Abstract
Glioblastoma multiforme (GBM) is the most common and deadly brain tumor, with a mean survival time of only 21 months. Despite the dramatic improvements in our understanding of GBM fueled by recent revolutions in molecular and systems biology, treatment advances for GBM have progressed inadequately slowly, which is due in part to the wide cellular and molecular heterogeneity both across tumors and within a single tumor. Thus, there is increasing clinical interest in targeting cell-extrinsic factors as way of slowing or halting the progression of GBM. These cell-extrinsic factors, collectively termed the microenvironment, include the extracellular matrix, blood vessels, stromal cells that surround tumor cells, and all associated soluble and scaffold-bound signals. In this review, we will first describe the regulation of GBM tumors by these microenvironmental factors. Next, we will discuss the various in vitro approaches that have been exploited to recapitulate and model the GBM tumor microenvironment in vitro. We conclude by identifying future challenges and opportunities in this field, including the development of microenvironmental platforms amenable to high-throughput discovery and screening. We anticipate that these ongoing efforts will prove to be valuable both as enabling tools for accelerating our understanding of microenvironmental regulation in GBM and as foundations for next-generation molecular screening platforms that may serve as a conceptual bridge between traditional reductionist systems and animal or clinical studies.
1. Introduction
Glioblastoma multiforme (GBM) is the most common and deadly form of primary brain cancer, accounting for approximately 54% of all brain tumors in the United States [1]. Despite its prevalence and lethality, there is currently no definitive treatment for patients afflicted with GBM. This lack of treatments is often attributed to the diffuse and unrelenting infiltration of tumor cells throughout the brain, [2] a phenomenon famously observed by neurosurgeon Dr. Walter Dandy in the 1920s, when he took the extreme step of surgically removing entire brain hemispheres of two comatose patients afflicted with GBM, only to see the tumor return post-resection [3].
While current treatment options are significantly more sophisticated than those exercised by Dr. Dandy, patient outcomes still remain poor. Standard therapy consists of the combination of tumor removal through surgical resection, radiotherapy, and chemotherapy. Following resection, image-guided radiotherapy is typically applied to the tumor margins, often including concomitant treatment with the alkylating agent temozolomide (TMZ) [4]. Despite this aggressive treatment regimen, tumor recurrence at the margin of the resection occurs in approximately 90% of patients and mean survival time is only around 21 months [4,5]. One of the main difficulties in effectively treating GBM with conventional therapies is that tumors that appear similarly in histopathological presentation are often in fact quite distinct at the cellular and molecular levels. For example, recent genomic analysis of many patient-derived GBM samples revealed at least three distinct subtypes of GBM, each of which contains specific genomic lesions relative to matched normal brain tissue (Figure 1) [6,7]. Furthermore, there is substantial cellular heterogeneity within a single tumor, with mounting evidence supporting the idea that tumor progression is driven by a subpopulation of glioma stem/initiating cells, which have high tumor-forming potential and express many neural stem cell markers [8]. Because cells in each tumor are distinct from other tumors classified as GBM, conventional treatments targeting intracellular signaling pathways, such as those regulating proliferation, will likely only be effective for a small subset of patients, and perhaps then only transiently as resistance evolves.
Figure 1.

Heterogeneity in GBM tumors. Hierarchical clustering of 200 tumors and 1740 genes revealed four distinct, statistically significant subtypes in GBM samples, which can be minimally represented by a predictive 840 gene sample (A). Red depicts genes that are overexpressed relative to normal tissue, while green depicts genes that are underexpressed. The four subtypes are named according to the lineage the tumor type most resembles. Performing the same analysis on either previously published data (B) or xenografts taken from mice (C) confirm the presence of four distinct subtypes. Figure adapted from Verhaak et al (2010), with permission.
Motivated by these findings, recent clinical trials have begun to explore new directions in the treatment of GBM with the aim of targeting the few common features shared across GBM subtypes. Instead of targeting cell-intrinsic pathways, these trials seek to intervene by manipulating the extracellular environment and the interactions of tumor cells with this environment, which is beginning to be recognized as a critical regulator of tumor progression [9–11]. Important components of the microenvironment include: 1) the extracellular matrix (ECM), the biopolymeric scaffold surrounding tumor cells, 2) non-tumor cells near or within the tumor, such as astrocytes, macrophages, endothelial cells, and fibroblasts, and 3) soluble and scaffold-bound signals such as growth and differentiation factors. Particularly intriguing is treatment with anti-angiogenesis drugs such as bevacizumab, which targets vascular-endothelial growth factor (VEGF), thereby reducing tumor-induced vascular recruitment. Bevacizumab has been shown to increase progression-free survival in phase III clinical trials when added to a regimen of radio- and chemo-therapy, but does not significantly improve overall survival [12–15]. In another novel modality of GBM treatment, directing cell migration towards an external chemotherapeutic sink with an implanted, migration-promoting hydrogel significantly reduced glioma tumor size in a mouse model [16]. The preliminary successes of these interventions hint at the promise of targeting the microenvironmental interactions of tumor cells as a viable treatment strategy in GBM. The identification of such targets for possible intervention therefore is a critical direction for GBM research, and this has motivated the development of advanced methodologies for studying interactions between GBM and the tumor microenvironment.
The extraordinary complexity of the tumor microenvironment has made it exceedingly difficult to determine how individual parameters in the microenvironment contribute to tumor progression in vivo. Because of this, there has been growing interest in developing simple, reductionist systems that model individual features of the microenvironment to isolate the effect of these features while offering a degree of reproducibility and interpretability not achievable with in vivo systems. This review will highlight the roles of the microenvironment on tumor development and progression, specifically focusing on the development of in vitro systems that mimic the tumor ECM. We will begin with a brief overview of how the microenvironment is thought to regulate GBM progression, then we will discuss in depth the experimental systems used to model this complexity in vitro. Finally, we will highlight existing technologies and model systems that may be adapted for the study of GBM.
2. Role of Microenvironment in GBM Progression
The microenvironment of GBM cells is extraordinarily complex and is composed of various types of ECM proteins and glycosaminoglycans (GAGs) as well as a diversity of cell types. This section provides a brief overview of the roles of the microenvironment in GBM, a subject that has been reviewed much more extensively elsewhere [17–19].
2.1 ECM Components of the Microenvironment
The ECM of the tumor microenvironment serves not only as a passive scaffold in which GBM tumor cells grow and migrate, but also plays a significant and active role in directing cellular behavior. The main component of brain ECM is the polysaccharide hyaluronic acid (HA) [20,21], which is a glycosaminoglycan made up of repeating disaccharides of D-glucuronic acid and N- acetyl-D-glucosamine. HA is constitutively produced within GBM tumors, [22,23] and its abundance is associated with cell proliferation and infiltration. Its cellular receptor CD44 is also overexpressed in GBM, and cells containing CD44 localize to the normal brain-tumor interface in vivo, suggesting that CD44-enriched cells are more efficient at invading the brain parenchyma [24,25]. Ligation of CD44 with HA activates key pro-tumorigenic signals including the Rho family of small GTPases [26,27], which are known to affect motility and proliferation, as well as PI3 kinase [28], which is known to affect cell growth, proliferation and differentiation. HA may also be endocytosed after CD44 binding, and its smaller degradation products have been shown to promote tumor progression through mechanisms that remain unclear [29].
The high vascularization of GBM also contributes ECM cues to GBM cells. For example, the basolateral membrane of GBM-associated vessels contains collagen IV, collagen V, fibronectin, and laminin, all of which are comparatively less abundant in avascular regions of the brain [30–33]. These molecules have been found repeatedly to enhance cell survival, proliferation, and migration in vitro and in vivo [34–38]. Furthermore, the proteoglycan tenascin-C is produced by tumor-associated endothelial cells and its presence correlates with angiognenesis and the progression from grade II to grade III glioma [39,40].
Attachment to many types of ECM, including fibronectin, laminin, and collagen, is controlled by a class of membrane-spanning proteins known as integrins. Interestingly, adhesion of integrins to their extracellular ligands may significantly desensitize GBM cells to therapy [41]. For example, α1 integrin signaling has been negatively correlated with drug induced apoptosis in GBM [42]. Additionally, α6 integrin is necessary for GBM stem cell self-renewal, proliferation, and tumor-forming capacity [37,43].
In addition to responding to existing ECM-based cues, GBM tumor cells also have the ability to actively shape the ECM for optimal cell growth and infiltration. Specifically, tumor cells may remodel the ECM through cell-secreted proteases, which “prime” the microenvironment for tumor progression by removing steric barriers for cell migration and angiogenesis, leading to enhanced GBM growth and dissemination. Urokinase plasminogen activator (uPA), cathespin B, and matrix metalloproteinases (MMP) have been shown to be upregulated in GBM and high levels of these proteases correlate with poor prognosis in patients [44,45].
2.2 The Mechanical Properties of the Extracellular Matrix and their Role in Tumor Progression
The mechanical properties of the ECM have been increasingly recognized as critical to the progression of advanced tumors in vivo. Cells interact with their mechanical environment, and these interactions have been shown to influence such tumor-critical processes as migration, differentiation, apoptosis, and proliferation.
Cells mechanically engage their environment and respond to microenvironmental forces using several distinct mechanisms whose actions are collectively termed mechanosensing or mechanotransduction [46]. In order to probe the local mechanical properties of its environment, a cell must deform it by actively applying a force. In one common paradigm, cells apply forces to the extracellular environment through the structural positioning [47] and contraction of actin-myosin complexes [48], with the resulting force being transmitted along actin filaments to membrane-spanning focal adhesion complexes that are physically connected to the extracellular environment through integrin-based complexes [49,50]. Focal adhesions then respond to the stress and/or strain across the adhesion, which initiates a signaling cascade with downstream targets such as changes in actin polymerization and crosslinking [51], changes in focal adhesion size [52], and regulation of gene transcription [53].
Numerous clinical observations support the hypothesis that tissue stiffness is critical for the progression of a variety of solid tumors, especially breast tumors [11]. GBM tumors are also thought to be stiffer than the normal brain tissue surrounding the tumor, as suggested by ultrasound imaging of strain magnitudes during neurosurgery [54]. Although the detailed spatial variation of stiffness in GBM tumor tissue and the contributing mechanisms are yet to be described, there is a strong possibility that increased tumor stiffness may contribute to malignancy as has been explored extensively in epithelial cancers [55,56]. Furthermore, observations by Hans Scherer in the 1940s identified key tracks for GBM invasion into the brain, such as the vasculature beds and white matter tracts [57,58]. Now known as Structures of Scherer, these components of the brain are known to be significantly stiffer than the surrounding parenchyma, suggesting GBM guidance may be influenced by the mechanical properties of the Structures of Scherer.
