Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Tissue mechanics promote IDH1-dependent HIF1α–tenascin C feedback to regulate glioblastoma aggression

An Author Correction to this article was published on 04 April 2023

This article has been updated

Abstract

Increased overall survival for patients with glioma brain tumours is associated with mutations in the metabolic regulator isocitrate dehydrogenase 1 (IDH1). Gliomas develop within a mechanically challenged microenvironment that is characterized by a dense extracellular matrix (ECM) that compromises vascular integrity to induce hypoxia and activate HIF1α. We found that glioma aggression and patient prognosis correlate with HIF1α levels and the stiffness of a tenascin C (TNC)-enriched ECM. Gain- and loss-of-function xenograft manipulations demonstrated that a mutant IDH1 restricts glioma aggression by reducing HIF1α-dependent TNC expression to decrease ECM stiffness and mechanosignalling. Recurrent IDH1-mutant patient gliomas had a stiffer TNC-enriched ECM that our studies attributed to reduced miR-203 suppression of HIF1α and TNC mediated via a tension-dependent positive feedback loop. Thus, our work suggests that elevated ECM stiffness can independently foster glioblastoma aggression and contribute to glioblastoma recurrence via bypassing the protective activity of IDH1 mutational status.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: ECM stiffness associates with IDH1 mutations in primary GBM tumours.
Figure 2: TNC modifies ECM stiffness and mechanosignalling.
Figure 3: HIF1α directly regulates TNC expression.
Figure 4: R132H IDH1 primary GBMs cannot tune HIF1α.
Figure 5: Mechanosignalling promotes R132H IDH1 tumour aggression.
Figure 6: miR-203 targets HIF1α and TNC.

Similar content being viewed by others

Change history

References

  1. Brat, D. J. et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 372, 2481–2498 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Van den Bent, M. J. et al. Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J. Clin. Oncol. 31, 344–350 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Cairncross, G. et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J. Clin. Oncol. 31, 337–343 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Losman, J.-A. J. A. & Kaelin, W. G. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev. 27, 836–852 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhao, S. et al. Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1. Science 324, 261–265 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Koivunen, P. et al. Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature 483, 484–488 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mouw, J. K. et al. Tissue mechanics modulate microRNA-dependent PTEN expression to regulate malignant progression. Nat. Med. 20, 360–367 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kumar, S. & Weaver, V. M. Mechanics, malignancy, and metastasis: the force journey of a tumor cell. Cancer Metastasis Rev. 28, 113–127 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Lopez, J. I., Kang, I., You, W.-K., McDonald, D. M. & Weaver, V. M. In situ force mapping of mammary gland transformation. Integr. Biol. (Camb) 3, 910–921 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Geiss, G. K. et al. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat. Biotechnol. 26, 317–325 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Colman, H. et al. A multigene predictor of outcome in glioblastoma. Neuro-Oncol. 12, 49–57 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Ruoslahti, E. Brain extracellular matrix. Glycobiology 6, 489–492 (1996).

    Article  CAS  PubMed  Google Scholar 

  15. Wade, A. et al. Proteoglycans and their roles in brain cancer. FEBS J. 280, 2399–2417 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zimmermann, D. R. & Dours-Zimmermann, M. T. Extracellular matrix of the central nervous system: from neglect to challenge. Histochem. Cell Biol. 130, 635–653 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Cancer Genome Atlas Research NetworkComprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  18. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Masson, N. & Ratcliffe, P. J. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab. 2, 3 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Koperek, O., Akin, E., Asari, R., Niederle, B. & Neuhold, N. Expression of hypoxia-inducible factor 1 α in papillary thyroid carcinoma is associated with desmoplastic stromal reaction and lymph node metastasis. Virchows Arch. 463, 795–802 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Midwood, K. S. & Orend, G. The role of tenascin-C in tissue injury and tumorigenesis. J. Cell Commun. Signal. 3, 287–310 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Turcan, S. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chesnelong, C. et al. Lactate dehydrogenase A silencing in IDH mutant gliomas. Neuro-Oncol. 16, 686–695 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Tarhonskaya, H. et al. Non-enzymatic chemistry enables 2-hydroxyglutarate-mediated activation of 2-oxoglutarate oxygenases. Nat. Commun. 5, 3423 (2014).

