Elsevier

Biomaterials

Volume 35, Issue 31, October 2014, Pages 8846-8853
Biomaterials

A composite hydrogel platform for the dissection of tumor cell migration at tissue interfaces

https://doi.org/10.1016/j.biomaterials.2014.07.003Get rights and content

Abstract

Glioblastoma multiforme (GBM), the most prevalent primary brain cancer, is characterized by diffuse infiltration of tumor cells into brain tissue, which severely complicates surgical resection and contributes to tumor recurrence. The most rapid mode of tissue infiltration occurs along blood vessels or white matter tracts, which represent topological interfaces thought to serve as “tracks” that speed cell migration. Despite this observation, the field lacks experimental paradigms that capture key features of these tissue interfaces and allow reductionist dissection of mechanisms of this interfacial motility. To address this need, we developed a culture system in which tumor cells are sandwiched between a fibronectin-coated ventral surface representing vascular basement membrane and a dorsal hyaluronic acid (HA) surface representing brain parenchyma. We find that inclusion of the dorsal HA surface induces formation of adhesive complexes and significantly slows cell migration relative to a free fibronectin-coated surface. This retardation is amplified by inclusion of integrin binding peptides in the dorsal layer and expression of CD44, suggesting that the dorsal surface slows migration through biochemically specific mechanisms rather than simple steric hindrance. Moreover, both the reduction in migration speed and assembly of dorsal adhesions depend on myosin activation and the stiffness of the ventral layer, implying that mechanochemical feedback directed by the ventral layer can influence adhesive signaling at the dorsal surface.

Introduction

Cell migration and the mechanisms that underlie specific migratory phenotypes are increasingly recognized to depend on extracellular context, especially the structure and mechanics of the extracellular matrix (ECM) [1], [2], [3]. On planar two-dimensional substrates, migration is typically described as being driven by a balance between actin polymerization at the cell front and actomyosin contraction at the cell rear that is transmitted to the ECM via adhesions [4]. In three-dimensional ECMs, migration can take various forms including mesenchymal migration (perhaps most analogous to classical two-dimensional migration) to amoeboid migration, which is less adhesion-dependent and leverages intracellular hydrostatic pressure generated by actomyosin contractility to extrude the cell body through matrix pores [5]. Importantly, the molecular mechanisms that control these migration modes are as diverse as the number of migratory phenotypes. In fact, many cells dynamically switch from one mode to another as they encounter and navigate different microenvironments, highlighting the importance of studying cell migration in culture systems that capture defining architectural features of tissue [6], [7], [8].

Cell migration is often guided by heterogeneous structures within the ECM; for example, a diverse variety of invasive solid tumors proceed along pre-existing anatomical structures [9], [10], [11], [12]. Metastastatic tumor cells have been clinically observed to preferentially migrate in bone cavities or between adipocytes, suggesting that the topographies of these structures may facilitate tissue dissemination [10]. Migration in this context may be regarded as being “interfacial” in nature, in that cells translocate along a ventral two-dimensional surface while surrounded on their dorsolateral surface by an amorphous ECM of a different composition. Other examples of interfacial migration are tumor cells that migrate between bundles of myelinated axons and connective brain tissue [10], [13].

A particularly important example of interfacial migration is the invasion of glioblastoma multiforme (GBM), the most common and deadly primary brain tumor. The extreme lethality of this malignancy is attributed in part to its diffuse and unrelenting infiltration of brain tissue, effectively precluding complete surgical resection [14]. GBM invasion patterns are unlike most other aggressive malignancies, in that GBM cells rarely intravasate and metastasize to distant tissues, instead remaining within the brain [14], [15]. The pre-existing structures that guide GBM, collectively known as the secondary structures of Scherer, include the subpial space, white matter tracts, and vascular beds [16]. While these structures are widely acknowledged to facilitate invasive migration, relatively little is known about the biophysical and molecular mechanisms through which they do so. For example, cells migrating along vascular beds simultaneously experience strong integrin-based inputs via fibronectin and laminin in the vascular basement membrane [15] while also receiving adhesive inputs from hyaluronic acid (HA) in the brain parenchyma, which can be mediated by HA receptors such as CD44 and RHAMM [17], [18]. There are also substantial biophysical asymmetries within this adhesive microenvironment, as vascular beds tend to be orders of magnitude stiffer than the surrounding parenchyma [19], [20], [21]. How these asymmetric signals are integrated to regulate migration in GBM remains unknown.

Despite the acknowledged importance of migration along asymmetric tissue interfaces in many tumors, comparatively little is known about the molecular mechanisms that underlie this process. The fact that migration mechanisms depend strongly on context has created an unmet need for experimental paradigms that recapitulate key aspects of these interfaces. To address this need, we developed a simple experimental system that features asymmetric ECM signals representative of the brain parenchyma–vascular interface, and used it to investigate molecular mechanisms of adhesion and motility.