Equally important to these clinical observations is the fact that GBM cells often acquire abnormalities in the mechanosensory machinery, including aberrant expression of key molecular components. For example, the critical mechanosensory protein focal adhesion kinase (FAK) is overexpressed in many GBM tumors. [59]. Additionally, GBM often exhibits altered expression of integrins, which, as discussed above, are critical for the physical transmission of force to the extracellular matrix from the actin cytoskeleton, in addition to simply mediating attachment to the ECM [60–62]. Finally, the leaky vasculature of GBM tumors contributes to peritumoral edema and increased interstitial fluid pressure (IFP) [63,64], which routinely causes morbidity and hinders drug delivery to the tumor, but can also alter the mechanical microenvironment through modulation of cell-ECM tension in both tumor and stromal cells [65]. All of these observations imply that an intricate link exists between the mechanical environment of the brain and GBM progression, although the precise molecular details remain incompletely understood.
2.3 GBM Interactions with Cells
GBM tumors are often composed of not only tumor cells but also parenchymal cells, which contribute to tumor progression and may in fact contribute more than 30% of the mass of a tumor [17]. This section will provide an overview of the types of interactions observed between GBM cells and parenchymal cells.
2.3.1 Perivascular Niche Cells
There is increasing evidence that the vasculature of GBM serves not only to provide an exchange medium for nutrients and waste products, but also that anatomical structures formed by tumor-associated vessels provide a pro-survival environment for GBM cells. These structures, known as perivascular niches, are composed of endothelial cells, pericytes, and astrocytes [17]. GBM vasculature is different than non-diseased vasculature throughout the brain and is often characterized by endothelial cell proliferation and hyperplasia [66]. Recruitment of vascular smooth muscle cells and pericytes is critical for forming a perivascular niche and for the survival of tumor-associated endothelial cells [67] and have been implicated in the progression of malignant gliomas [68].
Perivascular niche-associated cells, particularly astrocytes, deploy autocrine signaling to promote tumor malignancy and survival. GBM cells residing in the perivascular niche are often associated with reactive astrocytes which may induce GBM growth through the secretion of stromal cell-derived factor 1 (SDF-1) [69] and astrocyte elevated gene-1 (AEG-1) [70], both of which are frequently overexpressed in human brain tumors [71,72]. Interestingly, suppression of AEG-1 activity in a GBM mouse model diminished tumor growth, highlighting the importance of the perivascular niche to GBM progression [71]. Additional chemokines may serve as malignancy-promoting signals transduced by receptors that have been found on glioma cells including, chemoattractant protein-1 (CCL2), interleukin-8 (CXCL8), and RANTES (CCL5) [73]. Conversely, gliomas are also known to influence the proliferation, motility, and secretion of MMPs from astrocytes through soluble signaling cascades, suggesting that gliomas may leverage astrocytes to enhance their invasion [74–76].
The perivascular niche may also be important for the maintenance of brain tumor stem cells [77]. Because neural progenitor cells also associate with perivascular niches [78,79], these environments have been hypothesized to incubate GBM tumor-initiating cells and/or induce de-differentiation of tumor cells to a tumor-initiator like state [80,81]. Tumor cells that express stem cell markers CD133 and nestin have been shown to preferentially associate with the vasculature [80,82] and, remarkably, preferentially proliferate at vascular branch points [83]. Paracrine nitric oxide signaling has been suggested as a potential mediator of tumor stem cell maintenance, working through the Notch signaling axis to enhance self-renewal [84]. Similarly, interleukin-8 secretion by endothelial cells has been shown to enhance the proliferation and migration of GBM tumor stem cells [85]. Intriguingly, this effect was enhanced when the endothelial cells were cultured as three-dimensional networks compared to conventional monolayer culture, underscoring the importance of microenvironment dimensionality and mechanics in the malignant behavior of tumor-associated stromal cells. The field’s understanding of the cross-talk between tumor-associated endothelia and tumor stem cells is complicated by recent studies that suggest GBM stem cells may themselves transdifferentiate to become endothelial cells and incorporate into the tumor vasculature [86–88].
2.3.2 Microglia
The majority of non-tumor cells within a tumor are tumor-associated macrophages, which are most often differentiated macrophages known as microglia and have suppressed immune functions [89,90]. In fact, microglia may modulate the growth and migration of glioma cells [91,92]. Microglia have been found to produce MMPs, including MT-MMP1, in response to soluble factors secreted by glioma cells, likely through the activation of toll-like receptors on the microglia surface which further activates the MAPK pathway [92]. Additionally, in glioma mouse models that exhibit impaired microglia or lack microglia altogether, glioma tumor size was significantly less than those with normal microglia [93].
3. Engineering strategies to model the GBM microenvironment
As discussed above, the microenvironment of brain tumors plays a very significant role in GBM development, progression, and treatment. In this section, we will discuss strategies that have been adopted to recapitulate some of the key features of the GBM microenvironment in vitro. These strategies borrow technologies from surface science, polymeric materials, and microfabrication to manipulate the architectural and molecular features of biomaterial scaffolds and present cells with highly sophisticated and controlled microenvironmental cues.
3.1. Limitations of traditional cell culture systems
The role of ECM components in tumor pathophysiology has traditionally been studied using two-dimensional (2D) monolayer cell culture on glass or plastic substrates. This involves functionalizing the surface with the protein or proteoglycan of interest, typically by adsorbing it from solution, or by adding the molecule to the cell culture medium to observe effects of soluble signaling mediated by the ECM component. This simple method has enabled the study of tumor cell behavior in culture and has yielded a wealth of information on the role of several ECM molecules in tumor initiation and progression, often laying the groundwork for further studies using more physiologically appropriate models. For example, Berens and co-workers adopted this technique in a series of pivotal studies delineating the role of ECM components such as fibronectin, vitronectin, and hyaluronic acid in promoting the adhesion and migration of astrocytoma cells [94,95]. Similarly, the role of the matrix metalloproteases (MMPs) in glioma invasion was first established using monolayer culture systems [96]. Despite their utility in basic functional assays, simple monolayer culture suffers from several significant drawbacks when employed to study complex cell-ECM interactions. First, it is now well understood that cells in three-dimensional (3D) culture behave very differently than on flat 2D substrates: essential differences in the presentation, organization, and polarity of ECM proteins in 3D matrix result in concomitant modifications in the architecture and composition of cell-ECM adhesions and downstream signaling events [97,98]. Second, the stiffness of typical plastic or glass substrates exceeds 3 GPa, which is many orders of magnitude stiffer than brain, a tissue whose stiffness typically ranges from 100–5000 Pa [55]. Third, essential features of the 3D tumor microenvironment including hypoxia [99], increased interstitial fluid pressure [100], and cytokine concentration gradients are poorly reproduced by 2D culture. Finally, flat monolayer culture by its very nature is not naturally suited to studying the effects of matrix remodeling or cell-cell interactions. To overcome these obstacles, the field has turned to engineered microenvironments that can offer exquisite control over material composition, stiffness, and architecture in both 2D and 3D culture to systematically study the effects of complex cell-ECM interactions on GBM tumor pathology.
3.2. Studying the role of ECM stiffness
As discussed earlier, a wealth of indirect evidence based on clinical and empirical observations led to a suspected role for cell-ECM biophysical interactions in the pathophysiology of glioma. However, systematic mechanistic studies in this area have only become possible due to the development of cell culture substrata with independently controllable elastic modulus and biochemical functionalization. This is typically achieved by controlling the elastic modulus via the degree of crosslinking of a polymeric hydrogel matrix, which is then chemically functionalized – either by covalent attachment or adsorption from solution – by the ECM-derived protein or peptide ligand of interest to facilitate glioma cell attachment. The most commonly used such system is based on crosslinked polyacrylamide (PAA) and was originally introduced by Wang and co-workers in seminal studies that established the role of ECM stiffness on fibroblast morphology and motility [52,101]. This platform has since become widely adopted and has motivated the creation of multiple step-by-step protocols for generating PAA gels of tunable elasticity and ligand density [102–105]. Using these tunable PAA gels functionalized with fibronectin, our group demonstrated for the first time that the morphology, cytoskeletal architecture, proliferation, and motility of human GBM cells were exquisitely sensitive to the stiffness of the underlying substrate [106]. Cells were rounded and largely immobile on soft substrates, whereas increasing ECM stiffness resulted in a dramatic increase in cell area, focal adhesion formation, proliferation, and motility. Further, we found that this mechanosensitivity was dependent on non-muscle myosin II-mediated cellular contractility, such that inhibition of contractility using the small-molecule drug blebbistatin rescued invasion on soft substrates. In follow-up studies, we have explored the role of the focal adhesion proteins α-actinin [107] and talin [108] in transducing mechanical cues, as well as the central role of the Rho GTPase RhoA in mediating the resulting contractility-dependent cellular phenotype [109]. As a potential caveat, a recent study suggested that changing the stiffness of the PAA substrate is potentially communicated to cells through alterations in the anchoring density of the tethered ECM protein [110]. However, stiffness-dependent phenoptypes observed on PAA can often be reproduced with other, unrelated materials, including hyaluronic acid [111] and PDMS-based micropost arrays [112], suggesting that these PAA-based behaviors may be correctly interpreted as mechanosensory events. In any case, these tunable-stiffness culture systems can be used to delineate the molecular mechanisms by which ECM-encoded cues are sensed and processed by glioma cells and therefore may help identify molecular targets against tumor progression. For instance, through combined studies of glioma invasion in brain slice cultures [113] and mouse models [114], Rosenfeld, Canoll, and colleagues have shown that pharmacologic inhibition of myosin can severely limit tumor invasion even in the presence of potent pro-motility cytokines.
3.3. Modeling the glioma microenvironment in 3D ECMs
There is an increasing impetus in the field of tumor biology to study cell behavior in fully three-dimensional matrices that are significantly better at recapitulating physiological features than 2D monolayer culture. In addition to the fact that cells in 3D ECMs adopt shapes and adhesions that are much closer to those in vivo [115], 3D culture also facilitates the investigation of processes such as matrix remodeling and proteolytic degradation, which are central to tumor invasion [116]. Consequently, there have been strong efforts to study glioma cell-ECM interactions in 3D matrices to elucidate the role of matrix density, stiffness, and architecture in glioma progression.
3.3.1 Self-assembled biopolymer gels
Much early effort to investigate glioma invasion in 3D matrices emphasized hydrogels composed of native biopolymers such as collagen I, or Matrigel, a laminin-rich ECM extract from mouse sarcoma tumors. For instance, initial studies of U87-MG glioma spheroid expansion in Matrigel indicated that the growing spheroid exerted compressive forces whereas invading cells exerted traction forces on the ECM, showcasing the varied nature of physical interactions involved [117]. To further analyze how glioma cells interact with the ECM, Kaufman and colleagues studied glioma spheroid invasion in collagen I gels of varying concentration [35]. Although increasing collagen I concentration facilitated spheroid invasion due to an increase in fibers available for cell-ECM adhesions and traction, spheroid growth was inhibited at higher collagen I concentrations, perhaps because of the inhibitory effect of growing against a dense matrix. This latter observation was broadly confirmed with a wider panel of glioma cell types, where the cellular levels of cadherin and matrix metalloprotease (MMP) expression were also found to impact the invasive pattern[118]. Indeed, degradation of the collagen matrix by MMP activation – for example, upon Epidermal Growth Factor (EGF) stimulation - is one mechanism by which glioma cells can overcome the inhibitory effect of a denser ECM on cell migration [119]. A recent study employed temperature-controlled nucleation of collagen I fibers to partly deconvolute collagen concentration and matrix pore size. Collagen matrices nucleated at 22 °C as opposed to 37 °C exhibited a more porous network architecture, and permitted greater glioma spheroid invasion upon increasing collagen concentration, thus establishing the dominant role of matrix porosity in glioma invasion [120].