    Article  PubMed  Google Scholar 

  25. Wierenga, A. T. J., Vellenga, E. & Schuringa, J. J. Convergence of hypoxia and TGFβ pathways on cell cycle regulation in human hematopoietic stem/progenitor cells. PLoS ONE 9, e93494 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Marucci, G. et al. Pathological spectrum in recurrences of glioblastoma multiforme. Pathologica 107, 1–8 (2015).

    CAS  PubMed  Google Scholar 

  27. Li, R. et al. Genetic and clinical characteristics of primary and secondary glioblastoma is associated with differential molecular subtype distribution. Oncotarget 6, 7318–7324 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Barcellos-Hoff, M. H. & Ravani, S. A. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60, 1254–1260 (2000).

    CAS  PubMed  Google Scholar 

  29. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Miroshnikova, Y. A. et al. Engineering strategies to recapitulate epithelial morphogenesis within synthetic three-dimensional extracellular matrix with tunable mechanical properties. Phys. Biol. 8, 026013 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chappell, W. H. et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health. Oncotarget 2, 135–164 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Chang, J. H. et al. MicroRNA-203 modulates the radiation sensitivity of human malignant glioma cells. Int. J. Radiat. Oncol. Biol. Phys. 94, 412–420 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Liao, H. et al. MiR-203 downregulation is responsible for chemoresistance in human glioblastoma by promoting epithelial-mesenchymal transition via SNAI2. Oncotarget 6, 8914–8928 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Le, L. T.-N. et al. Loss of miR-203 regulates cell shape and matrix adhesion through ROBO1/Rac/FAK in response to stiffness. J. Cell Biol. 212, 707–719 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Scherr, M. et al. Lentivirus-mediated antagomir expression for specific inhibition of miRNA function. Nucleic Acids Res. 35, e149 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Williams, S. C. et al. R132H-mutation of isocitrate dehydrogenase-1 is not sufficient for HIF-1α upregulation in adult glioma. Acta Neuropathol. 121, 279–281 (2011).

    Article  PubMed  Google Scholar 

  37. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Noushmehr, H. et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17, 510–522 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Evans, S. M. et al. Hypoxia is important in the biology and aggression of human glial brain tumors. Clin. Cancer Res. 10, 8177–8184 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Vordermark, D. Significance of hypoxia in malignant glioma. Re: Evans et al. Hypoxia is important in the biology and aggression of human glial brain tumors. Clin Cancer Res 2004;10:8177–84. Clin. Cancer Res. 11, 3966–3967 (2005); Evans, S. M. et al. response 3967–3968.

    Article  PubMed  Google Scholar 

  41. Lu, K. V. et al. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 22, 21–35 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Johnson, K. R., Leight, J. L. & Weaver, V. M. Demystifying the effects of a three-dimensional microenvironment in tissue morphogenesis. Methods Cell Biol. 83, 547–583 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sarkaria, J. N. et al. Use of an orthotopic xenograft model for assessing the effect of epidermal growth factor receptor amplification on glioblastoma radiation response. Clin. Cancer Res. 12, 2264–2271 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Turcan, S. et al. Efficient induction of differentiation and growth inhibition in IDH1 mutant glioma cells by the DNMT inhibitor decitabine. Oncotarget 4, 1729–1736 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kriegel, A. J. & Liang, M. MicroRNA in situ hybridization for formalin fixed kidney tissues. J. Vis. Exp. 81, 50785 (2013).

    Google Scholar 

  46. Lakins, J. N., Chin, A. R. & Weaver, V. M. Exploring the link between human embryonic stem cell organization and fate using tension-calibrated extracellular matrix functionalized polyacrylamide gels. Methods Mol. Biol. 916, 317–350 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Lu for the B20 mouse experiment with input from G. Bergers; C. Cowdrey and A. Shai for tissue collection support; and I. Acerbi for IRB paperwork. Animal handling and tissue preparation was supported by L. Korets and N. Korets. This work was supported by US National Institutes of Health NCI R01 grants CA138818-01A1, CA192914-01, CA174929-01, and 2R01CA085482-11A1 (V.M.W.), U54 grants CA163155-01 and CA143836-01 (V.M.W.), US National Institutes of Health NCI F31CA180422-01A1 (Y.A.M.), NSF GRFP 1144247 (Y.A.M.), US DOD BCRP grant W81XWH-07-1-0538 (J.K.M.), NINDS R01 NS081117 (J.J.P.), NINDS R21 NS088114 and NBTS (A.I.P.).