Section snippets

HA-methacrylate synthesis

Methacrylated HA was synthesized as described previously [22]. Briefly, high molecular weight HA (66 kDa-90 kDa; Lifecore technologies) was dissolved at 1 wt% in deionized water, and then a six-fold molar excess of methacrylic anhydride (Sigma) was added dropwise to the solution on ice. The pH of the reaction was adjusted to a value greater than 8, where it was held for the duration of the experiment. The reaction was allowed to proceed overnight. HA-methacrylate was isolated by the addition of

Modeling perivascular migration with interfacial hydrogels

As described earlier, vascular structures are an important secondary Structure of Scherer along which GBM cells invade brain tissue. To model this process in culture, we developed an in vitro overlay culture system that recapitulates key features of the matrix environment found at this interface and allows for systematic and independent control of the biophysical and biochemical properties of each matrix (Fig. 1A). To mimic the fibronectin-rich basolateral membrane of the vasculature, we used

Discussion

Much previous research has illustrated the importance of studying cell migration in a context that recapitulates critical biochemical and biophysical features of the corresponding tissue microenvironment. In this study, we created a simple, reproducible system that allowed us to systematically modulate the asymmetric biophysical and biochemical properties of the dorsal and ventral surfaces of a model tissue interface. This interface could be tailored to include features of the vascular basement

Conclusion

We have created a simple model system to recapitulate migration of GBM cells along vascular interfaces. By manipulating both adhesive receptors and matrix properties, we discovered that cells migrate most efficiently when adhesion to the dorsal surface is weak, whether that adhesion was mediated by CD44 or integrins. We anticipate that this paradigm could readily be adapted to other tumor systems and will facilitate deeper investigation into mechanisms through which these tumors infiltrate

Acknowledgments

This work was supported by awards from the NIH (1F32CA174361 to AR, 1DP2OD004213 and 1R21CA174573 to SK), an NSF/NCI Physical and Engineering Sciences in Oncology Award (CMMI 1105539 to SK), and the W. M. Keck Foundation.

References (48)

  • G. Mehta et al.

    Synergistic effects of tethered growth factors and adhesion ligands on DNA synthesis and function of primary hepatocytes cultured on soft synthetic hydrogels

    Biomaterials

    (2010)
  • A. Pathak et al.

    Biophysical regulation of tumor cell invasion: moving beyond matrix stiffness

    Integr Biol

    (2011)
  • E. Sahai et al.

    Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis

    Nat Cell Biol

    (2003)
  • A.D. Doyle et al.

    One-dimensional topography underlies three-dimensional fibrillar cell migration

    J Cell Biol

    (2009)
  • P. Friedl et al.

    Tumour-cell invasion and migration: diversity and escape mechanisms

    Nat Rev Cancer

    (2003)
  • P. Friedl et al.

    Plasticity of cell migration: a multiscale tuning model

    J Cell Biol

    (2009)
  • M. Tozluoğlu et al.

    Matrix geometry determines optimal cancer cell migration strategy and modulates response to interventions

    Nat Cell Biol

    (2013)
  • R.J. Petrie et al.

    Nonpolarized signaling reveals two distinct modes of 3D cell migration

    J Cell Biol

    (2012)
  • J. Condeelis et al.

    Intravital imaging of cell movement in tumours

    Nat Rev Cancer

    (2003)
  • S. Alexander et al.

    Dynamic imaging of cancer growth and invasion: a modified skin-fold chamber model

    Histochem Cell Biol

    (2008)
  • G.P. Gritsenko et al.

    Interstitial guidance of cancer invasion

    J Pathol

    (2012)
  • S. Agnihotri et al.

    Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies

    Arch Immunol Ther Exp

    (2013)
  • J.J. Bernstein et al.

    Glioblastoma cells do not intravasate into blood vessels

    Neurosurgery

    (1995)
  • H.J. Scherer

    The forms of growth in gliomas and their practical significance

    Brain

    (1940)
  • Cited by (38)

    • Hydrogel matrix presence and composition influence drug responses of encapsulated glioblastoma spheroids

      2021, Acta Biomaterialia
      Citation Excerpt :

      Glioblastoma multiforme (GBM) is the most malignant primary tumor of the brain with a median patient survival rate of 12–15 months [1,2]. Although it rarely metastasizes to distant tissues, it grows characteristically by infiltrating the surrounding brain tissue [3]. The standard of care for GBM patients includes surgical resection of the bulk tumor mass followed by systemic chemotherapy with temozolomide (TMZ) coupled with local radiation therapy [4,5].

    • Piezo-type mechanosensitive ion channel component 1 (Piezo1) in human cancer

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      In vitro studies have shown that Piezo1 is an important target for the biological function of OSCC cells and that elevated expression of Piezo1 promotes the proliferation, migration, and invasion of OSCC cells, induces tumor vascular mimicry, and promotes tumor progression [39,78]. Piezo1 promotes OSCC cell proliferation by acting as a transcriptional target of the yes-associated protein (YAP) signal [78] and interacting with the integrin focal adhesion kinase (FAK) signal, a pathway that promotes OSCC tissue sclerosis [79]. These findings suggest that Piezo1 is an important target for OSCC diagnosis and prognosis, and it is a new strategy for cancer treatment.

    • A Feedforward Mechanism Mediated by Mechanosensitive Ion Channel PIEZO1 and Tissue Mechanics Promotes Glioma Aggression

      2018, Neuron
      Citation Excerpt :

      Modeling the altered environmental mechanics and stiffness using a hydrogel-based cell culture platform has revealed that glioma cells interact with the ECM to regulate its expression of the oncogenic microRNA miR18a (Rape et al., 2015). The complex physical environment and varying stiffness encountered by different areas of the glioma cells can modulate their migratory capacity (Rape and Kumar, 2014). Increased environmental stiffness can also promote glioma cell proliferation by spatially and biochemically amplifying epidermal growth factor receptor (EGFR) signaling (Umesh et al., 2014).

    View all citing articles on Scopus
    View full text