemmons, D.R.; Baxter, R.C. Mutagenesis of basic amino acids in the carboxyl-terminal region of insulin-like growth factor binding protein-5 affects acid-labile subunit binding. Endocrinology 2001, 142, 2147. [Google Scholar] [CrossRef] [PubMed]
  42. Twigg, S.M.; Kiefer, M.C.; Zapf, J.; Baxter, R.C. Insulin-like growth factor-binding protein 5 complexes with the acid-labile subunit. Role of the carboxyl-terminal domain. J. Biol. Chem. 1998, 273, 28791–28798. [Google Scholar] [CrossRef] [PubMed]
  43. Cerro, J.A.; Grewal, A.; Wood, T.L.; Pintar, J.E. Tissue-specific expression of the insulin-like growth factor binding protein (IGFBP) mRNAs in mouse and rat development. Regul. Pept. 1993, 48, 189–198. [Google Scholar] [CrossRef] [PubMed]
  44. Conover, C.A.; Oxvig, C.; Overgaard, M.T.; Christiansen, M.; Giudice, L.C. Evidence that the insulin-like growth factor binding protein-4 protease in human ovarian follicular fluid is pregnancy associated plasma protein-A. J. Clin. Endocrinol. Metab. 1999, 84, 4742–4745. [Google Scholar] [CrossRef] [PubMed]
  45. Laursen, L.S.; Overgaard, M.T.; Søe, R.; Boldt, H.B.; Sottrup-Jensen, L.; Giudice, L.C.; Conover, C.A.; Oxvig, C. Pregnancy-associated plasma protein-A (PAPP-A) cleaves insulin-like growth factor binding protein (IGFBP)-5 independent of IGF: Implications for the mechanism of IGFBP-4 proteolysis by PAPP-A. FEBS Lett. 2001, 504, 36–40. [Google Scholar] [CrossRef] [PubMed]
  46. Monget, P.; Mazerbourg, S.; Delpuech, T.; Maurel, M.-C.; Manière, S.; Zapf, J.; Lalmanach, G.; Oxvig, C.; Overgaard, M.T. Pregnancy-associated plasma protein-A is involved in insulin-like growth factor binding protein-2 (IGFBP-2) proteolytic degradation in bovine and porcine preovulatory follicles: Identification of cleavage site and characterization of IGFBP-2 degradation. Biol. Reprod. 2003, 68, 77–86. [Google Scholar] [CrossRef] [PubMed]
  47. Rorive, S.; Berton, A.; D’Haene, N.; Takacs, C.N.; Debeir, O.; Decaestecker, C.; Salmon, I. Matrix metalloproteinase-9 interplays with the IGFBP2-IGFII complex to promote cell growth and motility in astrocytomas. Glia 2008, 56, 1679–1690. [Google Scholar] [CrossRef] [PubMed]
  48. Jones, J.I.; Gockerman, A.; Busby, W.H.; Camacho-Hubner, C.; Clemmons, D.R. Extracellular matrix contains insulin-like growth factor binding protein-5: Potentiation of the effects of IGF-I. J. Cell Biol. 1993, 121, 679–687. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Bondy, C.A.; Werner, H.; Roberts, C.T.; LeRoith, D. Cellular pattern of insulin-like growth factor-I (IGF-I) and type I IGF receptor gene expression in early organogenesis: Comparison with IGF-II gene expression. Mol. Endocrinol. 1990, 4, 1386–1398. [Google Scholar] [CrossRef] [PubMed]
  50. Rotwein, P.; Burgess, S.K.; Milbrandt, J.D.; Krause, J.E. Differential expression of insulin-like growth factor genes in rat central nervous system. Proc. Natl. Acad. Sci. USA 1988, 85, 265–269. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Pera, E.M.; Wessely, O.; Li, S.Y.; De Robertis, E.M. Neural and head induction by insulin-like growth factor signals. Dev. Cell 2001, 1, 655–665. [Google Scholar] [CrossRef] [PubMed]
  52. Richard-Parpaillon, L.; Héligon, C.; Chesnel, F.; Boujard, D.; Philpott, A. The IGF pathway regulates head formation by inhibiting Wnt signaling in Xenopus. Dev. Biol. 2002, 244, 407–417. [Google Scholar] [CrossRef] [PubMed]
  53. Ayer-le Lievre, C.; Ståhlbom, P.A.; Sara, V.R. Expression of IGF-I and -II mRNA in the brain and craniofacial region of the rat fetus. Development 1991, 111, 105–115. [Google Scholar] [CrossRef] [PubMed]
  54. Bach, M.A.; Shen-Orr, Z.; Lowe, W.L.; Roberts, C.T.; LeRoith, D. Insulin-like growth factor I mRNA levels are developmentally regulated in specific regions of the rat brain. Brain Res. Mol. Brain Res. 1991, 10, 43–48. [Google Scholar] [CrossRef] [PubMed]
  55. Bondy, C.; Lee, W.H. Correlation between insulin-like growth factor (IGF)-binding protein 5 and IGF-I gene expression during brain development. J. Neurosci. 1993, 13, 5092–5104. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Freude, S.; Leeser, U.; Müller, M.; Hettich, M.M.; Udelhoven, M.; Schilbach, K.; Tobe, K.; Kadowaki, T.; Köhler, C.; Schröder, H.; et al. IRS-2 branch of IGF-1 receptor signaling is essential for appropriate timing of myelination. J. Neurochem. 2008, 107, 907–917. [Google Scholar] [CrossRef] [PubMed]
  57. Russo, V.C.; Werther, G.A. Des (1-3) IGF-I potently enhances differentiated cell growth in olfactory bulb organ culture. Growth Factors 1994, 11, 301–311. [Google Scholar] [CrossRef] [PubMed]
  58. Shinar, Y.; McMorris, F.A. Developing oligodendroglia express mRNA for insulin-like growth factor-I, a regulator of oligodendrocyte development. J. Neurosci. Res. 1995, 42, 516–527. [Google Scholar] [CrossRef] [PubMed]
  59. Bartlett, W.P.; Li, X.S.; Williams, M. Expression of IGF-1 mRNA in the murine subventricular zone during postnatal development. Brain Res. Mol. Brain Res. 1992, 12, 285–291. [Google Scholar] [CrossRef] [PubMed]
  60. Bartlett, W.P.; Li, X.S.; Williams, M.; Benkovic, S. Localization of insulin-like growth factor-1 mRNA in murine central nervous system during postnatal development. Dev. Biol. 1991, 147, 239–250. [Google Scholar] [CrossRef] [PubMed]
  61. Ye, P.; Umayahara, Y.; Ritter, D.; Bunting, T.; Auman, H.; Rotwein, P.; D’ercole, A.J. Regulation of insulin-like growth factor I (IGF-I) gene expression in brain of transgenic mice expressing an IGF-I-luciferase fusion gene. Endocrinology 1997, 138, 5466–5475. [Google Scholar] [CrossRef] [PubMed]
  62. Logan, A.; Gonzalez, A.M.; Hill, D.J.; Berry, M.; Gregson, N.A.; Baird, A. Coordinated pattern of expression and localization of insulin-like growth factor-II (IGF-II) and IGF-binding protein-2 in the adult rat brain. Endocrinology 1994, 135, 2255–2264. [Google Scholar] [CrossRef] [PubMed]
  63. Charalambous, M.; Menheniott, T.R.; Bennett, W.R.; Kelly, S.M.; Dell, G.; Dandolo, L.; Ward, A. An enhancer element at the Igf2/H19 locus drives gene expression in both imprinted and non-imprinted tissues. Dev. Biol. 2004, 271, 488–497. [Google Scholar] [CrossRef] [PubMed]
  64. Hynes, M.A.; Brooks, P.J.; Van Wyk, J.J.; Lund, P.K. Insulin-like growth factor II messenger ribonucleic acids are synthesized in the choroid plexus of the rat brain. Mol. Endocrinol. 1988, 2, 47–54. [Google Scholar] [CrossRef] [PubMed]
  65. Stylianopoulou, F.; Efstratiadis, A.; Herbert, J.; Pintar, J. Pattern of the insulin-like growth factor II gene expression during rat embryogenesis. Development 1988, 103, 497–506. [Google Scholar] [CrossRef] [PubMed]
  66. Daughaday, W.H.; Trivedi, B. Measurement of derivatives of proinsulin-like growth factor-II in serum by a radioimmunoassay directed against the E-domain in normal subjects and patients with nonislet cell tumor hypoglycemia. J. Clin. Endocrinol. Metab. 1992, 75, 110–115. [Google Scholar] [CrossRef] [PubMed]
  67. Scalia, P.; Marino, I.R.; Asero, S.; Pandini, G.; Grimberg, A.; El-Deiry, W.S.; Williams, S.J. Autocrine IGF-II-Associated Cancers: From a Rare Paraneoplastic Event to a Hallmark in Malignancy. Biomedicines 2023, 12, 40. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Haselbacher, G.; Humbel, R. Evidence for two species of insulin-like growth factor II (IGF II and “big” IGF II) in human spinal fluid. Endocrinology 1982, 110, 1822–1824. [Google Scholar] [CrossRef] [PubMed]
  69. Haselbacher, G.K.; Schwab, M.E.; Pasi, A.; Humbel, R.E. Insulin-like growth factor II (IGF II) in human brain: Regional distribution of IGF II and of higher molecular mass forms. Proc. Natl. Acad. Sci. USA 1985, 82, 2153–2157. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Lee, W.H.; Wang, G.M.; Seaman, L.B.; Vannucci, S.J. Coordinate IGF-I and IGFBP5 gene expression in perinatal rat brain after hypoxia-ischemia. J. Cereb. Blood Flow Metab. 1996, 16, 227–236. [Google Scholar] [CrossRef] [PubMed]
  71. Walter, H.J.; Berry, M.; Hill, D.J.; Logan, A. Spatial and temporal changes in the insulin-like growth factor (IGF) axis indicate autocrine/paracrine actions of IGF-I within wounds of the rat brain. Endocrinology 1997, 138, 3024–3034. [Google Scholar] [CrossRef] [PubMed]
  72. Brar, A.K.; Chernausek, S.D. Localization of insulin-like growth factor binding protein-4 expression in the developing and adult rat brain: Analysis by in situ hybridization. J. Neurosci. Res. 1993, 35, 103–114. [Google Scholar] [CrossRef] [PubMed]
  73. Delhanty, P.J.; Hill, D.J.; Shimasaki, S.; Han, V.K. Insulin-like growth factor binding protein-4, -5 and -6 mRNAs in the human fetus: Localization to sites of growth and differentiation? Growth Regul. 1993, 3, 8–11. [Google Scholar] [PubMed]
  74. Stenvers, K.L.; Zimmermann, E.M.; Gallagher, M.; Lund, P.K. Expression of insulin-like growth factor binding protein-4 and -5 mRNAs in adult rat forebrain. J. Comp. Neurol. 1994, 339, 91–105. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, W.H.; Michels, K.M.; Bondy, C.A. Localization of insulin-like growth factor binding protein-2 messenger RNA during postnatal brain development: Correlation with insulin-like growth factors I and II. Neuroscience 1993, 53, 251–265. [Google Scholar] [CrossRef] [PubMed]
  76. Ocrant, I.; Fay, C.T.; Parmelee, J.T. Characterization of insulin-like growth factor binding proteins produced in the rat central nervous system. Endocrinology 1990, 127, 1260–1267. [Google Scholar] [CrossRef] [PubMed]
  77. Wood, T.L.; Brown, A.L.; Rechler, M.M.; Pintar, J.E. The expression pattern of an insulin-like growth factor (IGF)-binding protein gene is distinct from IGF-II in the midgestational rat embryo. Mol. Endocrinol. 1990, 4, 1257–1263. [Google Scholar] [CrossRef] [PubMed]
  78. Reinhardt, R.R.; Bondy, C.A. Insulin-like growth factors cross the blood-brain barrier. Endocrinology 1994, 135, 1753–1761. [Google Scholar] [CrossRef] [PubMed]
  79. Altman, J. Autoradiographic investigation of cell proliferation in the brains of rats and cats. Anat. Rec. 1963, 145, 573–591. [Google Scholar] [CrossRef] [PubMed]
  80. Altman, J.; Das, G.D. Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J. Comp. Neurol. 1965, 124, 319–335. [Google Scholar] [CrossRef] [PubMed]
  81. de Carlos, J.A.; López-Mascaraque, L.; Valverde, F. Dynamics of cell migration from the lateral ganglionic eminence in the rat. J. Neurosci. 1996, 16, 6146–6156. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  82. Fuentealba, L.C.; Rompani, S.B.; Parraguez, J.I.; Obernier, K.; Romero, R.; Cepko, C.L.; Alvarez-Buylla, A. Embryonic Origin of Postnatal Neural Stem Cells. Cell 2015, 161, 1644–1655. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Merkle, F.T.; Tramontin, A.D.; García-Verdugo, J.M.; Alvarez-Buylla, A. Radial glia give rise to adult neural stem cells in the subventricular zone. Proc. Natl. Acad. Sci. USA 2004, 101, 17528–17532. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Yuzwa, S.A.; Borrett, M.J.; Innes, B.T.; Voronova, A.; Ketela, T.; Kaplan, D.R.; Bader, G.D.; Miller, F.D. Developmental Emergence of Adult Neural Stem Cells as Revealed by Single-Cell Transcriptional Profiling. Cell Rep. 2017, 21, 3970–3986. [Google Scholar] [CrossRef] [PubMed]
  85. Eriksson, P.S.; Perfilieva, E.; Björk-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] [PubMed]
  86. Doetsch, F.; García-Verdugo, J.M.; Alvarez-Buylla, A. Cellular composition and three-dimensional organization of the subventricular germinal zone in the adult mammalian brain. J. Neurosci. 1997, 17, 5046–5061. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  87. Hu, X.-L.; Chen, G.; Zhang, S.; Zheng, J.; Wu, J.; Bai, Q.-R.; Wang, Y.; Wang, H.; Feng, H.; Li, J.; et al. Persistent Expression of VCAM1 in Radial Glial Cells Is Required for the Embryonic Origin of Postnatal Neural Stem Cells. Neuron 2017, 95, 309–325.e6. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, H.-K.; Belz, T.; Bock, D.; Takacs, A.; Wu, H.; Lichter, P.; Chai, M.; Schütz, G. The nuclear receptor tailless is required for neurogenesis in the adult subventricular zone. Genes Dev. 2008, 22, 2473–2478. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Menn, B.; Garcia-Verdugo, J.M.; Yaschine, C.; Gonzalez-Perez, O.; Rowitch, D.; Alvarez-Buylla, A. Origin of oligodendrocytes in the subventricular zone of the adult brain. J. Neurosci. 2006, 26, 7907–7918. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Fukuda, S.; Kato, F.; Tozuka, Y.; Yamaguchi, M.; Miyamoto, Y.; Hisatsune, T. Two distinct subpopulations of nestin-positive cells in adult mouse dentate gyrus. J. Neurosci. 2003, 23, 9357–9366. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  91. Garcia, A.D.; Doan, N.B.; Imura, T.; Bush, T.G.; Sofroniew, M.V. GFAP-expressing progenitors are the principal source of constitutive neurogenesis in adult mouse forebrain. Nat. Neurosci. 2004, 7, 1233–1241. [Google Scholar] [CrossRef] [PubMed]
  92. Steiner, B.; Kronenberg, G.; Jessberger, S.; Brandt, M.D.; Reuter, K.; Kempermann, G. Differential regulation of gliogenesis in the context of adult hippocampal neurogenesis in mice. Glia 2004, 46, 41–52. [Google Scholar] [CrossRef] [PubMed]
  93. Suh, H.; Consiglio, A.; Ray, J.; Sawai, T.; D’Amour, K.A.; Gage, F.H. In vivo fate analysis reveals the multipotent and self-renewal capacities of Sox2+ neural stem cells in the adult hippocampus. Cell Stem Cell 2007, 1, 515–528. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  94. Merkle, F.T.; Mirzadeh, Z.; Alvarez-Buylla, A. Mosaic organization of neural stem cells in the adult brain. Science 2007, 317, 381–384. [Google Scholar] [CrossRef] [PubMed]
  95. Deng, W.; Saxe, M.D.; Gallina, I.S.; Gage, F.H. Adult-born hippocampal dentate granule cells undergoing maturation modulate learning and memory in the brain. J. Neurosci. 2009, 29, 13532–13542. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  96. Zhao, C.; Teng, E.M.; Summers, R.G.; Ming, G.L.; Gage, F.H. Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus. J. Neurosci. 2006, 26, 3–11. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  97. Smith, A.W.; Parashar, B.; Wernicke, A.G. Subventricular zone-associated glioblastoma: A call for translational research to guide clinical decision making. Neurogenesis 2016, 3, e1225548. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  98. Sanai, N.; Tramontin, A.D.; Quiñones-Hinojosa, A.; Barbaro, N.M.; Gupta, N.; Kunwar, S.; Lawton, M.T.; McDermott, M.W.; Parsa, A.T.; García-Verdugo, J.M.; 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] [PubMed]
  99. 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] [PubMed] [PubMed Central]
  100. Kriegstein, A.; Alvarez-Buylla, A. The glial nature of embryonic and adult neural stem cells. Annu. Rev. Neurosci. 2009, 32, 149–184. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  101. Zhao, C.; Deng, W.; Gage, F.H. Mechanisms and functional implications of adult neurogenesis. Cell 2008, 132, 645–660. [Google Scholar] [CrossRef] [PubMed]
  102. Passarelli, J.P.; Nimjee, S.M.; Townsend, K.L. Stroke and Neurogenesis: Bridging Clinical Observations to New Mechanistic Insights from Animal Models. Transl. Stroke Res. 2024, 15, 53–68. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, C.; Hu, J.; Ralls, S.; Kitamura, T.; Loh, Y.P.; Yang, Y.; Mukouyama, Y.-S.; Ahn, S. The molecular profiles of neural stem cell niche in the adult subventricular zone. PLoS ONE 2012, 7, e50501. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  104. Matarredona, E.R.; Talaverón, R.; Pastor, A.M. Interactions Between Neural Progenitor Cells and Microglia in the Subventricular Zone: Physiological Implications in the Neurogenic Niche and After Implantation in the Injured Brain. Front. Cell Neurosci. 2018, 12, 268. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  105. Dause, T.J.; Denninger, J.K.; Smith, B.M.; Kirby, E.D. The neural stem cell secretome across neurodevelopment. Exp. Neurol. 2022, 355, 114142. [Google Scholar] [CrossRef] [PubMed]
  106. Morizur, L.; Chicheportiche, A.; Gauthier, L.R.; Daynac, M.; Boussin, F.D.; Mouthon, M.A. Distinct Molecular Signatures of Quiescent and Activated Adult Neural Stem Cells Reveal Specific Interactions with Their Microenvironment. Stem Cell Rep. 2018, 11, 565–577. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Vogel, A.D.; Upadhya, R.; Shetty, A.K. Neural stem cell derived extracellular vesicles: Attributes and prospects for treating neurodegenerative disorders. eBioMedicine 2018, 38, 273–282. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  108. Komada, M.; Saitsu, H.; Kinboshi, M.; Miura, T.; Shiota, K.; Ishibashi, M. Hedgehog signaling is involved in development of the neocortex. Development 2008, 135, 2717–2727. [Google Scholar] [CrossRef] [PubMed]
  109. Daynac, M.; Tirou, L.; Faure, H.; Mouthon, M.-A.; Gauthier, L.R.; Hahn, H.; Boussin, F.D.; Ruat, M. Hedgehog Controls Quiescence and Activation of Neural Stem Cells in the Adult Ventricular-Subventricular Zone. Stem Cell Rep. 2016, 7, 735–748. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  110. Červenka, J.; Tylečková, J.; Skalníková, H.K.; Kepková, K.V.; Poliakh, I.; Valeková, I.; Pfeiferová, L.; Kolář, M.; Vaškovičová, M.; Pánková, T.; et al. Proteomic Characterization of Human Neural Stem Cells and Their Secretome During. Front. Cell Neurosci. 2020, 14, 612560. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Muffley, L.A.; Pan, S.C.; Smith, A.N.; Ga, M.; Hocking, A.M.; Gibran, N.S. Differentiation state determines neural effects on microvascular endothelial cells. Exp. Cell Res. 2012, 318, 2085–2093. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  112. Tham, M.; Ramasamy, S.; Gan, H.T.; Ramachandran, A.; Poonepalli, A.; Yu, Y.H.; Ahmed, S. CSPG is a secreted factor that stimulates neural stem cell survival possibly by enhanced EGFR signaling. PLoS ONE 2010, 5, e15341. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Dulken, B.W.; Leeman, D.S.; Boutet, S.C.; Hebestreit, K.; Brunet, A. Single-Cell Transcriptomic Analysis Defines Heterogeneity and Transcriptional Dynamics in the Adult Neural Stem Cell Lineage. Cell Rep. 2017, 18, 777–790. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. Denninger, J.K.; Chen, X.; Turkoglu, A.M.; Sarchet, P.; Volk, A.R.; Rieskamp, J.D.; Yan, P.; Kirby, E.D. Defining the adult hippocampal neural stem cell secretome: In vivo versus in vitro transcriptomic differences and their correlation to secreted protein levels. Brain Res. 2020, 1735, 146717. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Dause, T.J.; Denninger, J.K.; Osap, R.; Walters, A.E.; Rieskamp, J.D.; Kirby, E.D. Autocrine VEGF drives neural stem cell proximity to the adult hippocampus vascular niche. Life Sci. Alliance 2024, 7, e202402659. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Kirby, E.D.; Kuwahara, A.A.; Messer, R.L.; Wyss-Coray, T. Adult hippocampal neural stem and progenitor cells regulate the neurogenic niche by secreting VEGF. Proc. Natl. Acad. Sci. USA 2015, 112, 4128–4133. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  117. Lun, M.P.; Monuki, E.S.; Lehtinen, M.K. Development and functions of the choroid plexus-cerebrospinal fluid system. Nat. Rev. Neurosci. 2015, 16, 445–457. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  118. Walter, H.J.; Berry, M.; Hill, D.J.; Cwyfan-Hughes, S.; Holly, J.M.; Logan, A. Distinct sites of insulin-like growth factor (IGF)-II expression and localization in lesioned rat brain: Possible roles of IGF binding proteins (IGFBPs) in the mediation of IGF-II activity. Endocrinology 1999, 140, 520–532. [Google Scholar] [CrossRef] [PubMed]
  119. Spector, R.; Robert Snodgrass, S.; Johanson, C.E. A balanced view of the cerebrospinal fluid composition and functions: Focus on adult humans. Exp. Neurol. 2015, 273, 57–68. [Google Scholar] [CrossRef] [PubMed]
  120. Lehtinen, M.K.; Zappaterra, M.W.; Chen, X.; Yang, Y.J.; Hill, A.D.; Lun, M.; Maynard, T.; Gonzalez, D.; Kim, S.; Ye, P.; et al. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron 2011, 69, 893–905. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  121. 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] [PubMed] [PubMed Central]
  122. Silva-Vargas, V.; Maldonado-Soto, A.R.; Mizrak, D.; Codega, P.; Doetsch, F. Age-Dependent Niche Signals from the Choroid Plexus Regulate Adult Neural Stem Cells. Cell Stem Cell 2016, 19, 643–652. [Google Scholar] [CrossRef] [PubMed]
  123. Mashayekhi, F.; Draper, C.E.; Bannister, C.M.; Pourghasem, M.; Owen-Lynch, P.J.; Miyan, J.A. Deficient cortical development in the hydrocephalic Texas (H-Tx) rat: A role for CSF. Brain 2002, 125 Pt 8, 1859–1874. [Google Scholar] [CrossRef] [PubMed]
  124. Owen-Lynch, P.J.; Draper, C.E.; Mashayekhi, F.; Bannister, C.M.; Miyan, J.A. Defective cell cycle control underlies abnormal cortical development in the hydrocephalic Texas rat. Brain 2003, 126 Pt 3, 623–631. [Google Scholar] [CrossRef] [PubMed]
  125. Sawamoto, K.; Wichterle, H.; Gonzalez-Perez, O.; Cholfin, J.A.; Yamada, M.; Spassky, N.; Murcia, N.S.; Garcia-Verdugo, J.M.; Marin, O.; Rubenstein, J.L.R.; et al. New neurons follow the flow of cerebrospinal fluid in the adult brain. Science 2006, 311, 629–632. [Google Scholar] [CrossRef] [PubMed]
  126. Ziegler, A.N.; Levison, S.W.; Wood, T.L. Insulin and IGF receptor signalling in neural-stem-cell homeostasis. Nat. Rev. Endocrinol. 2015, 11, 161–170. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  127. Lehtinen, M.K.; Walsh, C.A. Neurogenesis at the brain-cerebrospinal fluid interface. Annu. Rev. Cell Dev. Biol. 2011, 27, 653–679. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  128. Dziegielewska, K.; Evans, C.; Lai, P.; Lorscheider, F.; Malinowska, D.; Møllgrd, K.; Saunders, N. Proteins in cerebrospinal fluid and plasma of fetal rats during development. Dev. Biol. 1981, 83, 193–200. [Google Scholar] [CrossRef] [PubMed]
  129. Marques, F.; Sousa, J.C.; Coppola, G.; Gao, F.; Puga, R.; Brentani, H.; Geschwind, D.H.; Sousa, N.; Correia-Neves, M.; Palha, J.A. Transcriptome signature of the adult mouse choroid plexus. Fluids Barriers CNS 2011, 8, 10. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  130. Parada, C.; Gato, A.; Bueno, D. Mammalian embryonic cerebrospinal fluid proteome has greater apolipoprotein and enzyme pattern complexity than the avian proteome. J. Proteome Res. 2005, 4, 2420–2428. [Google Scholar] [CrossRef] [PubMed]
  131. Zappaterra, M.D.; Lisgo, S.N.; Lindsay, S.; Gygi, S.P.; Walsh, C.A.; Ballif, B.A. A comparative proteomic analysis of human and rat embryonic cerebrospinal fluid. J. Proteome Res. 2007, 6, 3537–3548. [Google Scholar] [CrossRef] [PubMed]
  132. Grove, E.A.; Tole, S.; Limon, J.; Yip, L.; Ragsdale, C.W. The hem of the embryonic cerebral cortex is defined by the expression of multiple Wnt genes and is compromised in Gli3-deficient mice. Development 1998, 125, 2315–2325. [Google Scholar] [CrossRef] [PubMed]
  133. Haskell, G.T.; LaMantia, A.S. Retinoic acid signaling identifies a distinct precursor population in the developing and adult forebrain. J. Neurosci. 2005, 25, 7636–7647. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  134. Hébert, J.M.; Mishina, Y.; McConnell, S.K. BMP signaling is required locally to pattern the dorsal telencephalic midline. Neuron 2002, 35, 1029–1041. [Google Scholar] [CrossRef] [PubMed]
  135. Shimogori, T.; Banuchi, V.; Ng, H.Y.; Strauss, J.B.; Grove, E.A. Embryonic signaling centers expressing BMP, WNT and FGF proteins interact to pattern the cerebral cortex. Development 2004, 131, 5639–5647. [Google Scholar] [CrossRef] [PubMed]
  136. Lun, M.P.; Johnson, M.B.; Broadbelt, K.G.; Watanabe, M.; Kang, Y.-J.; Chau, K.F.; Springel, M.W.; Malesz, A.; Sousa, A.M.; Pletikos, M.; et al. Spatially heterogeneous choroid plexus transcriptomes encode positional identity and contribute to regional CSF production. J. Neurosci. 2015, 35, 4903–4916. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  137. Hunter, N.L.; Dymecki, S.M. Molecularly and temporally separable lineages form the hindbrain roof plate and contribute differentially to the choroid plexus. Development 2007, 134, 3449–3460. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Bartke, A.; Chandrashekar, V.; Dominici, F.; Turyn, D.; Kinney, B.; Steger, R.; Kopchick, J. Insulin-like growth factor 1 (IGF-1) and aging: Controversies and new insights. Biogerontology 2003, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  139. Ziegler, A.N.; Schneider, J.S.; Qin, M.; Tyler, W.A.; Pintar, J.E.; Fraidenraich, D.; Wood, T.L.; Levison, S.W. IGF-II promotes stemness of neural restricted precursors. Stem Cells 2012, 30, 1265–1276. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  140. Chen, R.L.; Kassem, N.A.; Sadeghi, M.; Preston, J.E. Insulin-like growth factor-II uptake into choroid plexus and brain of young and old sheep. J. Gerontol. A Biol. Sci. Med. Sci. 2008, 63, 141–148. [Google Scholar] [CrossRef] [PubMed]