Author information

Authors and Affiliations

Authors

Contributions

V.M.W., Y.A.M. and J.K.M. conceived the project. T.R.M., K.L., J.J.P. and Y.A.M. were involved in patient recruitment and human tissue collection. J.J.P. performed human patient sample selection and performed NanoString nCounter gene expression analysis with G.F.R. Y.A.M. and J.M.B. performed mechanical testing. Y.A.M. and J.K.M. performed immunohistological analyses, imaging analyses, and quantification. J.M.B. conducted animal injections. J.M.B., Y.A.M. and J.K.M. monitored mouse colonies, and processed and analysed mouse and human tissues. J.K.M. and Y.A.M. performed all in vitro studies. J.N.L. and Y.A.M. designed and built all shRNA, microRNA and IDH1 expression constructs. M.W.P. performed the bioinformatics analysis. A.I.P. assisted Y.A.M. with primary GBM culture and manipulations. E.C.H. and Y.K. provided the TS603 and TS667 glioma cells. V.M.W., J.K.M. and Y.A.M. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Valerie M. Weaver.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Cellularity, fibrillar collagen and vascularity are independent of ECM stiffness.

(a) Scatter plots of ECM stiffness measured by AFM in a human patient sample analyzed immediately post resection from the operating room (Fresh), post snap freezing and analyzed either within 30 minutes of being thawed in a cocktail of protein inhibitors (Frozen) or treated with either PBS (PBS) or hyaluronidase (HAse) for 60 minutes (6 samples/group were tested for all groups with the exception of the “HAse 60 mins” group where 5 samples were assessed). All samples were pooled per group with 5-6 regions tested per sample (pooled group means indicated with red lines). Group means are indicated in red (6 samples/group were tested for all groups with the exception of the “HAse 60 mins” group where 5 samples were assessed). (b) Immunohistochemistry images of patient tissue immunostained for neuronal (SMI31) or astrocytic (GFAP) processes indicating a switch in the type but not density of the cellular processes in normal versus GBM brains. Scale Bar 70 μm. (c) Correlation between measured ECM stiffness and cell number in the analyzed area revealing no relationship between the two variables (linear and exponential regressions, n = 100 areas (10 patient biopsies with 10 areas/patient), insert: representative image of the AFM method for tissue analysis). Scale Bar 150 μm. (d) Second harmonic generation (SHG) image of fibrillary collagen (purple) and CD31-immunostained vasculature (red) in human GBMs (insert: composite image of SHG and CD31 in a normal human brain). Scale Bar 70 μm. (e) Immunofluorescence images of astrocytic processes (GFAP, red) and vasculature (Collagen 1, green) in human gliotic (top) and GBM (bottom) biopsies. Scale Bar 50 μm. (f) Correlation between measured ECM stiffness and percent vascularity of the tissue (percent area positive for FVIII stain) revealing no relationship between the two variables (linear and exponential regression, n = 10 patients). (g) Distribution of ECM stiffness measured by AFM in two (labeled as mGBM model 1 and mGBM model 2) mouse xenograft models of GBM (n = 4 mice/group), normal mouse brain (n = 3 mice) as well as mGBM model 2 treated with an antiangiogenic agent, avastin (labeled as mGBM model 2 + B20, n = 5 mice) revealing a slight but non-significant reduction in ECM stiffness upon B20-mediated normalization of GBM vasculature in mouse models of GBM. All samples were pooled per group with 5 regions tested per sample (pooled group means indicated with yellow lines).

Supplementary Figure 2 R132H IDH1 expression modifies TNC expression and ECM stiffness.