3.3.2 Synthetic ECMs
Although the studies described above using biopolymer gels have yielded a great deal of insight into the regulation of glioma by ECM, their interpretation and applicability to the physiological situation are complicated by two factors. One, the normal brain ECM, as discussed above, is almost entirely devoid of fibrillar collagens, being instead composed of a dense non-fibrillar matrix based on a HA-proteoglycan-tenascin network [121]. Further, as discussed above, changing the concentration of the gel-forming biopolymers simultaneously affects ECM ligand density, network architecture, and stiffness, making it difficult to assess the independent contributions of these parameters in regulating glioma invasion. These drawbacks of native biopolymer gels can be partly overcome by synthetic ECM platforms based on crosslinked natural or synthetic polymers, where ECM stiffness and biochemical ligand functionalization can be adjusted independently. Over the last few decades, a number of ECM platforms have been developed to address these needs (as previously reviewed [122] and [123]), some of which are now commercially available. Here we restrict our focus to the application of these biomaterials for studying glioma progression in 3D.
Our laboratory adopted an approach for decoupling ECM stiffness from protein concentration in collagen I hydrogels by mixing the protein with agarose, an inert biopolymer that forms a meshwork entangled with the collagen and serves to stiffen the hydrogels with modest alterations in the fiber architecture [124]. Using this platform, we found that increasing matrix stiffness in 3D inhibited and eventually abrogated glioma spheroid invasion – a result opposite to that observed on 2D substrates [106]. This apparent contradiction is explained by the fact that migration in 3D collagen matrices requires cellular traction-mediated remodeling of collagen fibers while navigating steric barriers within the matrix, which are both impeded by hydrogel stiffening due to added agarose [125]. Further, single-cell migration in denser, agarose-rich matrices showed a phenotype reminiscent of amoeboid motility, with cells exhibiting dynamic path-finding protrusions at the leading edge and constrictions in the cell body to pass through narrow spaces in the ECM. Amoeboid motility has previously been observed in cells lacking protease activity [126,127] and is thought to be a mechanism by which tumor cells can escape therapeutic interventions that inhibit MMPs [128]. To further elucidate the role of matrix density in governing the mode of 3D glioma cell motility, we sought to create synthetic ECMs that more closely mimicked the architecture of native brain tissue. To this end, we adopted an ECM platform consisting of methacrylated HA functionalized with ECM-derived peptide ligands, such as the cell-adhesive RGD sequence, and crosslinked by dithiols such as dithiothreitol (DTT), enabling independent control of cell adhesivity and stiffness over ~3 orders of magnitude [111,129]. Importantly, these HA ECMs were devoid of fibrillar structures and had a high density with submicron porosity, similar to brain matrix [121]. U373-MG glioma spheroid invasion in HA ECMs was again inhibited by increasing ECM stiffness, as observed before; further, cells invading in the dense HA ECMs displayed a phenotype unlike that observed in fibrillar collagen matrices but strikingly similar to that previously observed in brain slice cultures [113] (Figure 2). This result raises the exciting possibility that HA-based synthetic ECMs can be used to dissect the signaling mechanisms that underlie the aggressive invasiveness of glioma cells, eventually leading to new molecular targets. These HA-based ECMs could also serve as a reductionist platform for studying the mechanisms by which the HA receptor CD44, which as described earlier activates intracellular signals that increase glioma cell proliferation and invasion [130,131].
Figure 2.

GBM cell motility in dense ECMs. U373-MG cells migrating in dense 3D hyaluronic acid-based hydrogels adopted a phenotype displaying dynamic, branched leading protrusions with abrupt cell-body movements, distinct from mesenchymal motility in 3D Collagen gels but strikingly similar to the motility observed in brain tissue. Figure adapted from Ananthanarayanan et al (2011), with permission.
3.4. Microfabricated platforms for studying cell-ECM interactions
An emerging trend in studies of tumor physiology in vitro is the use of microfabricated platforms that offer exquisite control over the topography, roughness, elasticity, and biochemical functionalization of the cell culture interface. Although these systems may not have the full complement of biochemical information encoded in native biopolymer gels or enable fully 3D studies of tumor invasion as in synthetic hydrogels, their specific advantage is in presenting a complex set of physico-chemical cues with control over spatial presentation at length scales relevant to cellular interactions, i.e. 10 nm – 10 μm. (as previously reviewed [132], [133], [134]). Here we shall focus on the application of these microfabricated systems to elucidate key aspects of glioma pathophysiology.
Zhu et al. used direct laser irradiation to create periodic grooves spaced ~ 200 nm apart on a polystyrene film and reported robust alignment of C6 rat glioma cells along the groove orientation [135]. This phenomenon, known broadly as contact guidance, has been reported widely for many neural cell types in culture and verifies the sensitivity of cell shape and cytoskeletal structure to physical constrains imposed upon it by the topography of the environment. A recent study used soft lithography molding of poly(dimethylsiloxane) (PDMS) to create lines and grooves and showed that primary glioma tumor cells showed directionally persistent migration along the lines on the surface [136]. To directly investigate the interplay between topographical confinement as experienced by a cell migrating in a dense 3D environment, and the stiffness of the matrix – which are intricately coupled in 3D culture platforms – our laboratory studied glioma cell migration in microfabricated polyacrylamide channels with independently tunable channel width and wall stiffness [137]. We found that confinement in narrow channels (< 10 μm width) increased glioma cell migration speed across all ECM stiffnesses, relieving the inhibitory effect of high ECM stiffness on 2D unconfined migration (Figure 3). Further, this confinement sensitivity was mediated by myosin II-mediated polarization of cytoskeletal traction forces, consistent with an established role for myosin II in enabling glioma cell migration in the dense brain environment [113]. We later used this platform to explore the interplay of these microenvironmental parameters with the expression of canonical oncogenes [138]. Another recent study from a different set of investigators used microcontact printing of adhesive fibronectin islands at different densities to simulate the physical compaction that is experienced by a growing tumor in vivo [139]. This study found that compaction increased expression of collagens IV and VI as well as the collagen crosslinking enzyme lysyl oxidase, which led to increased VEGF-mediated angiogenesis. Thus, microfabricated ECMs can be used to simulate several key features of the glioma microenvironment and uncover novel mechanisms that contribute to glioma progression.
Figure 3.

Orthogonal regulation of GBM cell motility by ECM stiffness and topographical confinement. Migration of U373-MG cells was studied in microfabricated fibronectin-coated polyacrylamide substrates with independently varying channel widths cw and wall stiffness E. Confinement in channels of narrow width increased migration speed for all values of wall stiffness (A) and relieved the inhibitory effect of high stiffness (B). Phase contrast images of the migrating cells shown in (C). Figure adapted from Pathak et al (2012), with permission
4. Future directions in modeling GBM in vitro
One of the outstanding challenges in cancer drug discovery is to improve the success rate of potential anticancer agents that succeed in clinical trials, which is currently around 5% [140]. This low success rate is partly due to the poor predictive ability of preclinical models of human cancers, such as mouse xenograft models [141], which in turn is due to their inability to faithfully reproduce non-cell-autonomous contributions to tumor pathology. The field of ‘tumor engineering’ seeks to bridge the gap by building sophisticated models of human cancer in vitro by leveraging advances in tissue engineering and biomaterials to recapitulate the tumor microenvironment with greater fidelity while retaining the essential genetic and epigenetic background through the use of appropriately sourced human cells [142–144]. In the next section, we will discuss the ways in which this strategy may aid in modeling important features of GBM in vitro.
4.1. Modeling glioma motility in 3D
Because GBM aggressively invades brain tissue, effective inhibition of tumor cell migration is widely considered an important therapeutic objective [145,146]. Interestingly, glioma cells invading the dense brain parenchyma have been observed to exhibit a distinctive type of motility that features highly dynamic protrusions and saltatory forward movements and requires myosin II-mediated contractility to squeeze the cell-body through narrow spaces [113]. However, glioma cells undergoing perivascular migration along blood vessels exhibit conventional mesenchymal motility with an elongated cellular phenotype [147]. These observations reinforce the fact that many tumor cells display significant plasticity in their modes of motility, implying a redundancy in the underlying mechanisms that allows cells to ‘tune’ their motility apparatus to effectively migrate in a variety of environments [148,149]. It is apparent that the diversity in cellular motility modes is in part reflective of the varied microenvironmental barriers cells must overcome to migrate in 3D [150]; however, we still have a very limited understanding of how these extracellular inputs are processed and integrated to enable the proficient invasion that is the hallmark of glioma. Our work using collagen-agarose ECMs and HA-based hydrogels indicated that increasing density of the 3D matrix causes a transition from an elongated mesenchymal mode to a contractility-dominated amoeboid-like phenotype [111,124]; however, the molecular mechanisms underlying this switch are only beginning to be elucidated. Several studies have indicated a role for the Rho GTPases Rac1, RhoA, and proteins that regulate the balance of their activity in governing different modes of glioma migration [147,151–153]. It is also becoming evident that the size and deformability of the nucleus often represent the rate-limiting step in migration through dense environments such as brain tissue [113,154]. Future work in this area using the appropriate 3D ECM models is very much needed for obtaining a coherent overall picture of the regulation of glioma migration and how it may be effectively inhibited in prospective therapies.
4.2 Modeling Interactions between GBM Tumor Cells and Other Cells
4.2.1 Myelinated Axons
Conventional substrates for the study of GBM migration are problematic in that they do not adequately represent the migration along the Secondary Structures of Scherer, which are perhaps the most common routes of dissemination for GBM in vivo. Myelinated axons, the main component of white matter tracts [155], have significant research value to many other research fields, such as the study of multiple sclerosis [156]. As such, significant effort has been made to create in vitro cultures that produce myelinated axons, primarily by the co-culture of immature oligodendrocytes with neurons [157]. Unfortunately, these myelinated axons have yet to be significantly incorporated into in vitro models of GBM, perhaps due to the significant technical burden associated with isolating, culturing, and patterning these cells. To achieve simpler and more reproducible systems, researchers have begun adapting approaches from materials science to mimic the aligned and discontinuous nature of the Structures of Scherer.
Electrospinning is a simple and effective method that applies high electrical potentials to polymer solution droplets to produce thin polymer fibers that can range from around 2 nm to hundreds of microns in diameter [158]. Electrospun fibers can be aligned as they are collected and functionalized to permit cell adhesion. Electrospun fibers have been widely exploited as tissue engineering scaffolds, specifically in the subfield of neural regeneration, and have been used effectively as substrates to investigate and promote neural stem cell differentiation [159], Schwann cell maturation [160], and as guides for neural migration following repair from injury [161,162]. Recently, these substrates have been adopted for the study of GBM[163,164]. For example, poly-ε-caprolactone (PCL) electrospun fibers have been shown to recapitulate important features of GBM migration observed in vitro. Using a core-spinning technique to independently vary the physical and chemical properties of PCL fibers, Rao et al. found that cell morphology, FAK expression, and myosin light chain-2 expression all strongly depend on fiber modulus [164]. Future effort will be needed to improve the degree to which these systems mimic tissue architecture, particularly with respect to incorporation of physiologically relevant ECM ligands and development of three-dimensional topologies.
4.2.2 Endothelial Cells
There has been substantial effort to understand the coordinated chemical signaling between blood vessels and glioma cells. Most in vitro efforts to model this have used simple co-culture models in which endothelial cells are cultured on the same surface as glioma cells [165–167]. These systems have revealed that endothelial and glioma cells can communicate through soluble paracrine signals and also physical signals when the cells are in direct contact. While these methods are useful for studying biochemical signaling between the vasculature and tumor, much work remains to be done to improve the degree to which these systems capture the cellular organization and architecture of the vascular-tumor cell interface.
GBM researchers may benefit from leveraging the methods developed by tissue engineers to produce realistic 3D vascular structures. Endothelial cells cultured on carrier beads and embedded in 3D hydrogels will spontaneously grow to form preliminary vessels [168]. Furthermore, spatially patterning PEG hydrogels with light to contain localized zones of bound integrin-adhesive RGD peptide or VEGF can guide the formation of blood vessels in vitro [169]. More sophisticated approaches have recently been developed that use a sacrificial 3D printed sugar-glass layer to form the structure of the endothelial cell network or microfluidics molds to produce realistic blood vessels [170–172]. Perhaps adopting these approaches to form model vasculature, in combination with glioma cells, will lead to further insights into the mechanism of intravasation of GBM cells and the perivascular niche regulation of tumor initiating cells, as discussed previously [85].
4.2.3 Microglia, Astrocytes, and Tumor-Associated Fibroblasts
Microglia, astrocytes, and tumor-associated fibroblasts profoundly affect tumor formation in vivo, as discussed earlier [91–93]. While simple co-culture studies have revealed much about the nature of the soluble paracrine signaling between these cell types [173], it is likely that they may also physically communicate. As such, GBM researchers may benefit from the ability to control seeding density and cell-cell contact by using spatially controlled coculture systems developed for other cell types. There are many systems that have been developed to spatially localize multiple cell types in vitro [174], including simple selective patterning, most often using microcontact printing of multiple cell-adherent proteins, where one cells can bind one, but not both, of the patterned ligands [175]. Microfluidic platforms have also been designed to control spatial localization of cells. [176] Additionally, systems have been developed in which adhesion can be controlled both temporally and spatially, with heat or electrical charge, which allow seeding of one cell type followed by the exposure of more ligand that allows the second cell type to adhere in a spatially controlled manner [177,178].
Spatially controlled co-culture is also possible in three dimensions. When dispersed in mineral oil, small, cell-laden hydrogel building blocks will assemble into complex and controlled structures which may then be crosslinked into a continuous gel using UV light [179]. Alternatively, 3D tissues can be assembled by the sequential layering of detached cell monolayers [180]. Spatially controlled 3D cultures may also be formed through microfluidics-based approaches, where different solutions are slowly flowed through microchannels to spatially localize individual components [181]. The adoption by cancer biologists of these complex and controlled systems will likely aid in understanding the complex interactions between tumor and stromal cells.
4.3 High Throughput approaches to ECM Screening
As discussed previously, microenvironmental regulation of GBM is complex and multi-factorial. Because of this, conventional low-throughput discovery platforms will likely not be able to fully uncover the specifics of the ECM-regulation of GBM cells. Furthermore, since chemoresistance is influenced by integrin adhesion to the ECM, multiplexed arrays with well-controlled cell-ECM interactions may facilitate both our fundamental understanding of how this adhesion drives biology and screening of chemotherapeutic drugs in a more physiologically-mimetic context than conventional screening assays.
Robotic spotting of ECM proteins has proven to be an extremely useful, versatile, and high-throughput means to test the effects of ECM on cells [182]. By combinatorially and sequentially varying the amounts of collagens I, III, IV, laminin and fibronectin, Alberti et al. found that specific combinations of these molecules controlled embryonic stem cell fate in ways that would have been nearly impossible to predict using traditional, lower-throughput paradigms [183]. Using the same approach, they were then able to devolve combinatorial effects of growth factor signaling and ECM signaling. Similarly, one can vary the mechanical properties of hydrogels using a robotic spotter and UV light to initiate photopolymerization, with one study screening 17,000 distinct combinations of stiffness and ligand in one experiment (Figure 4) [184,185] These high-throughput platforms need to be leveraged for constructing more sophisticated and physiologically relevant in vitro models of GBM that can be used for drug discovery screens.
Figure 4.

High throughput combinatorial screening of matrix configurations for human embryonic stem cell culture. Robotic spotting technologies were used to create highly precise arrays of 576 unique configurations of polymer gels (A, B). The arrays were then seeded with human embryonic stem cells and assayed for cell growth by quantifying the percent area of the island occupied (C). The high degree of multiplexing allowed for the identification of specific conditions that encourage embryonic stem cell growth in a manner that would have been largely impossible using conventional methods. This technology could be adapted to study the combinatorial effects of different ligand combinations on GBM behavior. Figure adapted from Anderson et al (2004), with permission.
5. Conclusions
Despite recent revolutions in molecular and systems biology that have facilitated our fundamental understanding of cell-intrinsic regulation of GBM, the prognosis for this disease remains quite poor, suggesting that cell-extrinsic or microenvironmental interactions may also substantially contribute to disease progression. Extensive research has revealed a significant role for the microenvironment - including adjacent cells, the ECM, and the vasculature - in maintenance of GBM. The extreme complexity and potential importance of the microenvironment in vivo has motivated enormous effort to develop next-generation platforms for capturing key components of the GBM tumor microenvironment in vitro, which is in turn allowing unprecedented dissection of microenvironmental regulation of GBM.
While significant groundwork has now been laid in this direction, significant challenges remain. First, the extreme complexity of the in vivo environment remains to be modeled in a reproducible and highly controlled fashion. By integrating multiple reductionist approaches systematically and deploying them in a high-throughput fashion, researchers should be able to create more complete, reproducible, and controllable in vitro GBM microenvironments. In addition to the complexity of the microenvironment, significant knowledge gaps remain in our understanding of tumor complexity and heterogeneity, both within a single tumor (as exemplified in the increasingly recognized role of tumor stem cells) and across different GBM tumors (as embodied by the four distinct GBM subtypes). It is possible, perhaps even likely, that different tumor cell types will respond differently to the same constellation of microenvironmental cues. Finally, it will be important for investigators to leverage these increasingly complex in vitro systems for the high-throughput drug discovery and screening, which we envision will help bridge the gap often found between highly reductionist systems, animal studies, and clinical trials.
Acknowledgments
B.A. gratefully acknowledges the support of a postdoctoral fellowship from the California Institute for Regenerative Medicine (TG2-01164). A.R. gratefully acknowledges the support of an NIH NRSA postdoctoral fellowship (1F32CA174361-01). S.K. gratefully acknowledges support from the NIH (1R211EB016359), NSF (CMMI 1105539), and W. M. Keck Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Agnihotri S, Burrell KE, Wolf A, Jalali S, Hawkins C, et al. Glioblastoma, a Brief Review of History, Molecular Genetics, Animal Models and Novel Therapeutic Strategies. Arch Immunol Ther Exp. 2013;61:25–41. doi: 10.1007/s00005-012-0203-0. [DOI] [PubMed] [Google Scholar]
- 2.Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, et al. The 2007 WHO Classification of Tumours of the Central Nervous System. Acta Neuropathol. 2007;114:97–109. doi: 10.1007/s00401-007-0243-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.DANDY WE. Removal of right cerebral hemisphere for certain tumors with hemiplegia: Preliminary report. JAMA. 1928;90:823–825. doi: 10.1001/jama.1928.02690380007003. [DOI] [Google Scholar]