  141. Lozano-Ureña, A.; Lázaro-Carot, L.; Jiménez-Villalba, E.; Montalbán-Loro, R.; Mateos-White, I.; Duart-Abadía, P.; Martínez-Gurrea, I.; Nakayama, K.I.; Fariñas, I.; Kirstein, M.; et al. IGF2 interacts with the imprinted gene Cdkn1c to promote terminal differentiation of neural stem cells. Development 2023, 150, dev200563. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  142. Deng, Y.; Wang, L.; Ge, L.; Duan, D.; Zhuo, Y.; Yuan, T.; Yan, W.; Huang, P.; Teng, X.; Lu, M. Effects of IGFBP-2 on proliferation and differentiation in neural stem cell line C17.2. J. Neurorestoratology 2017, 5, 143–153. [Google Scholar] [CrossRef]
  143. Shahin, W.S.; Ebed, S.O.; Tyler, S.R.; Miljkovic, B.; Choi, S.H.; Zhang, Y.; Zhou, W.; Evans, I.A.; Yeaman, C.; Engelhardt, J.F. Redox-dependent Igfbp2 signaling controls Brca1 DNA damage response to govern neural stem cell fate. Nat. Commun. 2023, 14, 444. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  144. Iqbal, M.A.; Eftekharpour, E. Regulatory Role of Redox Balance in Determination of Neural Precursor Cell Fate. Stem Cells Int. 2017, 2017, 9209127. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  145. Nickenig, G.; Baudler, S.; Müller, C.; Werner, C.; Werner, N.; Welzel, H.; Strehlow, K.; Böhm, M. Redox-sensitive vascular smooth muscle cell proliferation is mediated by GKLF and Id3 in vitro and in vivo. FASEB J. 2002, 16, 1077–1086. [Google Scholar] [CrossRef] [PubMed]
  146. Ghosh, M.; Shubham, S.; Mandal, K.; Trivedi, V.; Chauhan, R.; Naseera, S. Survival and prognostic factors for glioblastoma multiforme: Retrospective single-institutional study. Indian J. Cancer 2017, 54, 362–367. [Google Scholar] [CrossRef] [PubMed]
  147. Peeters, M.C.M.; Dirven, L.; Koekkoek, J.A.F.; Gortmaker, E.G.; Fritz, L.; Vos, M.J.; Taphoorn, M.J.B. Prediagnostic symptoms and signs of adult glioma: The patients’ view. J. Neurooncol. 2020, 146, 293–301. [Google Scholar] [CrossRef] [PubMed]
  148. Ohgaki, H.; Kleihues, P. Genetic pathways to primary and secondary glioblastoma. Am. J. Pathol. 2007, 170, 1445–1453. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  149. 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] [PubMed] [PubMed Central]
  150. 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]
  151. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro-Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  152. 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] [PubMed]
  153. Furth, J.; Kahn, C.M.; Breedis, C. The Transmission of Leukemia of Mice with a Single Cell. Cancer Res. 1937, 31, 276–282. [Google Scholar]
  154. 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] [PubMed] [PubMed Central]
  155. Collins, A.T.; Berry, P.A.; Hyde, C.; Stower, M.J.; Maitland, N.J. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005, 65, 10946–10951. [Google Scholar] [CrossRef] [PubMed]
  156. 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] [PubMed] [PubMed Central]
  157. Li, C.; Heidt, D.G.; Dalerba, P.; Burant, C.F.; Zhang, L.; Adsay, V.; Wicha, M.; Clarke, M.F.; Simeone, D.M. Identification of pancreatic cancer stem cells. Cancer Res. 2007, 67, 1030–1037. [Google Scholar] [CrossRef] [PubMed]
  158. O’Brien, C.A.; Pollett, A.; Gallinger, S.; Dick, J.E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007, 445, 106–110. [Google Scholar] [CrossRef] [PubMed]
  159. 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, 111–115. [Google Scholar] [CrossRef] [PubMed]
  160. 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] [PubMed]
  161. 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] [PubMed]
  162. Bachoo, R.M.; Maher, E.A.; Ligon, K.L.; Sharpless, N.E.; Chan, S.S.; You, M.J.; Tang, Y.; DeFrances, J.; Stover, E.; Weissleder, R.; et al. Epidermal growth factor receptor and Ink4a/Arf: Convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis. Cancer Cell 2002, 1, 269–277. [Google Scholar] [CrossRef] [PubMed]
  163. 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] [PubMed] [PubMed Central]
  164. Sutter, R.; Yadirgi, G.; Marino, S. Neural stem cells, tumour stem cells and brain tumours: Dangerous relationships? Biochim. Biophys. Acta 2007, 1776, 125–137. [Google Scholar] [CrossRef] [PubMed]
  165. 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] [PubMed]
  166. MacLeod, G.; Bozek, D.A.; Rajakulendran, N.; Monteiro, V.; Ahmadi, M.; Steinhart, Z.; Kushida, M.M.; Yu, H.; Coutinho, F.J.; Cavalli, F.M.G.; et al. Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Rep. 2019, 27, 971–986.e9. [Google Scholar] [CrossRef] [PubMed]
  167. 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] [PubMed] [PubMed Central]
  168. Llaguno, S.A.; 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] [PubMed Central]
  169. Llaguno, S.R.A.; Wang, Z.; Sun, D.; Chen, J.; Xu, J.; Kim, E.; Hatanpaa, K.J.; Raisanen, J.M.; Burns, D.K.; Johnson, J.E.; et al. Adult Lineage-Restricted CNS Progenitors Specify Distinct Glioblastoma Subtypes. Cancer Cell 2015, 28, 429–440. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  170. Chow, L.M.; Endersby, R.; Zhu, X.; Rankin, S.; Qu, C.; Zhang, J.; Broniscer, A.; Ellison, D.W.; Baker, S.J. Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. Cancer Cell 2011, 19, 305–316. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  171. Galvao, R.P.; Kasina, A.; McNeill, R.S.; Harbin, J.E.; Foreman, O.; Verhaak, R.G.W.; Nishiyama, A.; Miller, C.R.; Zong, H. Transformation of quiescent adult oligodendrocyte precursor cells into malignant glioma through a multistep reactivation process. Proc. Natl. Acad. Sci. USA 2014, 111, E4214–E4223. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  172. 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] [PubMed] [PubMed Central]
  173. Assanah, M.; Lochhead, R.; Ogden, A.; Bruce, J.; Goldman, J.; Canoll, P. Glial progenitors in adult white matter are driven to form malignant gliomas by platelet-derived growth factor-expressing retroviruses. J. Neurosci. 2006, 26, 6781–6790. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  174. Dawson, M.R.; Polito, A.; Levine, J.M.; Reynolds, R. NG2-expressing glial progenitor cells: An abundant and widespread population of cycling cells in the adult rat CNS. Mol. Cell Neurosci. 2003, 24, 476–488. [Google Scholar] [CrossRef] [PubMed]
  175. Dimou, L.; Simon, C.; Kirchhoff, F.; Takebayashi, H.; Götz, M. Progeny of Olig2-expressing progenitors in the gray and white matter of the adult mouse cerebral cortex. J. Neurosci. 2008, 28, 10434–10442. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  176. Ligon, K.L.; Huillard, E.; Mehta, S.; Kesari, S.; Liu, H.; Alberta, J.A.; Bachoo, R.M.; Kane, M.; Louis, D.N.; DePinho, R.A.; et al. Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma. Neuron 2007, 53, 503–517. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  177. Jackson, E.L.; Garcia-Verdugo, J.M.; Gil-Perotin, S.; Roy, M.; Quinones-Hinojosa, A.; VandenBerg, S.; Alvarez-Buylla, A. PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling. Neuron 2006, 51, 187–199. [Google Scholar] [CrossRef] [PubMed]
  178. Ligon, K.L.; Alberta, J.A.; Kho, A.T.; Weiss, J.; Kwaan, M.R.; Nutt, C.L.; Louis, D.N.; Stiles, C.D.; Rowitch, D.H. The oligodendroglial lineage marker OLIG2 is universally expressed in diffuse gliomas. J. Neuropathol. Exp. Neurol. 2004, 63, 499–509. [Google Scholar] [CrossRef] [PubMed]
  179. Lu, Q.R.; Park, J.K.; Noll, E.; Chan, J.A.; Alberta, J.; Yuk, D.; Alzamora, M.G.; Louis, D.N.; Stiles, C.D.; Rowitch, D.H.; et al. Oligodendrocyte lineage genes (OLIG) as molecular markers for human glial brain tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 10851–10856. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  180. 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]
  181. The Telomeres Mendelian Randomization Collaboration; Haycock, P.C.; Burgess, S.; Nounu, A.; Zheng, J.; Okoli, G.N.; Bowden, J.; Wade, K.H.; Timpson, N.J.; Evans, D.M.; et al. Association Between Telomere Length and Risk of Cancer and Non-Neoplastic Diseases: A Mendelian Randomization Study. JAMA Oncol. 2017, 3, 636–651. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  182. Howell, A.E.; Robinson, J.W.; Wootton, R.E.; McAleenan, A.; Tsavachidis, S.; Ostrom, Q.T.; Bondy, M.; Armstrong, G.; Relton, C.; Haycock, P.; et al. Testing for causality between systematically identified risk factors and glioma: A Mendelian randomization study. BMC Cancer 2020, 20, 508. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  183. Fontán-Lozano, Á.; Morcuende, S.; Davis-López de Carrizosa, M.A.; Benítez-Temiño, B.; Mejías, R.; Matarredona, E.R. To Become or Not to Become Tumorigenic: Subventricular Zone. Front. Oncol. 2020, 10, 602217. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  184. Carrano, A.; Zarco, N.; Phillipps, J.; Lara-Velazquez, M.; Suarez-Meade, P.; Norton, E.S.; Chaichana, K.L.; Quiñones-Hinojosa, A.; Asmann, Y.W.; Guerrero-Cázares, H. Human Cerebrospinal Fluid Modulates Pathways Promoting Glioblastoma Malignancy. Front. Oncol. 2021, 11, 624145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  185. 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] [PubMed] [PubMed Central]
  186. Norton, E.S.; Whaley, L.A.; Ulloa-Navas, M.J.; García-Tárraga, P.; Meneses, K.M.; Lara-Velazquez, M.; Zarco, N.; Carrano, A.; Quiñones-Hinojosa, A.; García-Verdugo, J.M.; et al. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022, 19, 58. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  187. Adeberg, S.; Bostel, T.; König, L.; Welzel, T.; Debus, J.; Combs, S.E. A comparison of long-term survivors and short-term survivors with glioblastoma, subventricular zone involvement: A predictive factor for survival? Radiat. Oncol. 2014, 9, 95. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  188. 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] [PubMed]
  189. Jafri, N.F.; Clarke, J.L.; Weinberg, V.; Barani, I.J.; Cha, S. Relationship of glioblastoma multiforme to the subventricular zone is associated with survival. Neuro-Oncol. 2013, 15, 91–96. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  190. Young, G.S.; Macklin, E.A.; Setayesh, K.; Lawson, J.D.; Wen, P.Y.; Norden, A.D.; Drappatz, J.; Kesari, S. Longitudinal MRI evidence for decreased survival among periventricular glioblastoma. J. Neurooncol. 2011, 104, 261–269. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  191. Mummareddy, N.; Salwi, S.R.; Kumar, N.G.; Zhao, Z.; Ye, F.; Le, C.H.; Mobley, B.C.; Thompson, R.C.; Chambless, L.B.; Mistry, A.M. Prognostic relevance of CSF and peri-tumoral edema volumes in glioblastoma. J. Clin. Neurosci. 2021, 84, 1–7. [Google Scholar] [CrossRef] [PubMed]
  192. 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] [PubMed] [PubMed Central]
  193. Sonoda, Y.; Saito, R.; Kanamori, M.; Kumabe, T.; Uenohara, H.; Tominaga, T. The association of subventricular zone involvement at recurrence with survival after repeat surgery in patients with recurrent glioblastoma. Neurol. Med.-Chir. 2014, 54, 302–309. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  194. Alonso, M.I.; Martín, C.; Carnicero, E.; Bueno, D.; Gato, A. Cerebrospinal fluid control of neurogenesis induced by retinoic acid during early brain development. Dev. Dyn. 2011, 240, 1650–1659. [Google Scholar] [CrossRef] [PubMed]
  195. Chang, J.T.; Lehtinen, M.K.; Sive, H. Zebrafish cerebrospinal fluid mediates cell survival through a retinoid signaling pathway. Dev. Neurobiol. 2016, 76, 75–92. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  196. Huang, X.; Liu, J.; Ketova, T.; Fleming, J.T.; Grover, V.K.; Cooper, M.K.; Litingtung, Y.; Chiang, C. Transventricular delivery of Sonic hedgehog is essential to cerebellar ventricular zone development. Proc. Natl. Acad. Sci. USA 2010, 107, 8422–8427. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  197. Kaiser, K.; Gyllborg, D.; Procházka, J.; Salašová, A.; Kompaníková, P.; Molina, F.L.; Laguna-Goya, R.; Radaszkiewicz, T.; Harnoš, J.; Procházková, M.; et al. WNT5A is transported via lipoprotein particles in the cerebrospinal fluid to regulate hindbrain morphogenesis. Nat. Commun. 2019, 10, 1498. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  198. Martín, C.; Bueno, D.; Alonso, M.; Moro, J.; Callejo, S.; Parada, C.; Martín, P.; Carnicero, E.; Gato, A. FGF2 plays a key role in embryonic cerebrospinal fluid trophic properties over chick embryo neuroepithelial stem cells. Dev. Biol. 2006, 297, 402–416. [Google Scholar] [CrossRef] [PubMed]
  199. Hamilton, L.K.; Dufresne, M.; Joppé, S.E.; Petryszyn, S.; Aumont, A.; Calon, F.; Barnabé-Heider, F.; Furtos, A.; Parent, M.; Chaurand, P.; et al. Aberrant Lipid Metabolism in the Forebrain Niche Suppresses Adult Neural Stem Cell Proliferation in an Animal Model of Alzheimer’s Disease. Cell Stem Cell 2015, 17, 397–411. [Google Scholar] [CrossRef] [PubMed]
  200. Muthusamy, N.; Sommerville, L.J.; Moeser, A.J.; Stumpo, D.J.; Sannes, P.; Adler, K.; Blackshear, P.J.; Weimer, J.M.; Ghashghaei, H.T. MARCKS-dependent mucin clearance and lipid metabolism in ependymal cells are required for maintenance of forebrain homeostasis during aging. Aging Cell 2015, 14, 764–773. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  201. Banizs, B.; Pike, M.M.; Millican, C.L.; Ferguson, W.B.; Komlosi, P.; Sheetz, J.; Bell, P.D.; Schwiebert, E.M.; Yoder, B.K. Dysfunctional cilia lead to altered ependyma and choroid plexus function, and result in the formation of hydrocephalus. Development 2005, 132, 5329–5339. [Google Scholar] [CrossRef] [PubMed]
  202. Olstad, E.W.; Ringers, C.; Hansen, J.N.; Wens, A.; Brandt, C.; Wachten, D.; Yaksi, E.; Jurisch-Yaksi, N. Ciliary Beating Compartmentalizes Cerebrospinal Fluid Flow in the Brain and Regulates Ventricular Development. Curr. Biol. 2019, 29, 229–241.e6. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  203. Siyahhan, B.; Knobloch, V.; de Zélicourt, D.; Asgari, M.; Daners, M.S.; Poulikakos, D.; Kurtcuoglu, V. Flow induced by ependymal cilia dominates near-wall cerebrospinal fluid dynamics in the lateral ventricles. J. R. Soc. Interface 2014, 11, 20131189. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  204. Rickman, D.S.; Bobek, M.P.; Misek, D.E.; Kuick, R.; Blaivas, M.; Kurnit, D.M.; Taylor, J.; Hanash, S.M. Distinctive molecular profiles of high-grade and low-grade gliomas based on oligonucleotide microarray analysis. Cancer Res. 2001, 61, 6885–6891. [Google Scholar] [PubMed]
  205. Freije, W.A.; Castro-Vargas, F.E.; Fang, Z.; Horvath, S.; Cloughesy, T.; Liau, L.M.; Mischel, P.S.; Nelson, S.F. Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 2004, 64, 6503–6510. [Google Scholar] [CrossRef] [PubMed]
  206. Nigro, J.M.; Misra, A.; Zhang, L.; Smirnov, I.; Colman, H.; Griffin, C.; Ozburn, N.; Chen, M.; Pan, E.; Koul, D.; et al. Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res. 2005, 65, 1678–1686. [Google Scholar] [CrossRef] [PubMed]
  207. Nutt, C.L.; Mani, D.R.; Betensky, R.A.; Tamayo, P.; Cairncross, J.G.; Ladd, C.; Pohl, U.; Hartmann, C.; E McLaughlin, M.; Batchelor, T.T.; et al. Gene expression-based classification of malignant gliomas correlates better with survival than histological classification. Cancer Res. 2003, 63, 1602–1607. [Google Scholar] [PubMed]
  208. Phillips, H.S.; Kharbanda, S.; Chen, R.; Forrest, W.F.; Soriano, R.H.; Wu, T.D.; Misra, A.; Nigro, J.M.; Colman, H.; Soroceanu, L.; et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006, 9, 157–173. [Google Scholar] [CrossRef] [PubMed]
  209. McDonald, K.L.; O’Sullivan, M.G.; Parkinson, J.F.; Shaw, J.M.; Payne, C.A.; Brewer, J.M.; Young, L.; Reader, D.J.; Wheeler, H.T.; Cook, R.J.; et al. IQGAP1 and IGFBP2: Valuable biomarkers for determining prognosis in glioma patients. J. Neuropathol. Exp. Neurol. 2007, 66, 405–417. [Google Scholar] [CrossRef] [PubMed]
  210. Colman, H.; Zhang, L.; Sulman, E.P.; McDonald, J.M.; Shooshtari, N.L.; Rivera, A.; Popoff, S.; Nutt, C.L.; Louis, D.N.; Cairncross, J.G.; et al. A multigene predictor of outcome in glioblastoma. Neuro-Oncol. 2010, 12, 49–57. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  211. Ceccarelli, M.; Barthel, F.P.; Malta, T.M.; Sabedot, T.S.; Salama, S.R.; Murray, B.A.; Morozova, O.; Newton, Y.; Radenbaugh, A.; Pagnotta, S.M.; et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016, 164, 550–563. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  212. Prasad, B.; Tian, Y.; Li, X. Large-Scale Analysis Reveals Gene Signature for Survival Prediction in Primary Glioblastoma. Mol. Neurobiol. 2020, 57, 5235–5246. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  213. Yin, W.; Tang, G.; Zhou, Q.; Cao, Y.; Li, H.; Fu, X.; Wu, Z.; Jiang, X. Expression Profile Analysis Identifies a Novel Five-Gene Signature to Improve Prognosis Prediction of Glioblastoma. Front. Genet. 2019, 10, 419. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  214. Yu, Z.; Du, M.; Lu, L. A Novel 16-Genes Signature Scoring System as Prognostic Model to Evaluate Survival Risk in Patients with Glioblastoma. Biomedicines 2022, 10, 317. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  215. Liu, Y.; Shen, S.; Yan, Z.; Yan, L.; Ding, H.; Wang, A.; Xu, Q.; Sun, L.; Yuan, Y. Expression characteristics and their functional role of IGFBP gene family in pan-cancer. BMC Cancer 2023, 23, 371. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  216. Fukushima, T.; Tezuka, T.; Shimomura, T.; Nakano, S.; Kataoka, H. Silencing of insulin-like growth factor-binding protein-2 in human glioblastoma cells reduces both invasiveness and expression of progression-associated gene CD24. J. Biol. Chem. 2007, 282, 18634–18644. [Google Scholar] [CrossRef] [PubMed]
  217. Patil, S.S.; Gokulnath, P.; Bashir, M.; Shwetha, S.D.; Jaiswal, J.; Shastry, A.H.; Arimappamagan, A.; Santosh, V.; Kondaiah, P. Insulin-like growth factor binding protein-2 regulates β-catenin signaling pathway in glioma cells and contributes to poor patient prognosis. Neuro-Oncol. 2016, 18, 1487–1497. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  218. Sehgal, P.; Kumar, N.; Kumar, V.R.P.; Patil, S.; Bhattacharya, A.; Kumar, M.V.; Mukherjee, G.; Kondaiah, P. Regulation of protumorigenic pathways by insulin like growth factor binding protein2 and its association along with β-catenin in breast cancer lymph node metastasis. Mol. Cancer 2013, 12, 63. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  219. Chen, X.; Zhang, Y.; Zhang, P.; Wei, M.; Tian, T.; Guan, Y.; Han, C.; Wei, W.; Ma, Y. IGFBP2 drives epithelial-mesenchymal transition in hepatocellular carcinoma via activating the Wnt/β-catenin pathway. Infect. Agent. Cancer 2023, 18, 73. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  220. Hsieh, D.; Hsieh, A.; Stea, B.; Ellsworth, R. IGFBP2 promotes glioma tumor stem cell expansion and survival. Biochem. Biophys. Res. Commun. 2010, 397, 367–372. [Google Scholar] [CrossRef] [PubMed]
  221. Robilliard, L.D.; Yu, J.; Anchan, A.; Finlay, G.; Angel, C.E.; Graham, E.S. Comprehensive Assessment of Secreted Immuno-Modulatory Cytokines by Serum-Differentiated and Stem-like Glioblastoma Cells Reveals Distinct Differences between Glioblastoma Phenotypes. Int. J. Mol. Sci. 2022, 23, 14164. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  222. Berezovsky, A.D.; Poisson, L.M.; Cherba, D.; Webb, C.P.; Transou, A.D.; Lemke, N.W.; Hong, X.; Hasselbach, L.A.; Irtenkauf, S.M.; Mikkelsen, T.; et al. Sox2 promotes malignancy in glioblastoma by regulating plasticity and astrocytic differentiation. Neoplasia 2014, 16, 193–206.e25. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  223. Kunhiraman, H.; McSwain, L.; Shahab, S.W.; Gershon, T.R.; MacDonald, T.J.; Kenney, A.M. IGFBP2 promotes proliferation and cell migration through STAT3 signaling in Sonic hedgehog medulloblastoma. Acta Neuropathol. Commun. 2023, 11, 62. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  224. Vanner, R.J.; Remke, M.; Gallo, M.; Selvadurai, H.J.; Coutinho, F.; Lee, L.; Kushida, M.; Head, R.; Morrissy, S.; Zhu, X.; et al. Quiescent sox2(+) cells drive hierarchical growth and relapse in sonic hedgehog subgroup medulloblastoma. Cancer Cell 2014, 26, 33–47. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  225. Friend, K.E.; Khandwala, H.M.; Flyvbjerg, A.; Hill, H.; Li, J.; McCutcheon, I.E. Growth hormone and insulin-like growth factor-I: Effects on the growth of glioma cell lines. Growth Horm. IGF Res. 2001, 11, 84–91. [Google Scholar] [CrossRef] [PubMed]
  226. Schlenska-Lange, A.; Knüpfer, H.; Lange, T.J.; Kiess, W.; Knüpfer, M. Cell proliferation and migration in glioblastoma multiforme cell lines are influenced by insulin-like growth factor I in vitro. Anticancer Res. 2008, 28, 1055–1060. [Google Scholar] [PubMed]
  227. Zamykal, M.; Martens, T.; Matschke, J.; Günther, H.S.; Kathagen, A.; Schulte, A.; Peters, R.; Westphal, M.; Lamszus, K. Inhibition of intracerebral glioblastoma growth by targeting the insulin-like growth factor 1 receptor involves different context-dependent mechanisms. Neuro-Oncol. 2015, 17, 1076–1085. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  228. Antoniades, H.N.; Galanopoulos, T.; Neville-Golden, J.; Maxwell, M. Expression of insulin-like growth factors I and II and their receptor mRNAs in primary human astrocytomas and meningiomas; in vivo studies using in situ hybridization and immunocytochemistry. Int. J. Cancer 1992, 50, 215–222. [Google Scholar] [CrossRef] [PubMed]
  229. Hirano, H.; Lopes, M.B.S.; Laws, E.R.; Asakura, T.; Goto, M.; Carpenter, J.E.; Karns, L.R.; VandenBerg, S.R. Insulin-like growth factor-1 content and pattern of expression correlates with histopathologic grade in diffusely infiltrating astrocytomas. Neuro-Oncol. 1999, 1, 109–119. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  230. Merrill, M.J.; Edwards, N.A. Insulin-like growth factor-I receptors in human glial tumors. J. Clin. Endocrinol. Metab. 1990, 71, 199–209. [Google Scholar] [CrossRef] [PubMed]
  231. Soroceanu, L.; Kharbanda, S.; Chen, R.; Soriano, R.H.; Aldape, K.; Misra, A.; Zha, J.; Forrest, W.F.; Nigro, J.M.; Modrusan, Z.; et al. Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma. Proc. Natl. Acad. Sci. USA 2007, 104, 3466–3471. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  232. Suvasini, R.; Shruti, B.; Thota, B.; Shinde, S.V.; Friedmann-Morvinski, D.; Nawaz, Z.; Prasanna, K.V.; Thennarasu, K.; Hegde, A.S.; Arivazhagan, A.; et al. Insulin growth factor-2 binding protein 3 (IGF2BP3) is a glioblastoma-specific marker that activates phosphatidylinositol 3-kinase/mitogen-activated protein kinase (PI3K/MAPK) pathways by modulating IGF-2. J. Biol. Chem. 2011, 286, 25882–25890. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  233. Sandberg-Nordqvist, A.C.; Ståhlbom, P.A.; Reinecke, M.; Collins, V.P.; von Holst, H.; Sara, V. Characterization of insulin-like growth factor 1 in human primary brain tumors. Cancer Res. 1993, 53, 2475–2478. [Google Scholar] [PubMed]
  234. Sandberg, A.C.; Engberg, C.; Lake, M.; von Holst, H.; Sara, V.R. The expression of insulin-like growth factor I and insulin-like growth factor II genes in the human fetal and adult brain and in glioma. Neurosci. Lett. 1988, 93, 114–119. [Google Scholar] [CrossRef] [PubMed]
  235. Dmitrenko, V.V.; Kavsan, V.M.; Boyko, O.I.; Rymar, V.I.; Stepanenko, A.A.; Balynska, O.V.; Mausheva, T.A.; Rozumenko, V.D.; Zozulya, Y.P. Expression of genes belonging to the IGF-system in glial tumors. Tsitol Genet. 2011, 45, 41–57. [Google Scholar] [CrossRef] [PubMed]
  236. Kavsan, V.; Shostak, K.; Dmitrenko, V.; Zozulya, Y.; Rozumenko, V.; Demotes-Mainard, J. Characterization of genes with increased expression in human glioblastomas. Tsitol Genet. 2005, 39, 37–49. [Google Scholar] [PubMed]
  237. Maris, C.; D’Haene, N.; Trépant, A.-L.; Le Mercier, M.; Sauvage, S.; Allard, J.; Rorive, S.; Demetter, P.; Decaestecker, C.; Salmon, I. IGF-IR: A new prognostic biomarker for human glioblastoma. Br. J. Cancer 2015, 113, 729–737. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  238. Mohlin, S.; Hamidian, A.; Påhlman, S. HIF2A and IGF2 expression correlates in human neuroblastoma cells and normal immature sympathetic neuroblasts. Neoplasia 2013, 15, 328–334. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  239. Mu, Q.; Wang, L.; Yu, F.; Gao, H.; Lei, T.; Li, P.; Liu, P.; Zheng, X.; Hu, X.; Chen, Y.; et al. Imp2 regulates GBM progression by activating IGF2/PI3K/Akt pathway. Cancer Biol. Ther. 2015, 16, 623–633. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  240. Bendall, S.C.; Stewart, M.H.; Menendez, P.; George, D.; Vijayaragavan, K.; Werbowetski-Ogilvie, T.; Ramos-Mejia, V.; Rouleau, A.; Yang, J.; Bossé, M.; et al. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature 2007, 448, 1015–1021. [Google Scholar] [CrossRef] [PubMed]
  241. Patel, A.P.; Tirosh, I.; Trombetta, J.J.; Shalek, A.K.; Gillespie, S.M.; Wakimoto, H.; Cahill, D.P.; Nahed, B.V.; Curry, W.T.; Martuza, R.L.; et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014, 344, 1396–1401. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  242. 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] [PubMed] [PubMed Central]
  243. 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, 522–526. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  244. Ferguson, K.M.; Blin, C.; Alfazema, N.; Gangoso, E.; Pollard, S.M.; Marques-Torrejon, M.A. Lrig1 regulates the balance between proliferation and quiescence in glioblastoma stem cells. Front. Cell Dev. Biol. 2022, 10, 983097. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  245. Marqués-Torrejón, M.Á.; Williams, C.A.C.; Southgate, B.; Alfazema, N.; Clements, M.P.; Garcia-Diaz, C.; Blin, C.; Arranz-Emparan, N.; Fraser, J.; Gammoh, N.; et al. LRIG1 is a gatekeeper to exit from quiescence in adult neural stem cells. Nat. Commun. 2021, 12, 2594. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  246. Elmlinger, M.W.; Deininger, M.H.; Schuett, B.S.; Meyermann, R.; Duffner, F.; Grote, E.H.; Ranke, M.B. In vivo expression of insulin-like growth factor-binding protein-2 in human gliomas increases with the tumor grade. Endocrinology 2001, 142, 1652–1658. [Google Scholar] [CrossRef] [PubMed]
  247. Fuller, G.N.; Rhee, C.H.; Hess, K.R.; Caskey, L.S.; Wang, R.; Bruner, J.M.; Yung, W.K.; Zhang, W. Reactivation of insulin-like growth factor binding protein 2 expression in glioblastoma multiforme: A revelation by parallel gene expression profiling. Cancer Res. 1999, 59, 4228–4232. [Google Scholar] [PubMed]
  248. Tseng, L.Y.-H.; Brown, A.L.; Yang, Y.W.-H.; Romanus, J.A.; Orlowski, C.C.; Taylor, T.; Rechler, M.M. The fetal rat binding protein for insulin-like growth factors is expressed in the choroid plexus and cerebrospinal fluid of adult rats. Mol. Endocrinol. 1989, 3, 1559–1568. [Google Scholar] [CrossRef] [PubMed]
  249. Wood, T.L.; Streck, R.D.; Pintar, J.E. Expression of the IGFBP-2 gene in post-implantation rat embryos. Development 1992, 114, 59–66. [Google Scholar] [CrossRef] [PubMed]
  250. van Straaten, H.W.; Hekking, J.W.; Wiertz-Hoessels, E.J.; Thors, F.; Drukker, J. Effect of the notochord on the differentiation of a floor plate area in the neural tube of the chick embryo. Anat. Embryol. 1988, 177, 317–324. [Google Scholar] [CrossRef] [PubMed]
  251. Yamada, T.; Placzek, M.; Tanaka, H.; Dodd, J.; Jessell, T.M. Control of cell pattern in the developing nervous system: Polarizing activity of the floor plate and notochord. Cell 1991, 64, 635–647. [Google Scholar] [CrossRef] [PubMed]
  252. Shen, F.; Song, C.; Liu, Y.; Zhang, J.; Wei Song, S. IGFBP2 promotes neural stem cell maintenance and proliferation differentially associated with glioblastoma subtypes. Brain Res. 2019, 1704, 174–186. [Google Scholar] [CrossRef] [PubMed]
  253. Kaplan, M.P.; Chin, S.S.; Fliegner, K.H.; Liem, R.K. Alpha-internexin, a novel neuronal intermediate filament protein, precedes the low molecular weight neurofilament protein (NF-L) in the developing rat brain. J. Neurosci. 1990, 10, 2735–2748. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  254. Burgdorf, J.S.; Yoon, S.; Dos Santos, M.; Lammert, C.R.; Moskal, J.R.; Penzes, P. An IGFBP2-derived peptide promotes neuroplasticity and rescues deficits in a mouse model of Phelan-McDermid syndrome. Mol. Psychiatry 2023, 28, 1101–1111. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  255. Holmin, S.; Mathiesen, T.; Langmoen, I.A.; Sandberg Nordqvist, A.C. Depolarization induces insulin-like growth factor binding protein-2 expression in vivo via NMDA receptor stimulation. Growth Horm. IGF Res. 2001, 11, 399–406. [Google Scholar] [CrossRef] [PubMed]
  256. Khan, S.; Lu, X.; Huang, Q.; Tang, J.; Weng, J.; Yang, Z.; Lv, M.; Xu, X.; Xia, F.; Zhang, M.; et al. IGFBP2 Plays an Essential Role in Cognitive Development during Early Life. Adv. Sci. 2019, 6, 1901152. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  257. van Doorn, J. Insulin-like growth factor-II and bioactive proteins containing a part of the E-domain of pro-insulin-like growth factor-II. Biofactors 2020, 46, 563–578. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  258. Greenall, S.A.; Bentley, J.D.; Pearce, L.A.; Scoble, J.A.; Sparrow, L.G.; Bartone, N.A.; Xiao, X.; Baxter, R.C.; Cosgrove, L.J.; Adams, T.E. Biochemical characterization of individual human glycosylated pro-insulin-like growth factor (IGF)-II and big-IGF-II isoforms associated with cancer. J. Biol. Chem. 2013, 288, 59–68. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  259. PotalPotalitsyn, P.; Mrázková, L.; Selicharová, I.; Tencerová, M.; Ferenčáková, M.; Chrudinová, M.; Turnovská, T.; Brzozowski, A.M.; Marek, A.; Kaminský, J.; et al. Non-glycosylated IGF2 prohormones are more mitogenic than native IGF2. Commun. Biol. 2023, 6, 863. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  260. Frasca, F.; Pandini, G.; Scalia, P.; Sciacca, L.; Mineo, R.; Costantino, A.; Goldfine, I.D.; Belfiore, A.; Vigneri, R. Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol. Cell Biol. 1999, 19, 3278–3288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  261. Huang, J.; Morehouse, C.; Streicher, K.; Higgs, B.W.; Gao, J.; Czapiga, M.; Boutrin, A.; Zhu, W.; Brohawn, P.; Chang, Y.; et al. Altered expression of insulin receptor isoforms in breast cancer. PLoS ONE 2011, 6, e26177. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  262. Vella, V.; Milluzzo, A.; Scalisi, N.M.; Vigneri, P.; Sciacca, L. Insulin Receptor Isoforms in Cancer. Int. J. Mol. Sci. 2018, 19, 3615. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  263. Benabou, E.; Salamé, Z.; Wendum, D.; Lequoy, M.; Tahraoui, S.; Merabtene, F.; Chrétien, Y.; Scatton, O.; Rosmorduc, O.; Fouassier, L.; et al. Insulin receptor isoform A favors tumor progression in human hepatocellular carcinoma by increasing stem/progenitor cell features. Cancer Lett. 2019, 450, 155–168. [Google Scholar] [CrossRef] [PubMed]
  264. Martinez-Quetglas, I.; Pinyol, R.; Dauch, D.; Torrecilla, S.; Tovar, V.; Moeini, A.; Alsinet, C.; Portela, A.; Rodriguez-Carunchio, L.; Solé, M.; et al. IGF2 Is Up-regulated by Epigenetic Mechanisms in Hepatocellular Carcinomas and Is an Actionable Oncogene Product in Experimental Models. Gastroenterology 2016, 151, 1192–1205. [Google Scholar] [CrossRef] [PubMed]
  265. Song, P.; Kwon, Y.; Joo, J.Y.; Kim, D.G.; Yoon, J.H. Secretomics to Discover Regulators in Diseases. Int. J. Mol. Sci. 2019, 20, 3893. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  266. Martin, R.M.; Holly, J.M.; Davey Smith, G.; Gunnell, D. Associations of adiposity from childhood into adulthood with insulin resistance and the insulin-like growth factor system: 65-year follow-up of the Boyd Orr Cohort. J. Clin. Endocrinol. Metab. 2006, 91, 3287–3295. [Google Scholar] [CrossRef] [PubMed]
  267. Ziegler, A.N.; Feng, Q.; Chidambaram, S.; Testai, J.M.; Kumari, E.; Rothbard, D.E.; Constancia, M.; Sandovici, I.; Cominski, T.; Pang, K.; et al. Insulin-like Growth Factor II: An Essential Adult Stem Cell Niche Constituent in Brain and Intestine. Stem Cell Rep. 2019, 12, 816–830. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Figure 1. The insulin-like growth factor (IGF)/insulin axis: receptors and ligands. Receptors include transmembrane receptors (the IGF-1 receptor (IGF-1R) and the insulin receptor (IR)), hybrid heterodimers (IR-A and IR-B) and hybrid tetramers (IGF-1R/IR). Hybrid receptors are formed from the heterodimerisation of IGF-1R and IR subunits. Ligands include insulin, IGF-I, IGF-II and their variants (high-molecular-weight IGF-II). IGF-2R/mannose 6-phosphate receptor (M6PR) is a transmembrane glycoprotein without kinase activity. Relative binding affinities for each receptor–ligand interaction differ. Insulin binding affinity is strongest for the IR-A and IR-B homodimers and weaker for heterodimeric receptors. IGF-I binding affinity is strongest for the IGF-1R, IGF-1R/IR-A and IGF-1R/IR-B heterodimers. IGF-II binding affinity is strongest for the IGF-1R, IR-A and IGF/IR-A heterodimeric receptor. Overarching biological effects for receptor activation via these pathways include the modulation of metabolism and mitogenesis. Figure adapted from [27,28,29,30]. Created with BioRender.com.
Figure 1. The insulin-like growth factor (IGF)/insulin axis: receptors and ligands. Receptors include transmembrane receptors (the IGF-1 receptor (IGF-1R) and the insulin receptor (IR)), hybrid heterodimers (IR-A and IR-B) and hybrid tetramers (IGF-1R/IR). Hybrid receptors are formed from the heterodimerisation of IGF-1R and IR subunits. Ligands include insulin, IGF-I, IGF-II and their variants (high-molecular-weight IGF-II). IGF-2R/mannose 6-phosphate receptor (M6PR) is a transmembrane glycoprotein without kinase activity. Relative binding affinities for each receptor–ligand interaction differ. Insulin binding affinity is strongest for the IR-A and IR-B homodimers and weaker for heterodimeric receptors. IGF-I binding affinity is strongest for the IGF-1R, IGF-1R/IR-A and IGF-1R/IR-B heterodimers. IGF-II binding affinity is strongest for the IGF-1R, IR-A and IGF/IR-A heterodimeric receptor. Overarching biological effects for receptor activation via these pathways include the modulation of metabolism and mitogenesis. Figure adapted from [27,28,29,30]. Created with BioRender.com.
Ijms 26 04749 g001
Figure 3. Architecture and location of the choroid plexus (ChP) and SVZ. The cellular architecture of the lateral ventricle ChP is shown. The human SVZ is composed of four layers: (1) the ependymal cell layer, lining the lateral ventricle and separating the cerebral spinal fluid (CSF) from the SVZ; (2) the hypocellular layers composed of astrocytes and microglia; (3) the additional layer formed form three different types of astrocyte; (4) oligodendrocytes and myelinated axons. NSCs contact the CSF via apical cilia at the ventricular surface. The CSF and diffusible factors are released by the ChP which can intricately mediate NSC development. Adapted from [126,127]. Created with BioRender.com.
Figure 3. Architecture and location of the choroid plexus (ChP) and SVZ. The cellular architecture of the lateral ventricle ChP is shown. The human SVZ is composed of four layers: (1) the ependymal cell layer, lining the lateral ventricle and separating the cerebral spinal fluid (CSF) from the SVZ; (2) the hypocellular layers composed of astrocytes and microglia; (3) the additional layer formed form three different types of astrocyte; (4) oligodendrocytes and myelinated axons. NSCs contact the CSF via apical cilia at the ventricular surface. The CSF and diffusible factors are released by the ChP which can intricately mediate NSC development. Adapted from [126,127]. Created with BioRender.com.
Ijms 26 04749 g003
Figure 4. Schematic representing the findings in isocitrate dehydrogenase (IDH)-wildtype temporally located glioblastomas from Lee et al. (2018) [180]. Samples taken from SVZ free from tumours and the tumour itself were subject to whole-exome sequencing. Matching mutations between the two sites were found including single nucleotide variants in epidermal growth factor receptor (EGFR), tumour protein 53 (TP53) and telomerase reverse transcriptase (TERT). Insertions or deletions in phosphatase and tensin homolog (PTEN) and retinoblastoma protein (RB1) were also detected. The proportion of variant alleles or the variant allele frequency was much lower in SVZ samples than in tumour tissue (TERT C228T mutation SVZ: 1–22%, tumour: 22–52%). The figure is a visualisation of the results presented in [180]. Created with BioRender.com.
Figure 4. Schematic representing the findings in isocitrate dehydrogenase (IDH)-wildtype temporally located glioblastomas from Lee et al. (2018) [180]. Samples taken from SVZ free from tumours and the tumour itself were subject to whole-exome sequencing. Matching mutations between the two sites were found including single nucleotide variants in epidermal growth factor receptor (EGFR), tumour protein 53 (TP53) and telomerase reverse transcriptase (TERT). Insertions or deletions in phosphatase and tensin homolog (PTEN) and retinoblastoma protein (RB1) were also detected. The proportion of variant alleles or the variant allele frequency was much lower in SVZ samples than in tumour tissue (TERT C228T mutation SVZ: 1–22%, tumour: 22–52%). The figure is a visualisation of the results presented in [180]. Created with BioRender.com.
Ijms 26 04749 g004
Figure 5. Schematic showing disruption of the ependymal lateral ventricle wall by glioblastoma tumour cells. Reports by Norton et al. (2022) described the ability for patient-derived glioblastoma cells to contact CSF via invasion of the lateral ventricular lumen [186]. As a result, metabolic homeostasis is lost and ependymal lipid biosynthesis is disrupted [199,200]. Ependymal functional cilia for CSF circulation, neuroblast migration to the olfactory bulb and protection from fluid build-up (hydrocephalus) become shortened and dysfunctional [125,186,201,202,203]. It is proposed that CSF and protein contents could infiltrate through disruption of the ependymal monolayer into the tumour [186]. This is one proposed reason for lateral ventricle proximal glioblastomas exhibiting enhanced malignancy [186]. Adapted from [126,127,186]. Created with BioRender.com.
Figure 5. Schematic showing disruption of the ependymal lateral ventricle wall by glioblastoma tumour cells. Reports by Norton et al. (2022) described the ability for patient-derived glioblastoma cells to contact CSF via invasion of the lateral ventricular lumen [186]. As a result, metabolic homeostasis is lost and ependymal lipid biosynthesis is disrupted [199,200]. Ependymal functional cilia for CSF circulation, neuroblast migration to the olfactory bulb and protection from fluid build-up (hydrocephalus) become shortened and dysfunctional [125,186,201,202,203]. It is proposed that CSF and protein contents could infiltrate through disruption of the ependymal monolayer into the tumour [186]. This is one proposed reason for lateral ventricle proximal glioblastomas exhibiting enhanced malignancy [186]. Adapted from [126,127,186]. Created with BioRender.com.
Ijms 26 04749 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Harland, A.J.; Perks, C.M. IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis. Int. J. Mol. Sci. 2025, 26, 4749. https://doi.org/10.3390/ijms26104749

AMA Style

Harland AJ, Perks CM. IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis. International Journal of Molecular Sciences. 2025; 26(10):4749. https://doi.org/10.3390/ijms26104749

Chicago/Turabian Style

Harland, Abigail J., and Claire M. Perks. 2025. "IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis" International Journal of Molecular Sciences 26, no. 10: 4749. https://doi.org/10.3390/ijms26104749

APA Style

Harland, A. J., & Perks, C. M. (2025). IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis. International Journal of Molecular Sciences, 26(10), 4749. https://doi.org/10.3390/ijms26104749

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