(a) Immunofluorescence images and quantifications of WT and R132H IDH1 patient tissues immunostained for DAPI(blue), aggrecan (green), and versican (red) (mean ± s.e.m., n = 7 mice/group, ns by unpaired 2-sided t-test). (b) Immunofluorescence images of patient tissues immunostained for the lecticans, aggrecan (green) and versican (red). (c) Immunoblot analysis confirming the R132H and WT IDH1 status in U87 GBM line. (d) Kaplan-Meier graph showing survival of xenograft mice injected orthotopically with either U87 WT IDH1 (blue, n = 7 mice) or U87 R132H IDH1 (red, n = 7 mice) human GBM cells. (e) Histogram showing the distribution of ECM stiffness measured by AFM in xenograft tumors derived from U87 WT (blue) or R132H mutant (red) IDH1 cells (n = 5 mice/group WT IDH1, n = 4 mice/group R132H IDH1, two-sided Kolmogorov-Smirnov test P = 9.7e-4). (f) Immunofluorescence images and quantification of U87 IDH1 WT and R132H mutant xenograft tumors immunostained for pMLC2 (green, top) and pFAK397 (green, bottom) with propidium iodide (PI, red) (mean ± s.e.m., n = 7 mice/group, unpaired 2-sided t-test P = 0.0007). (g) Immunofluorescence images and quantification of TNC (green) with PI (red) in xenograft tumors derived from either WT or R132H IDH1 human primary cells (mean ± s.e.m., n = 7 mice/group, unpaired 2-sided t-test P = 0.0087). (h) Immunoblot analysis confirming lentiviral shRNA construct targeting of TNC (top). Immunofluorecence images of cellular morphology (F-actin in green and DAPI in blue) of scramble shRNA and TNC shRNA cells (bottom). Scale Bar 10 μm. (i) TNC knockdown in mouse xenograft tumors by mRNA (top) and immunofluorescence (bottom) in scramble and TNC shRNA constructs (mean ± s.d., n = 5 mice/group, unpaired 2-sided t-test P = 0.014). Scale Bars 50 μm, unless otherwise indicated.

Supplementary Figure 3 HIF1α and miR-203 regulate TNC expression.

(a) Distribution of ECM stiffness measured by AFM in xenograft tumors arising from primary R132H IDH1 cells in either HIF1α-positive or-negative regions surrounding areas of necrosis (n = 4 mice, two-sided Kolmogorov-Smirnov test P = 3.7e-2). All samples were pooled per group with 5 regions tested per sample (pooled group means indicated with yellow lines). (b) Immunofluorescence images of xenograft tumors derived from cells expressing a control scramble shRNA or an shRNA for TNC shRNA immunostained for HIF1α with PI (red). (c) mRNA expression of HIF1α (left) and TNC(right) in WT IDH1 cells expressing either scramble shRNA or shRNA targeting HIF1α, represented as % of shCNL (mean ± s.e.m., n = 6/group, unpaired 2-sided test P = 0.001). (d) Quantification of HIF1α (left) and TNC(right) protein expression in IDH1 WT cells with and without hypoxia, as indicated (mean ± s.e.m., n = 6/group, one-way ANOVA with Tukey’s multiple comparisons test P = 0.0013). (e) Representative immunoblot of WT and R132H IDH1 primary GBM cells plated on laminin-coated tissue culture plates under normoxic and hypoxic (1% oxygen) conditions probed for TNC and HIF1α. (f) Immunoblot of WT IDH1 primary human GBM cells, expressing either scramble control shRNA or shRNA targeting HIF1α, plated on lamin-coated tissue culture plates under normoxic or hypoxic (1% oxygen) conditions, as indicated, and probed for TNC induction. (g) Immunofluorescence images of xenograft tumors derived from either WT or R132H IDH1 primary human GBM cells immunostained for HIF1α (green) with PI (red); asterisks indicate areas of necrosis. Scale Bars 50 μm.

Supplementary Figure 4 Endogenous and ectopic R132H IDH1 1° GBMs cannot tune HIF1α.