- 4.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. New England Journal of Medicine. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- 5.Berens ME, Giese A. “…those left behind.” Biology and Oncology of Invasive Glioma Cells. Neoplasia. 1999;1:208–219. doi: 10.1038/sj.neo.7900034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McLendon R, Friedman A, Bigner D, Meir EGV, Brat DJ, et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110. doi: 10.1016/j.ccr.2009.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Altaner C. Glioblastoma and stem cells. Neoplasma. 2008;55:369–374. [PubMed] [Google Scholar]
- 9.Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007;7:139–147. doi: 10.1038/nrc2067. [DOI] [PubMed] [Google Scholar]
- 10.Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239–252. doi: 10.1038/nrc2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8:241–254. doi: 10.1016/j.ccr.2005.08.010. [DOI] [PubMed] [Google Scholar]
- 12.Chinot OL, de Rouge TLM, Moore N, Zeaiter A, Das A, et al. AVAglio: Phase 3 trial of bevacizumab plus temozolomide and radiotherapy in newly diagnosed glioblastoma multiforme. Adv Therapy. 2011;28:334–340. doi: 10.1007/s12325-011-0007-3. [DOI] [PubMed] [Google Scholar]
- 13.Kreisl TN, Kim L, Moore K, Duic P, Royce C, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27:740–745. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Narayana A, Gruber D, Kunnakkat S, Golfinos JG, Parker E, et al. A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma: Clinical article. Journal of Neurosurgery. 2012;116:341–345. doi: 10.3171/2011.9.JNS11656. [DOI] [PubMed] [Google Scholar]
- 15.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Dowell JM, Reardon DA, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253–1259. doi: 10.1158/1078-0432.CCR-06-2309. [DOI] [PubMed] [Google Scholar]
- 16.Jain A, Betancur M, Patel GD, Valmikinathan CM, Mukhatyar VJ, et al. Guiding intracortical brain tumour cells to an extracortical cytotoxic hydrogel using aligned polymeric nanofibres. Nat Mater. 2014;13:308–316. doi: 10.1038/nmat3878. [DOI] [PubMed] [Google Scholar]
- 17.Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia. 2011;59:1169–1180. doi: 10.1002/glia.21136. [DOI] [PubMed] [Google Scholar]
- 18.Payne LS, Huang P. The pathobiology of collagens in glioma. [Accessed 14 August 2013];Mol Cancer Res. 2013 doi: 10.1158/1541-7786.MCR-13-0236. Available: http://mcr.aacrjournals.org/content/early/2013/07/16/1541-7786.MCR-13-0236. [DOI] [PMC free article] [PubMed]
- 19.Ulrich T, Kumar S. Mechanobiology in Health and Disease in the Central Nervous System. In: Nagatomi J, editor. Mechanobiology Handbook. CRC Press; 2011. [Accessed 14 August 2013]. pp. 391–411. Available: http://www.crcnetbase.com/doi/abs/10.1201/b10780-23. [Google Scholar]
- 20.Laurent TC, Fraser JR. Hyaluronan. FASEB J. 1992;6:2397–2404. [PubMed] [Google Scholar]
- 21.Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer. 2004;4:528–539. doi: 10.1038/nrc1391. [DOI] [PubMed] [Google Scholar]
- 22.Delpech B, Maingonnat C, Girard N, Chauzy C, Maunoury R, et al. Hyaluronan and hyaluronectin in the extracellular matrix of human brain tumour stroma. Eur J Cancer. 1993;29A:1012–1017. doi: 10.1016/s0959-8049(05)80214-x. [DOI] [PubMed] [Google Scholar]
- 23.Wiranowska M, Ladd S, Moscinski LC, Hill B, Haller E, et al. Modulation of hyaluronan production by CD44 positive glioma cells. Int J Cancer. 2010;127:532–542. doi: 10.1002/ijc.25085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ariza A, López D, Mate JL, Isamat M, Musulén E, et al. Role of CD44 in the invasiveness of glioblastoma multiforme and the noninvasiveness of meningioma: an immunohistochemistry study. Hum Pathol. 1995;26:1144–1147. doi: 10.1016/0046-8177(95)90278-3. [DOI] [PubMed] [Google Scholar]
- 25.Wiranowska M, Ladd S, Smith SR, Gottschall PE. CD44 adhesion molecule and neuro-glial proteoglycan NG2 as invasive markers of glioma. Brain Cell Biol. 2006;35:159–172. doi: 10.1007/s11068-007-9009-0. [DOI] [PubMed] [Google Scholar]
- 26.Bourguignon LYW. Hyaluronan-mediated CD44 activation of RhoGTPase signaling and cytoskeleton function promotes tumor progression. Semin Cancer Biol. 2008;18:251–259. doi: 10.1016/j.semcancer.2008.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bourguignon LY, Zhu H, Shao L, Chen YW. CD44 interaction with tiam1 promotes Rac1 signaling and hyaluronic acid-mediated breast tumor cell migration. J Biol Chem. 2000;275:1829–1838. doi: 10.1074/jbc.275.3.1829. [DOI] [PubMed] [Google Scholar]
- 28.Herishanu Y, Gibellini F, Njuguna N, Hazan-Halevy I, Keyvanfar K, et al. CD44 signaling via PI3K/AKT and MAPK/ERK pathways protects CLL cells from spontaneous and drug induced apoptosis through MCL-1. Leuk Lymphoma. 2011;52:1758–1769. doi: 10.3109/10428194.2011.569962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Novak U, Stylli SS, Kaye AH, Lepperdinger G. Hyaluronidase-2 overexpression accelerates intracerebral but not subcutaneous tumor formation of murine astrocytoma cells. Cancer Res. 1999;59:6246–6250. [PubMed] [Google Scholar]
- 30.Giese A, Westphal M. Glioma invasion in the central nervous system. Neurosurgery. 1996;39:235–250. doi: 10.1097/00006123-199608000-00001. discussion 250–252. [DOI] [PubMed] [Google Scholar]
- 31.Knott JC, Mahesparan R, Garcia-Cabrera I, Bølge Tysnes B, Edvardsen K, et al. Stimulation of extracellular matrix components in the normal brain by invading glioma cells. Int J Cancer. 1998;75:864–872. doi: 10.1002/(sici)1097-0215(19980316)75:6<864::aid-ijc8>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 32.Tysnes BB, Mahesparan R, Thorsen F, Haugland HK, Porwol T, et al. Laminin expression by glial fibrillary acidic protein positive cells in human gliomas. Int J Dev Neurosci. 1999;17:531–539. doi: 10.1016/s0736-5748(99)00055-6. [DOI] [PubMed] [Google Scholar]
- 33.Yamamoto M, Mohanam S, Sawaya R, Fuller GN, Seiki M, et al. Differential expression of membrane-type matrix metalloproteinase and its correlation with gelatinase A activation in human malignant brain tumors in vivo and in vitro. Cancer Res. 1996;56:384–392. [PubMed] [Google Scholar]
- 34.Demuth T, Berens ME. Molecular Mechanisms of Glioma Cell Migration and Invasion. J Neurooncol. 2004;70:217–228. doi: 10.1007/s11060-004-2751-6. [DOI] [PubMed] [Google Scholar]
- 35.Kaufman LJ, Brangwynne CP, Kasza KE, Filippidi E, Gordon VD, et al. Glioma expansion in collagen I matrices: analyzing collagen concentration-dependent growth and motility patterns. Biophys J. 2005;89:635–650. doi: 10.1529/biophysj.105.061994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kawataki T, Yamane T, Naganuma H, Rousselle P, Andurén I, et al. Laminin isoforms and their integrin receptors in glioma cell migration and invasiveness: Evidence for a role of alpha5-laminin(s) and alpha3beta1 integrin. Exp Cell Res. 2007;313:3819–3831. doi: 10.1016/j.yexcr.2007.07.038. [DOI] [PubMed] [Google Scholar]
- 37.Lathia JD, Li M, Hall PE, Gallagher J, Hale JS, et al. Laminin alpha 2 enables glioblastoma stem cell growth. Ann Neurol. 2012;72:766–778. doi: 10.1002/ana.23674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ohnishi T, Hiraga S, Izumoto S, Matsumura H, Kanemura Y, et al. Role of fibronectin-stimulated tumor cell migration in glioma invasion in vivo: clinical significance of fibronectin and fibronectin receptor expressed in human glioma tissues. Clin Exp Metastasis. 1998;16:729–741. doi: 10.1023/a:1006532812408. [DOI] [PubMed] [Google Scholar]
- 39.Mahesparan R, Read T-A, Lund-Johansen M, Skaftnesmo KO, Bjerkvig R, et al. Expression of extracellular matrix components in a highly infiltrative in vivo glioma model. Acta Neuropathol. 2003;105:49–57. doi: 10.1007/s00401-002-0610-0. [DOI] [PubMed] [Google Scholar]
- 40.Zagzag D, Friedlander DR, Miller DC, Dosik J, Cangiarella J, et al. Tenascin expression in astrocytomas correlates with angiogenesis. Cancer Res. 1995;55:907–914. [PubMed] [Google Scholar]
- 41.Hehlgans S, Haase M, Cordes N. Signalling via integrins: implications for cell survival and anticancer strategies. Biochim Biophys Acta. 2007;1775:163–180. doi: 10.1016/j.bbcan.2006.09.001. [DOI] [PubMed] [Google Scholar]
- 42.Aoudjit F, Vuori K. Integrin signaling inhibits paclitaxel-induced apoptosis in breast cancer cells. Oncogene. 2001;20:4995–5004. doi: 10.1038/sj.onc.1204554. [DOI] [PubMed] [Google Scholar]
- 43.Lathia JD, Gallagher J, Heddleston JM, Wang J, Eyler CE, et al. Integrin alpha 6 regulates glioblastoma stem cells. Cell Stem Cell. 2010;6:421–432. doi: 10.1016/j.stem.2010.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141:52–67. doi: 10.1016/j.cell.2010.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rao JS. Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer. 2003;3:489–501. doi: 10.1038/nrc1121. [DOI] [PubMed] [Google Scholar]
- 46.Discher DE, Janmey P, Wang Y. Tissue Cells Feel and Respond to the Stiffness of Their Substrate. Science. 2005;310:1139–1143. doi: 10.1126/science.1116995. [DOI] [PubMed] [Google Scholar]
- 47.Oakes PW, Beckham Y, Stricker J, Gardel ML. Tension is required but not sufficient for focal adhesion maturation without a stress fiber template. J Cell Biol. 2012;196:363–374. doi: 10.1083/jcb.201107042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kolega J, Janson LW, Taylor DL. The role of solation-contraction coupling in regulating stress fiber dynamics in nonmuscle cells. J Cell Biol. 1991;114:993–1003. doi: 10.1083/jcb.114.5.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chang C-W, Kumar S. Vinculin tension distributions of individual stress fibers within cell–matrix adhesions. J Cell Sci. 2013;126:3021–3030. doi: 10.1242/jcs.119032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Geiger B, Bershadsky A. Assembly and mechanosensory function of focal contacts. Curr Opin Cell Biol. 2001;13:584–592. doi: 10.1016/s0955-0674(00)00255-6. [DOI] [PubMed] [Google Scholar]
- 51.Solon J, Levental I, Sengupta K, Georges PC, Janmey PA. Fibroblast Adaptation and Stiffness Matching to Soft Elastic Substrates. Biophys J. 2007;93:4453–4461. doi: 10.1529/biophysj.106.101386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Pelham RJ, Wang Y. Cell locomotion and focal adhesions are regulated by substrate flexibility. PNAS. 1997;94:13661–13665. doi: 10.1073/pnas.94.25.13661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Maniotis AJ, Chen CS, Ingber DE. Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. PNAS. 1997;94:849–854. doi: 10.1073/pnas.94.3.849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Selbekk T, Brekken R, Solheim O, Lydersen S, Hernes TAN, et al. Tissue Motion and Strain in the Human Brain Assessed by Intraoperative Ultrasound in Glioma Patients. Ultrasound in Medicine & Biology. 2010;36:2–10. doi: 10.1016/j.ultrasmedbio.2009.05.007. [DOI] [PubMed] [Google Scholar]
- 55.Elkin BS, Azeloglu EU, Costa KD, Morrison B., 3rd Mechanical heterogeneity of the rat hippocampus measured by atomic force microscope indentation. J Neurotrauma. 2007;24:812–822. doi: 10.1089/neu.2006.0169. [DOI] [PubMed] [Google Scholar]
- 56.Lefranc F, Brotchi J, Kiss R. Possible future issues in the treatment of glioblastomas: special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis. J Clin Oncol. 2005;23:2411–2422. doi: 10.1200/JCO.2005.03.089. [DOI] [PubMed] [Google Scholar]
- 57.Scherer H. Structural development in gliomas. Am J Cancer. 1938;34:333–348. [Google Scholar]
- 58.Scherer HJ. The Forms of Growth in Gliomas and Their Practical Significance. Brain. 1940;63:1–35. doi: 10.1093/brain/63.1.1. [DOI] [Google Scholar]
- 59.Rutka JT, Muller M, Hubbard SL, Forsdike J, Dirks PB, et al. Astrocytoma adhesion to extracellular matrix: functional significance of integrin and focal adhesion kinase expression. J Neuropathol Exp Neurol. 1999;58:198–209. doi: 10.1097/00005072-199902000-00009. [DOI] [PubMed] [Google Scholar]
- 60.Belot N, Rorive S, Doyen I, Lefranc F, Bruyneel E, et al. Molecular characterization of cell substratum attachments in human glial tumors relates to prognostic features. Glia. 2001;36:375–390. doi: 10.1002/glia.1124. [DOI] [PubMed] [Google Scholar]
- 61.Friedlander DR, Zagzag D, Shiff B, Cohen H, Allen JC, et al. Migration of brain tumor cells on extracellular matrix proteins in vitro correlates with tumor type and grade and involves alphaV and beta1 integrins. Cancer Res. 1996;56:1939–1947. [PubMed] [Google Scholar]
- 62.Paulus W, Baur I, Schuppan D, Roggendorf W. Characterization of integrin receptors in normal and neoplastic human brain. Am J Pathol. 1993;143:154–163. [PMC free article] [PubMed] [Google Scholar]
- 63.Boucher Y, Salehi H, Witwer B, Harsh GR, Jain RK. Interstitial fluid pressure in intracranial tumours in patients and in rodents. Br J Cancer. 1997;75:829–836. doi: 10.1038/bjc.1997.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jain RK, di Tomaso E, Duda DG, Loeffler JS, Sorensen AG, et al. Angiogenesis in brain tumours. Nat Rev Neurosci. 2007;8:610–622. doi: 10.1038/nrn2175. [DOI] [PubMed] [Google Scholar]
- 65.Polacheck WJ, German AE, Mammoto A, Ingber DE, Kamm RD. Mechanotransduction of fluid stresses governs 3D cell migration. PNAS. 2014:201316848. doi: 10.1073/pnas.1316848111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med. 2008;359:492–507. doi: 10.1056/NEJMra0708126. [DOI] [PubMed] [Google Scholar]
- 67.Song S, Ewald AJ, Stallcup W, Werb Z, Bergers G. PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 2005;7:870–879. doi: 10.1038/ncb1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chekenya M, Enger PØ, Thorsen F, Tysnes BB, Al-Sarraj S, et al. The glial precursor proteoglycan, NG2, is expressed on tumour neovasculature by vascular pericytes in human malignant brain tumours. Neuropathol Appl Neurobiol. 2002;28:367–380. doi: 10.1046/j.1365-2990.2002.00412.x. [DOI] [PubMed] [Google Scholar]
- 69.Barbero S, Bajetto A, Bonavia R, Porcile C, Piccioli P, et al. Expression of the chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1 in human brain tumors and their involvement in glial proliferation in vitro. Ann N Y Acad Sci. 2002;973:60–69. doi: 10.1111/j.1749-6632.2002.tb04607.x. [DOI] [PubMed] [Google Scholar]
- 70.Hoelzinger DB, Demuth T, Berens ME. Autocrine factors that sustain glioma invasion and paracrine biology in the brain microenvironment. J Natl Cancer Inst. 2007;99:1583–1593. doi: 10.1093/jnci/djm187. [DOI] [PubMed] [Google Scholar]
- 71.Emdad L, Sarkar D, Su Z-Z, Lee S-G, Kang D-C, et al. Astrocyte elevated gene-1: recent insights into a novel gene involved in tumor progression, metastasis and neurodegeneration. Pharmacol Ther. 2007;114:155–170. doi: 10.1016/j.pharmthera.2007.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rempel SA, Dudas S, Ge S, Gutiérrez JA. Identification and localization of the cytokine SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis in human glioblastoma. Clin Cancer Res. 2000;6:102–111. [PubMed] [Google Scholar]
- 73.Zhou Y, Larsen PH, Hao C, Yong VW. CXCR4 Is a Major Chemokine Receptor on Glioma Cells and Mediates Their Survival. J Biol Chem. 2002;277:49481–49487. doi: 10.1074/jbc.M206222200. [DOI] [PubMed] [Google Scholar]
- 74.Lal PG, Ghirnikar RS, Eng LF. Astrocyte-astrocytoma cell line interactions in culture. J Neurosci Res. 1996;44:216–222. doi: 10.1002/(SICI)1097-4547(19960501)44:3<216::AID-JNR2>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 75.Couldwell WT, Yong VW, Dore-Duffy P, Freedman MS, Antel JP. Production of soluble autocrine inhibitory factors by human glioma cell lines. J Neurol Sci. 1992;110:178–185. doi: 10.1016/0022-510x(92)90026-h. [DOI] [PubMed] [Google Scholar]
- 76.Le DM, Besson A, Fogg DK, Choi K-S, Waisman DM, et al. Exploitation of astrocytes by glioma cells to facilitate invasiveness: a mechanism involving matrix metalloproteinase-2 and the urokinase-type plasminogen activator-plasmin cascade. J Neurosci. 2003;23:4034–4043. doi: 10.1523/JNEUROSCI.23-10-04034.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gilbertson RJ, Rich JN. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer. 2007;7:733–736. doi: 10.1038/nrc2246. [DOI] [PubMed] [Google Scholar]
- 78.Mirzadeh Z, Merkle FT, Soriano-Navarro M, Garcia-Verdugo JM, 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. doi: 10.1016/j.stem.2008.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shen Q, Wang Y, Kokovay E, Lin G, Chuang S-M, et al. Adult SVZ stem cells lie in a vascular niche: A quantitative analysis of niche cell-cell interactions. Cell Stem Cell. 2008;3:289–300. doi: 10.1016/j.stem.2008.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, et al. A perivascular niche for brain tumor stem cells. Cancer Cell. 2007;11:69–82. doi: 10.1016/j.ccr.2006.11.020. [DOI] [PubMed] [Google Scholar]
- 81.Hambardzumyan D, Becher OJ, Rosenblum MK, Pandolfi PP, Manova-Todorova K, et al. PI3K pathway regulates survival of cancer stem cells residing in the perivascular niche following radiation in medulloblastoma in vivo. Genes Dev. 2008;22:436–448. doi: 10.1101/gad.1627008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, et al. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66:7843–7848. doi: 10.1158/0008-5472.CAN-06-1010. [DOI] [PubMed] [Google Scholar]
- 83.Farin A, Suzuki SO, Weiker M, Goldman JE, Bruce JN, et al. Transplanted glioma cells migrate and proliferate on host brain vasculature: a dynamic analysis. Glia. 2006;53:799–808. doi: 10.1002/glia.20334. [DOI] [PubMed] [Google Scholar]
- 84.Charles N, Ozawa T, Squatrito M, Bleau A-M, Brennan CW, et al. Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell. 2010;6:141–152. doi: 10.1016/j.stem.2010.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Infanger DW, Cho Y, Lopez BS, Mohanan S, Liu SC, et al. Glioblastoma stem cells are regulated by interleukin-8 signaling in a tumoral perivascular niche. Cancer Res. 2013;73:7079–7089. doi: 10.1158/0008-5472.CAN-13-1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature. 2010;468:824–828. doi: 10.1038/nature09557. [DOI] [PubMed] [Google Scholar]
- 87.Soda Y, Marumoto T, Friedmann-Morvinski D, Soda M, Liu F, et al. Transdifferentiation of glioblastoma cells into vascular endothelial cells. [Accessed 31 January 2013];PNAS. 2011 doi: 10.1073/pnas.1016030108. Available: http://www.pnas.org/content/early/2011/01/19/1016030108. [DOI] [PMC free article] [PubMed]
- 88.Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, et al. Glioblastoma stem-like cells give rise to tumour endothelium. Nature. 2010;468:829–833. doi: 10.1038/nature09624. [DOI] [PubMed] [Google Scholar]
- 89.Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors. Glia. 2002;40:252–259. doi: 10.1002/glia.10147. [DOI] [PubMed] [Google Scholar]
- 90.Watters JJ, Schartner JM, Badie B. Microglia function in brain tumors. J Neurosci Res. 2005;81:447–455. doi: 10.1002/jnr.20485. [DOI] [PubMed] [Google Scholar]
- 91.Bettinger I, Thanos S, Paulus W. Microglia promote glioma migration. Acta Neuropathol. 2002;103:351–355. doi: 10.1007/s00401-001-0472-x. [DOI] [PubMed] [Google Scholar]
- 92.Markovic DS, Glass R, Synowitz M, van Rooijen N, Kettenmann H. Microglia stimulate the invasiveness of glioma cells by increasing the activity of metalloprotease-2. J Neuropathol Exp Neurol. 2005;64:754–762. doi: 10.1097/01.jnen.0000178445.33972.a9. [DOI] [PubMed] [Google Scholar]
- 93.Galarneau H, Villeneuve J, Gowing G, Julien J-P, Vallières L. Increased glioma growth in mice depleted of macrophages. Cancer Res. 2007;67:8874–8881. doi: 10.1158/0008-5472.CAN-07-0177. [DOI] [PubMed] [Google Scholar]
- 94.Giese A, Loo MA, Rief MD, Tran N, Berens ME. Substrates for astrocytoma invasion. Neurosurgery. 1995;37:294–301. doi: 10.1227/00006123-199508000-00015. [DOI] [PubMed] [Google Scholar]
- 95.Koochekpour S, Pilkington GJ, Merzak A. Hyaluronic acid/CD44H interaction induces cell detachment and stimulates migration and invasion of human glioma cells in vitro. International Journal of Cancer. 1995;63:450–454. doi: 10.1002/ijc.2910630325. [DOI] [PubMed] [Google Scholar]
- 96.Belien ATJ, Paganetti PA, Schwab ME. Membrane-type 1 matrix metalloprotease (MT1-MMP) enables invasive migration of glioma cells in central nervous system white matter. Journal of Cell Biology. 1999;144:373–384. doi: 10.1083/jcb.144.2.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nature Reviews Molecular Cell Biology. 2006;7:211–224. doi: 10.1038/nrm1858. [DOI] [PubMed] [Google Scholar]
- 98.Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell. 2007;130:601–610. doi: 10.1016/j.cell.2007.08.006. [DOI] [PubMed] [Google Scholar]
- 99.Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11:393–410. doi: 10.1038/nrc3064. [DOI] [PubMed] [Google Scholar]
- 100.Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007;99:1441–1454. doi: 10.1093/jnci/djm135. [DOI] [PubMed] [Google Scholar]
- 101.Lo C-M, Wang H-B, Dembo M, Wang Y. Cell Movement Is Guided by the Rigidity of the Substrate. Biophysical Journal. 2000;79:144–152. doi: 10.1016/S0006-3495(00)76279-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cretu A, Castagnino P, Assoian R. Studying the Effects of Matrix Stiffness on Cellular Function using Acrylamide-based. Hydrogels. 2010:e2089. doi: 10.3791/2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Engler A, Bacakova L, Newman C, Hategan A, Griffin M, et al. Substrate compliance versus ligand density in cell on gel responses. Biophys J. 2004;86:617–628. doi: 10.1016/S0006-3495(04)74140-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Johnson KR, Leight JL, Weaver VM. Demystifying the Effects of a Three-Dimensional Microenvironment in Tissue Morphogenesis. Methods in Cell Biology. 2007;83:547–583. doi: 10.1016/s0091-679x(07)83023-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yeung T, Georges PC, Flanagan LA, Marg B, Ortiz M, et al. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil Cytoskeleton. 2005;60:24–34. doi: 10.1002/cm.20041. [DOI] [PubMed] [Google Scholar]
- 106.Ulrich TA, de Juan Pardo EM, Kumar S. The mechanical rigidity of the extracellular matrix regulates the structure, motility, and proliferation of glioma cells. Cancer Res. 2009;69:4167–4174. doi: 10.1158/0008-5472.CAN-08-4859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sen S, Dong MM, Kumar S. Isoform-specific contributions of alpha-actinin to glioma cell mechanobiology. PLoS ONE. 2009;4:e8427. doi: 10.1371/journal.pone.0008427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sen S, Ng WP, Kumar S. Contributions of talin-1 to glioma cell-matrix tensional homeostasis. Journal of The Royal Society Interface. 2011 doi: 10.1098/rsif.2011.0567. Available: http://rsif.royalsocietypublishing.org/content/early/2011/12/05/rsif.2011.0567.abstract. [DOI] [PMC free article] [PubMed]
- 109.MacKay J, Keung A, Kumar S. A Genetic Strategy for the Dynamic and Graded Control of Cell Mechanics, Motility, and Matrix Remodeling. Biophysical Journal. 2012;102:434–442. doi: 10.1016/j.bpj.2011.12.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Trappmann B, Gautrot JE, Connelly JT, Strange DGT, Li Y, et al. Extracellular-matrix tethering regulates stem-cell fate. 2012:642–649. doi: 10.1038/nmat3339. [DOI] [PubMed] [Google Scholar]
- 111.Ananthanarayanan B, Kim Y, Kumar S. Elucidating the mechanobiology of malignant brain tumors using a brain matrix-mimetic hyaluronic acid hydrogel platform. Biomaterials. 2011;32:7913–7923. doi: 10.1016/j.biomaterials.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Fu J, Wang Y-K, Yang MT, Desai RA, Yu X, et al. Mechanical regulation of cell function with geometrically modulated elastomeric substrates. Nat Meth. 2010;7:733–736. doi: 10.1038/nmeth.1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Beadle C, Assanah MC, Monzo P, Vallee R, Rosenfeld SS, et al. The role of Myosin II in glioma invasion of the brain. Molecular Biology of the Cell. 2008;19:3357–3368. doi: 10.1091/mbc.E08-03-0319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ivkovic S, Beadle C, Noticewala S, Massey SC, Swanson KR, et al. Direct inhibition of myosin II effectively blocks glioma invasion in the presence of multiple motogens. Molecular Biology of the Cell. 2012;23:533–542. doi: 10.1091/mbc.E11-01-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Cukierman E. Taking Cell-Matrix Adhesions to the Third Dimension. Science. 2001;294:1708–1712. doi: 10.1126/science.1064829. [DOI] [PubMed] [Google Scholar]
- 116.Lu P, Weaver VM, Werb Z. The extracellular matrix: A dynamic niche in cancer progression. The Journal of Cell Biology. 2012;196:395–406. doi: 10.1083/jcb.201102147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gordon VD, Valentine MT, Gardel ML, Andor-Ardó D, Dennison S, et al. Measuring the mechanical stress induced by an expanding multicellular tumor system: a case study. Experimental Cell Research. 2003;289:58–66. doi: 10.1016/S0014-4827(03)00256-8. [DOI] [PubMed] [Google Scholar]
- 118.Hegedus B, Marga F, Jakab K, Sharpe-Timms KL, Forgacs G. The interplay of cell-cell and cell-matrix interactions in the invasive properties of brain tumors. Biophysical Journal. 2006;91:2708–2716. doi: 10.1529/biophysj.105.077834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kim HD, Guo TW, Wu AP, Wells A, Gertler FB, et al. Epidermal Growth Factor-induced enhancement of glioblastoma cell migration in 3D arises from an intrinsic increase in speed but an extrinsic matrix- and proteolysis-dependent increase in persistence. Molecular Biology of the Cell. 2008;19:4249–4259. doi: 10.1091/mbc.E08-05-0501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yang Y, Motte S, Kaufman LJ. Pore size variable type I collagen gels and their interaction with glioma cells. Biomaterials. 2010;31:5678–5688. doi: 10.1016/j.biomaterials.2010.03.039. [DOI] [PubMed] [Google Scholar]
- 121.Thorne RG, Nicholson C. In vivo diffusion analysis with quantum dots and dextrans predicts the width of brain extracellular space. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:5567–5572. doi: 10.1073/pnas.0509425103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Seliktar D. Designing Cell-Compatible Hydrogels for Biomedical Applications. Science. 2012;336:1124–1128. doi: 10.1126/science.1214804. [DOI] [PubMed] [Google Scholar]
- 123.Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature Biotechnology. 2005;23:47–55. doi: 10.1038/nbt1055. [DOI] [PubMed] [Google Scholar]
- 124.Ulrich TA, Jain A, Tanner K, MacKay JL, Kumar S. Probing cellular mechanobiology in three-dimensional culture with collagen–agarose matrices. Biomaterials. 2010;31:1875–1884. doi: 10.1016/j.biomaterials.2009.10.047. [DOI] [PubMed] [Google Scholar]
- 125.Ulrich TA, Lee TG, Shon HK, Moon DW, Kumar S. Microscale mechanisms of agarose-induced disruption of collagen remodeling. Biomaterials. 2011;32:5633–5642. doi: 10.1016/j.biomaterials.2011.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nature Cell Biology. 2003;5:711–719. doi: 10.1038/ncb1019. [DOI] [PubMed] [Google Scholar]
- 127.Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, et al. Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. Journal of Cell Biology. 2003;160:267–277. doi: 10.1083/jcb.200209006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Friedl P, Wolf K. Tumour-cell invasion and migration: Diversity and escape mechanisms. Nature Reviews Cancer. 2003;3:362–374. doi: 10.1038/nrc1075. [DOI] [PubMed] [Google Scholar]
- 129.Marklein RA, Burdick JA. Spatially controlled hydrogel mechanics to modulate stem cell interactions. Soft Matter. 2010;6:136. doi: 10.1039/b916933d. [DOI] [Google Scholar]
- 130.Ponta H, Sherman L, Herrlich PA. CD44: From adhesion molecules to signalling regulators. Nature Reviews Molecular Cell Biology. 2003;4:33–45. doi: 10.1038/nrm1004. [DOI] [PubMed] [Google Scholar]
- 131.Xu Y, Stamenkovic I, Yu Q. CD44 attenuates activation of the Hippo signaling pathway and is a prime therapeutic target for glioblastoma. Cancer Research. 2010;70:2455–2464. doi: 10.1158/0008-5472.can-09-2505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Khademhosseini A, Langer R, Borenstein J, Vacanti JP. Microscale technologies for tissue engineering and biology. Proc Natl Acad Sci U S A. 2006;103:2480–2487. doi: 10.1073/pnas.0507681102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ross AM, Jiang ZX, Bastmeyer M, Lahann J. Physical Aspects of Cell Culture Substrates: Topography, Roughness, and Elasticity. Small. 2012;8:336–355. doi: 10.1002/smll.201100934. [DOI] [PubMed] [Google Scholar]
- 134.Polacheck WJ, Zervantonakis IK, Kamm RD. Tumor cell migration in complex microenvironments. Cellular and Molecular Life Sciences. 2013;70:1335–1356. doi: 10.1007/s00018-012-1115-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zhu BS, Zhang QQ, Lu QH, Xu YH, Yin J, et al. Nanotopographical guidance of C6 glioma cell alignment and oriented growth. Biomaterials. 2004;25:4215–4223. doi: 10.1016/j.biomaterials.2003.11.020. [DOI] [PubMed] [Google Scholar]
- 136.Gallego-Perez D, Higuita-Castro N, Denning L, DeJesus J, Dahl K, et al. Microfabricated mimics of in vivo structural cues for the study of guided tumor cell migration. Lab on a Chip. 2012;12:4424–4432. doi: 10.1039/c2lc40726d. [DOI] [PubMed] [Google Scholar]
- 137.Pathak A, Kumar S. Independent regulation of tumor cell migration by matrix stiffness and confinement. [Accessed 20 August 2012];PNAS. 2012 doi: 10.1073/pnas.1118073109. Available: http://www.pnas.org/content/early/2012/06/07/1118073109. [DOI] [PMC free article] [PubMed]
- 138.Pathak A, Kumar S. Transforming potential and matrix stiffness co-regulate confinement sensitivity of tumor cell migration. Integr Biol (Camb) 2013;5:1067–1075. doi: 10.1039/c3ib40017d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mammoto T, Jiang A, Jiang E, Panigrahy D, Kieran MW, et al. Role of Collagen Matrix in Tumor Angiogenesis and Glioblastoma Multiforme Progression. The American Journal of Pathology. 2013 doi: 10.1016/j.ajpath.2013.06.026. Available: http://www.sciencedirect.com/science/article/pii/S0002944013004823. [DOI] [PMC free article] [PubMed]
- 140.Sharpless NE, Depinho RA. The mighty mouse: genetically engineered mouse models in cancer drug development. Nat Rev Drug Discov. 2006;5:741–754. doi: 10.1038/nrd2110. [DOI] [PubMed] [Google Scholar]
- 141.Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 2001;84:1424–1431. doi: 10.1054/bjoc.2001.1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hutmacher DW. Biomaterials offer cancer research the third dimension. Nat Mater. 2010;9:90–93. doi: 10.1038/nmat2619. [DOI] [PubMed] [Google Scholar]
- 143.Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, et al. Engineering tumors with 3D scaffolds. Nat Methods. 2007;4:855–860. doi: 10.1038/nmeth1085. [DOI] [PubMed] [Google Scholar]
- 144.Liu Y, Shu XZ, Prestwich GD. Tumor engineering: orthotopic cancer models in mice using cell-loaded, injectable, cross-linked hyaluronan-derived hydrogels. Tissue Eng. 2007;13:1091–1101. doi: 10.1089/ten.2006.0297. [DOI] [PubMed] [Google Scholar]
- 145.Bravo-Cordero JJ, Hodgson L, Condeelis J. Directed cell invasion and migration during metastasis. Current Opinion in Cell Biology. 2012;24:277–283. doi: 10.1016/j.ceb.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wells A, Grahovac J, Wheeler S, Ma B, Lauffenburger D. Targeting tumor cell motility as a strategy against invasion and metastasis. Trends in Pharmacological Sciences. 2013;34:283–289. doi: 10.1016/j.tips.2013.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Hirata E, Yukinaga H, Kamioka Y, Arakawa Y, Miyamoto S, et al. In vivo fluorescence resonance energy transfer imaging reveals differential activation of Rho-family GTPases in glioblastoma cell invasion. Journal of Cell Science. 2012;125:858–868. doi: 10.1242/jcs.089995. [DOI] [PubMed] [Google Scholar]
- 148.Friedl P, Alexander S. Cancer Invasion and the Microenvironment: Plasticity and Reciprocity. Cell. 2011;147:992–1009. doi: 10.1016/j.cell.2011.11.016. [DOI] [PubMed] [Google Scholar]
- 149.Friedl P, Wolf K. Plasticity of cell migration: a multiscale tuning model. The Journal of Cell Biology. 2009;188:11–19. doi: 10.1083/jcb.200909003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Petrie RJ, Yamada KM. At the leading edge of three-dimensional cell migration. Journal of Cell Science. 2013 doi: 10.1242/jcs.093732. Available: http://jcs.biologists.org/content/early/2013/01/31/jcs.093732.abstract. [DOI] [PMC free article] [PubMed]
- 151.Ruiz-Ontanon P, Orgaz JL, Aldaz B, Elosegui-Artola A, Martino J, et al. Cellular Plasticity Confers Migratory and Invasive Advantages to a Population of Glioblastoma-Initiating Cells that Infiltrate Peritumoral Tissue. Stem Cells. 2013;31:1075–1085. doi: 10.1002/stem.1349. [DOI] [PubMed] [Google Scholar]
- 152.Weeks A, Okolowsky N, Golbourn B, Ivanchuk S, Smith C, et al. ECT2 and RASAL2 Mediate Mesenchymal-Amoeboid Transition In Human Astrocytoma Cells. The American Journal of Pathology. 2012;181:662–674. doi: 10.1016/j.ajpath.2012.04.011. [DOI] [PubMed] [Google Scholar]
- 153.Yamazaki D, Kurisu S, Takenawa T. Involvement of Rac and Rho signaling in cancer cell motility in 3D substrates. Oncogene. 2009;28:1570–1583. doi: 10.1038/onc.2009.2. [DOI] [PubMed] [Google Scholar]
- 154.Wolf K, te Lindert M, Krause M, Alexander S, te Riet J, et al. Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol. 2013;201:1069–1084. doi: 10.1083/jcb.201210152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Davies SJA, Fitch MT, Memberg SP, Hall AK, Raisman G, et al. Regeneration of adult axons in white matter tracts of the central nervous system. Nature. 1997;390:680–683. doi: 10.1038/37776. [DOI] [PubMed] [Google Scholar]
- 156.Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mörk S, et al. Axonal Transection in the Lesions of Multiple Sclerosis. New England Journal of Medicine. 1998;338:278–285. doi: 10.1056/NEJM199801293380502. [DOI] [PubMed] [Google Scholar]
- 157.Merrill JE. In vitro and in vivo pharmacological models to assess demyelination and remyelination. Neuropsychopharmacology. 2009;34:55–73. doi: 10.1038/npp.2008.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Bhardwaj N, Kundu SC. Electrospinning: A fascinating fiber fabrication technique. Biotechnology Advances. 2010;28:325–347. doi: 10.1016/j.biotechadv.2010.01.004. [DOI] [PubMed] [Google Scholar]
- 159.Christopherson GT, Song H, Mao H-Q. The influence of fiber diameter of electrospun substrates on neural stem cell differentiation and proliferation. Biomaterials. 2009;30:556–564. doi: 10.1016/j.biomaterials.2008.10.004. [DOI] [PubMed] [Google Scholar]
- 160.Chew SY, Mi R, Hoke A, Leong KW. The effect of the alignment of electrospun fibrous scaffolds on Schwann cell maturation. Biomaterials. 2008;29:653–661. doi: 10.1016/j.biomaterials.2007.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Corey JM, Gertz CC, Wang B-S, Birrell LK, Johnson SL, et al. The design of electrospun PLLA nanofiber scaffolds compatible with serum-free growth of primary motor and sensory neurons. Acta Biomater. 2008;4:863–875. doi: 10.1016/j.actbio.2008.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Prabhakaran MP, Venugopal JR, Ramakrishna S. Mesenchymal stem cell differentiation to neuronal cells on electrospun nanofibrous substrates for nerve tissue engineering. Biomaterials. 2009;30:4996–5003. doi: 10.1016/j.biomaterials.2009.05.057. [DOI] [PubMed] [Google Scholar]
- 163.Johnson J, Nowicki MO, Lee CH, Chiocca EA, Viapiano MS, et al. Quantitative Analysis of Complex Glioma Cell Migration on Electrospun Polycaprolactone Using Time-Lapse Microscopy. Tissue Engineering Part C: Methods. 2009;15:531–540. doi: 10.1089/ten.tec.2008.0486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Rao SS, Nelson MT, Xue R, DeJesus JK, Viapiano MS, et al. Mimicking white matter tract topography using core–shell electrospun nanofibers to examine migration of malignant brain tumors. Biomaterials. 2013;34:5181–5190. doi: 10.1016/j.biomaterials.2013.03.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Chen Z, Htay A, Dos Santos W, Gillies GT, Fillmore HL, et al. In vitro angiogenesis by human umbilical vein endothelial cells (HUVEC) induced by three-dimensional co-culture with glioblastoma cells. J Neurooncol. 2009;92:121–128. doi: 10.1007/s11060-008-9742-y. [DOI] [PubMed] [Google Scholar]
- 166.Hurst RD, Fritz IB. Properties of an immortalised vascular endothelial/glioma cell co-culture model of the blood-brain barrier. J Cell Physiol. 1996;167:81–88. doi: 10.1002/(SICI)1097-4652(199604)167:1<81::AID-JCP9>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
- 167.Zhu TS, Costello MA, Talsma CE, Flack CG, Crowley JG, et al. Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells. Cancer Res. 2011;71:6061–6072. doi: 10.1158/0008-5472.CAN-10-4269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Nehls V, Drenckhahn D. A Novel, Microcarrier-Based in Vitro Assay for Rapid and Reliable Quantification of Three-Dimensional Cell Migration and Angiogenesis. Microvascular Research. 1995;50:311–322. doi: 10.1006/mvre.1995.1061. [DOI] [PubMed] [Google Scholar]
- 169.Leslie-Barbick JE, Shen C, Chen C, West JL. Micron-scale spatially patterned, covalently immobilized vascular endothelial growth factor on hydrogels accelerates endothelial tubulogenesis and increases cellular angiogenic responses. Tissue Eng Part A. 2011;17:221–229. doi: 10.1089/ten.TEA.2010.0202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Baker BM, Trappmann B, Stapleton SC, Toro E, Chen CS. Microfluidics embedded within extracellular matrix to define vascular architectures and pattern diffusive gradients. Lab Chip. 2013;13:3246–3252. doi: 10.1039/C3LC50493J. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT, et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat Mater. 2012;11:768–774. doi: 10.1038/nmat3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Zheng Y, Chen J, Craven M, Choi NW, Totorica S, et al. In vitro microvessels for the study of angiogenesis and thrombosis. PNAS. 2012;109:9342–9347. doi: 10.1073/pnas.1201240109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Clark AK, Taubenberger AV, Taylor RA, Niranjan B, Chea ZY, et al. A bioengineered microenvironment to quantitatively measure the tumorigenic properties of cancer-associated fibroblasts in human prostate cancer. Biomaterials. 2013;34:4777–4785. doi: 10.1016/j.biomaterials.2013.03.005. [DOI] [PubMed] [Google Scholar]
- 174.Kaji H, Camci-Unal G, Langer R, Khademhosseini A. Engineering systems for the generation of patterned co-cultures for controlling cell-cell interactions. Biochim Biophys Acta. 2011;1810:239–250. doi: 10.1016/j.bbagen.2010.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Bhatia SN, Yarmush ML, Toner M. Controlling cell interactions by micropatterning in co-cultures: hepatocytes and 3T3 fibroblasts. J Biomed Mater Res. 1997;34:189–199. doi: 10.1002/(sici)1097-4636(199702)34:2<189::aid-jbm8>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 176.Chiu DT, Jeon NL, Huang S, Kane RS, Wargo CJ, et al. Patterned deposition of cells and proteins onto surfaces by using three-dimensional microfluidic systems. Proc Natl Acad Sci USA. 2000;97:2408–2413. doi: 10.1073/pnas.040562297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Kaji H, Kanada M, Oyamatsu D, Matsue T, Nishizawa M. Microelectrochemical approach to induce local cell adhesion and growth on substrates. Langmuir. 2004;20:16–19. doi: 10.1021/la035537f. [DOI] [PubMed] [Google Scholar]
- 178.Yamato M, Kwon OH, Hirose M, Kikuchi A, Okano T. Novel patterned cell coculture utilizing thermally responsive grafted polymer surfaces. J Biomed Mater Res. 2001;55:137–140. doi: 10.1002/1097-4636(200104)55:1<137::aid-jbm180>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 179.Du Y, Lo E, Ali S, Khademhosseini A. Directed assembly of cell-laden microgels for fabrication of 3D tissue constructs. Proc Natl Acad Sci USA. 2008;105:9522–9527. doi: 10.1073/pnas.0801866105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Harimoto M, Yamato M, Hirose M, Takahashi C, Isoi Y, et al. Novel approach for achieving double-layered cell sheets co-culture: overlaying endothelial cell sheets onto monolayer hepatocytes utilizing temperature-responsive culture dishes. J Biomed Mater Res. 2002;62:464–470. doi: 10.1002/jbm.10228. [DOI] [PubMed] [Google Scholar]
- 181.Wong AP, Perez-Castillejos R, Christopher Love J, Whitesides GM. Partitioning microfluidic channels with hydrogel to construct tunable 3-D cellular microenvironments. Biomaterials. 2008;29:1853–1861. doi: 10.1016/j.biomaterials.2007.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Fernandes TG, Diogo MM, Clark DS, Dordick JS, Cabral JMS. High-throughput cellular microarray platforms: applications in drug discovery, toxicology and stem cell research. Trends Biotechnol. 2009;27:342–349. doi: 10.1016/j.tibtech.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Alberti K, Davey RE, Onishi K, George S, Salchert K, et al. Functional immobilization of signaling proteins enables control of stem cell fate. Nat Methods. 2008;5:645–650. doi: 10.1038/nmeth.1222. [DOI] [PubMed] [Google Scholar]
- 184.Anderson DG, Levenberg S, Langer R. Nanoliter-scale synthesis of arrayed biomaterials and application to human embryonic stem cells. Nat Biotech. 2004;22:863–866. doi: 10.1038/nbt981. [DOI] [PubMed] [Google Scholar]
- 185.Nakajima M, Ishimuro T, Kato K, Ko I-K, Hirata I, et al. Combinatorial protein display for the cell-based screening of biomaterials that direct neural stem cell differentiation. Biomaterials. 2007;28:1048–1060. doi: 10.1016/j.biomaterials.2006.10.004. [DOI] [PubMed] [Google Scholar]