(a) Immunoblots and quantification of primary human glioma cells harboring an endogenous R132H IDH1 mutation (top: TS603, bottom: BT142) plated on soft (140 Pa) or stiff (>6 kPa) substrates under normoxic and hypoxic (1% oxygen) conditions and probed for HIF1α and TNC protein expressions, as well as actin (graph shows means of 2 biological replicate samples per group). (b) Immunoblots and quantification of WT IDH1 primary human glioma cells (TS667) plated on soft (140 Pa) or stiff (>6 kPa) substrates under normoxic and hypoxic (1% oxygen) conditions and probed for HIF1α and TNC protein expressions, as well as actin (graph shows means of 2 biological replicate samples per group,). (c) Immunoblot of R132H IDH1 primary human GBM cells expressing either vector control or constitutively active HIF1α (CA-HIF1α) plated on soft (140 Pa) gels under normoxic and hypoxic (1% oxygen) conditions, as indicated, and probed for HIF1α protein expression, as well as HIF1α -driven protein expressions of TNC and LOXL2.

Supplementary Figure 5 miR-203 targets the HIF1α and TNC 3’ UTRs.

(a) Diagram of miR-203 targeting seed sequences located in 3’-untranslated regions (3’ UTRs) of human HIF1α and TNC mRNAs. (b) Quantification of miR-203 expression after RNA immunoprecipitation (RIP) using AGO2 as a bait validates that antagomiR-mediated targeting of miR-203 reduces miR-203 loading into the RISC (graph shows mean of 2 IP lysates per group). (c) Immunoblots for TNC, HIF1α, LOXL2 (as HIF1α target gene) and actin from primary human GBM cells expressing R132H IDH1 (± an antagomiR control or an antagomiR targeting miR-203, ±1% hypoxia). (d) Quantification of HIF1α and TNC protein expressions in primary human GBM cells expressing R132H IDH1 (± an antagomiR control or an antagomiR targeting miR-203, ±1% hypoxia) (means ± s.e.m., n = 3 lysates/group, One-way ANOVA with Tukey’s multiple comparisons test P = 0.0011).

Supplementary Figure 6 Regional assessment of ECM stiffness and mechanosignaling.

(a) Maps of ECM stiffness are obtained using an AFM tip where a 5 μm diameter bead is attached to the end of a cantilever. The AFM tip was brought down to indent the sample, during which the motion of the z-piezo and the applied force were recorded. The ECM stiffness was computed from the displacement of the z-piezo minus the bending of the cantilever. The number of ECM stiffness measurements made was operator-determined by the x- and y-step sizes input for a 90 μm by 90 μm grid. The example shown demonstrates how step sizes of 14 (in the x- and y-directions) result in 196 independent mechanical measurements. (b) For regionally-matched correlations between ECM stiffness and mechanosignaling, patient (and xenograft) tissues were immunostained for TNC (green) and pFAK (red). Multiple specific, patient-representative regions (4-10 regions per tumor) were identified (with the x- and y-coordinates noted and images acquired) for further AFM testing in a serial section. For subsequent AFM testing, after patient (and xenograft) tumor sections were secured for testing, the identified testing region was located via x- and y-coordinates using the optical microscope. (c) Left: Immunohistochemistry images of patient tumors immunostained for TNC (green) and pFAK (red) with DAPI (blue) with regions for AFM analysis in serial sections denoted by white boxes. Graphs illustrate individual patient correlations between TNC and pFAK. Right: AFM maps of patient tumors. Graphs illustrate individual patient correlations between elastic modulus (stiffness) and pFAK. (d) The data underlying (c left) was combined into graphs depicting regional matching of ECM stiffness and pFAK (left) and TNC and pFAK (right) to illustrate not only the correlations between the variables, but also the variability between patients within the cohort (n = 3 patient samples/group, One-way ANOVA with Tukey’s comparison P = 0.0011). (e) The data underlying (c right) was combined into a graph depicting both the patient-specific trends in elastic modulus (stiffness) and the patient variability within the cohort (n = 10 patients, 4 regions/patient sample, linear regression analysis yielding p < 0.0001 for both plots with R2 = 0.4812 for pFAK:stiffness data and R2 = 0.5850 for pFAK:TNC data). Scale Bars 200 μm (left panels) and 20 μm (right panels), as indicated.

Supplementary Figure 7 Full scans of key blots.

Supplementary information

Supplementary Information

Supplementary Information (PDF 902 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miroshnikova, Y., Mouw, J., Barnes, J. et al. Tissue mechanics promote IDH1-dependent HIF1α–tenascin C feedback to regulate glioblastoma aggression. Nat Cell Biol 18, 1336–1345 (2016). https://doi.org/10.1038/ncb3429

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3429

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer