Solid stress in brain tumours causes neuronal loss and neurological dysfunction and can be reversed by lithium

Giorgio Seano, Hadi T Nia, Kyrre E Emblem, Meenal Datta, Jun Ren, Shanmugarajan Krishnan, Jonas Kloepper, Marco C Pinho, William W Ho, Mitrajit Ghosh, Vasileios Askoxylakis, Gino B Ferraro, Lars Riedemann, Elizabeth R Gerstner, Tracy T Batchelor, Patrick Y Wen, Nancy U Lin, Alan J Grodzinsky, Dai Fukumura, Peigen Huang, James W Baish, Timothy P Padera, Lance L Munn, Rakesh K Jain, Giorgio Seano, Hadi T Nia, Kyrre E Emblem, Meenal Datta, Jun Ren, Shanmugarajan Krishnan, Jonas Kloepper, Marco C Pinho, William W Ho, Mitrajit Ghosh, Vasileios Askoxylakis, Gino B Ferraro, Lars Riedemann, Elizabeth R Gerstner, Tracy T Batchelor, Patrick Y Wen, Nancy U Lin, Alan J Grodzinsky, Dai Fukumura, Peigen Huang, James W Baish, Timothy P Padera, Lance L Munn, Rakesh K Jain

Abstract

The compression of brain tissue by a tumour mass is believed to be a major cause of the clinical symptoms seen in patients with brain cancer. However, the biological consequences of these physical stresses on brain tissue are unknown. Here, via imaging studies in patients and by using mouse models of human brain tumours, we show that a subgroup of primary and metastatic brain tumours, classified as nodular on the basis of their growth pattern, exert solid stress on the surrounding brain tissue, causing a decrease in local vascular perfusion as well as neuronal death and impaired function. We demonstrate a causal link between solid stress and neurological dysfunction by applying and removing cerebral compression, which respectively mimic the mechanics of tumour growth and of surgical resection. We also show that, in mice, treatment with lithium reduces solid-stress-induced neuronal death and improves motor coordination. Our findings indicate that brain-tumour-generated solid stress impairs neurological function in patients, and that lithium as a therapeutic intervention could counter these effects.

Figures

Fig. 1.. Measuring how nodular tumors mechanically…
Fig. 1.. Measuring how nodular tumors mechanically affect the surrounding brain.
(a-c) Solid stress in mouse models of nodular (patient-derived U87 cell line) and infiltrative brain tumors (patient-derived MGG8 cell line) in nude mice. (a) High-resolution ultrasound imaging of the stress-induced deformation and representative stress profiles across the tumour diameter and the normal surrounding tissue. Scale bar: 1 mm. (b-c) Estimation of the circumferential (compression; σθθ) and radial (tension; σrr) stress in the surrounding tissue obtained from mathematical model (described in Figure S1b-d and Method section). Data are mean of three independent tumors ± s.e.m. (d) Micro-anatomic deformation of the brain tissue around nodular tumors. IHC of U87 and MGG8 tumors at the interface of the normal brain tissue. Neurons (NeuN staining) and vessels (Collagen IV staining). Arrows indicate the deformed region around the tumour, neurons are packed and vessels are displaced following the circumference of the tumour margin. Representative images from a cohort of 10 mice with tumour. Scale bar: 50 μm. (e) Representative T2W and FLAIR MRI of pre-surgery pre-treatment patients with perfectly-defined margins GBM (nodular) and ill-defined GBM (infiltrative). (f) Karnofsky performance score (KPS) histograms of patients with perfectly-defined margins (nodular) or ill-defined GBM (infiltrative). Cohort of 64 pre-surgery pre-treatment GBM patients.
Fig. 2.. Reduced vessel perfusion in the…
Fig. 2.. Reduced vessel perfusion in the brain tissue around nodular tumors.
(a) Longitudinal OCT intravital angiography (perfused vessels) of the nodular Gl261 mouse model. U87 and BT474 are presented in Fig. S3B, D. Scale bar: 1 mm. (b) Longitudinal quantitative analysis of the vascular perfused volume fraction in the surrounding tissue. GBM U87: Day17 p=0.025; Day18 p=0.011; Day19 p=0.045; day20 p=0.037 vs the red-dotted time-point. BC BT474: Day14 p=0.001; Day18 p=0.024; Day21 p=0.003; day25 p=0.005; day28 p=0.001; day32 p<0.001; day38 p=0.035 vs the red-dotted time-point. Data are mean ± s.e.m. (c) Intravital longitudinal multi-photon imaging depicting changes in vessel diameter in the nodular Gl261-GFP and in the infiltrative MGG8-GFP GBM mouse models. GFP (green, tumour cells) and TAMRA-Dextran (red, 2MDa-dextran, blood flow). VA = vascular area. Scale bar: 20 μm.
Fig. 3.. Neuronal deformation and death in…
Fig. 3.. Neuronal deformation and death in the brain tissue around nodular tumors.
(a) Intravital multi-photon imaging of brain nuclei around the nodular mouse Gl261-DsRed GBM implanted in H2B-EGFP mouse (CAG::H2B-EGFP C57/BL6). GFP (green, nuclei within the brain tissue), DsRed (red, tumour cells) and BlueCascade (blue, 2MDa-dextran, blood flow). Insets are magnifications of the yellow squares. Vascular area (VA) is the quantification (%) of these exact images. Scale bar: 20 μm. (b) IHC of neuronal nuclei (NeuN) in the surrounding brain tissue of mice with the nodular U87 tumour. Representative image from a cohort of 10 mice with tumour. Insets are magnifications of the red squares. Scale bar: 50 μm. (c) Quantification of neuronal nuclear area in the brain tissue around nodular or infiltrative tumors. Data are mean ± s.e.m. p-values are vs “300–450 from the tumour edge”. (d) Apoptosis (ApopTag) at the interface between the nodular U87 tumour and the normal brain tissue. Representative image from a cohort of 10 mice with tumour. Inset is magnification of the red square. Scale bar: 100 μm.
Fig. 4.. Reduced perfusion in the surrounding…
Fig. 4.. Reduced perfusion in the surrounding normal tissues in GBM and BC brain met patients.
(a) Quantification of the local perfusion in surrounding normal brain tissue from immediately adjacent to the peri-edematous region of the GBM to the region 12 mm away from the tumor edge. The cohort of 64 pre-surgery GBM patients was divided in patients with a reduced perfusion in the surrounding tissue (“reduced perf”) and patients with no difference in perfusion (“unchanged perf”). Data are mean ± s.e.m. Point 1.72 p<0.001 vs “unchanged perf”; point 3.44 p<0.001; point 5.15 p=0.001; point 6.87 p=0.014; point 8.59 p=0.019. (b) Representative pre-surgery GBM patient from a cohort of 33 patients with local reduced perfusion and fiber tensor signal in the surrounding tissue as well as midline shift. (c) Histograms of KPS in “reduced perf” and “unchanged perf” GBM patients. (d) CE-T1 and FLAIR-T2 volumes in the two classes with “reduced perf.” or “unchanged perf.” in the surrounding brain tissue. N.S., not significant. Data are mean ± s.e.m (N = 64 patients). (e) Classification of the patients in the two perfusion subclasses in based on the edematous (FLAIR-T2) MRI margins (N = 64 patients). Comparable result with CE-T1 MRI, Fig. S6B. (f) Quantification of the local perfusion in the brain tissue immediately surrounding the BC brain mets. Cohort of 34 BC patients (26 HER2-positive; 8 HER2-negative). Data are mean ± s.e.m. Point 0 p<0.001 vs point 1.2; point 1.2 p<0.001 vs point 2.4; point 3.6 p=0.010 vs point 4.8; point 4.8 p=0.013 vs point 6. (g) Representative BC HER2-negative patient from a cohort of 34 patients with local reduction of perfusion and fiber tensor signal in the surrounding tissue.
Fig. 5.. Direct chronic compression of brain…
Fig. 5.. Direct chronic compression of brain reduces vessel perfusion and induces neuronal damage.
(a) Schematic of the intravital compression device procedure. (b) Deformation of the compressed tissue. Hematoxylin-eosin (H&E) of the whole brain gradually and chronically compressed for 14 days. Representative image from a cohort of 7 mice with compression device. Insets are magnifications of the white squares. Scale bar: 1 mm. (c) OCT longitudinal intravital angiography of the brain seen from the compression apparatus (average projection of the 3D imaging). Vascular area (VA) is the quantification (%) of the shown images. Scale bar: 1 mm. (d) Ultrastructural imaging of nuclei in uncompressed and compressed cortexes via electromicroscopy. Representative images from a cohort of 3 mice per experimental point. Scale bar: 1 μm. Quantification of the percentage of condensed chromatin area in n nuclei. Data are mean ± s.e.m. (e-f) NeuN (neuron nuclei) in the compressed cortex. Representative images from a cohort of 4 mice per experimental point and quantification of number of neurons (NeuN+ nuclei) per cortex exposed to the cranial window. Data are mean ± s.e.m. Scale bar: 100 μm. (g) Quantification of neuronal nuclear area in uncompressed or compressed cortexes. Normalized results. Data are mean ± s.e.m. (h) Rotarod endurance (index of motor coordination and balance) after 14 days of compression of the motor and somatosensory cortex. Data are mean of 3 consecutive days (1 measure/day) and 5 mice per group ± s.e.m. (i) Static gait test (index of locomotion); footprint analysis of the stride length. Footprint in Fig. S8E. Data are mean of 3 technical replicates and n mice per group ± s.e.m.
Fig. 6.. Decompression restores vessel perfusion and…
Fig. 6.. Decompression restores vessel perfusion and partially rescues motor-coordination.
(a) OCT angiography after decompression of brains with nodular U87 tumors by removing cranial window (craniotomy). Complete time-course in Figure S9B-C. (b) Schematic of the long-term compression/decompression timeline via the in vivo chronic apparatus. (c) Longitudinal OCT angiography before and after decompression. Images of a representative mouse from a cohort of 3 mice. Insets are magnifications of the red squares. See Movie S7 for the complete time-lapse. Scale bar: 1 mm. (d) Representative cortexes stained with anti-NeuN from a cohort of 3 mice. The long-term compression induces tissue loss and the decompression partly restores the micro-anatomy of the cortex. Insets are magnifications of the red squares. Scale bar: 500 μm. (e) Quantification of the NeuN+ neurons in the cortexes, decompressed or compressed and released. Data are mean ± s.e.m. (f) Rotarod endurance (index of motor coordination and balance) 24–48 hours after decompression of the motor and somatosensory cortex. Data are mean of 2 consecutive days and 12 mice per group ± s.e.m (normalized with the value before decompression).
Fig. 7.. Systemic treatment with lithium alleviates…
Fig. 7.. Systemic treatment with lithium alleviates compression-mediated neuronal damage.
(a) Schematic of the treatments in long-term compression/decompression procedure. (b) Representative cortexes stained with anti-NeuN from a cohort of 5 mice per experimental point. Chronic lithium treatment preserves the cortex micro-anatomy after decompression. Insets are magnifications of the red squares. Scale bar: 500 μm. (c) Quantification of the NeuN+ neurons in the cortexes, decompressed or compressed and released. Data are mean ± s.e.m. (d) Ultrastructural imaging of nuclei in compressed cortexes treated with vehicle or lithium via electromicroscopy. Representative images from a cohort of 3 mice per experimental point. Scale bar: 1 μm. Quantification of the percentage of condensed chromatin area in n nuclei. Data are mean ± s.e.m. (e) Longitudinal analysis of the Rotarod endurance (index of motor coordination and balance). Data are mean of 2 consecutive days and 6 mice per group ± s.e.m. (f) Static gait test (index of locomotion); footprint analysis of the stride length at the time of highest compression (day 14). Data are mean of 3 technical replicates and n mice per group ± s.e.m. 115 steps measured for vehicle-treated mice and 62 for lithium-treated ones.
Fig. 8.. Solid stress from nodular brain…
Fig. 8.. Solid stress from nodular brain tumors compresses the surrounding brain tissue, thus causing neurological dysfunction.
(a) Heatmap illustrating normalized gene expression of differentially expressed genes (FDR<0.05) from RNA-Seq analysis of compressed/released cortexes under chronic lithium treatment. (b) Quantification of the NeuN+ neurons in the cortexes, compressed and released lithium-treated with treatment start when compression and symptoms were already present. Data are mean ± s.e.m. (c) Schematic summarizing the effects of nodular tumors and their resulting solid stress on the surrounding brain tissue. The focus of the manuscript is on nodular tumors and the effects of solid stress on the surrounding tissue, while infiltrative tumors appeared to exert lower solid stress to the brain. (d) Schematic summarizing the causes, modifiers and consequences of solid stress from nodular tumors.

References

    1. Goriely A et al. Mechanics of the brain: perspectives, challenges, and opportunities. Biomech Model Mechanobiol 14, 931–965, doi:10.1007/s10237-015-0662-4 (2015).
    1. Helmlinger G, Netti PA, Lichtenbeld HC, Melder RJ & Jain RK Solid stress inhibits the growth of multicellular tumor spheroids. Nature Biotechnology 15, 778–783, doi:10.1038/nbt0897-778 (1997).
    1. Stylianopoulos T et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc Natl Acad Sci U S A 109, 15101–15108, doi:10.1073/pnas.1213353109 (2012).
    1. Nia HT et al. Solid stress and elastic energy as measures of tumour mechanopathology. Nat Biomed Eng 1, doi:10.1038/s41551-016-0004 (2016).
    1. Jain RK, Martin JD & Stylianopoulos T The role of mechanical forces in tumor growth and therapy. Annual Review of Biomedical Engineering 16, 321 (2014).
    1. Chauhan VP et al. Compression of pancreatic tumor blood vessels by hyaluronan is caused by solid stress and not interstitial fluid pressure. Cancer Cell 26, 14–15, doi:10.1016/j.ccr.2014.06.003 (2014).
    1. Stylianopoulos T et al. Coevolution of solid stress and interstitial fluid pressure in tumors during progression: implications for vascular collapse. Cancer Res 73, 3833–3841, doi:10.1158/0008-5472.CAN-12-4521 (2013).
    1. Gamburg ES et al. The prognostic significance of midline shift at presentation on survival in patients with glioblastoma multiforme. Int J Radiat Oncol Biol Phys 48, 1359–1362 (2000).
    1. Nia HT et al. Quantifying solid stress and elastic energy from excised or in situ tumors. Nat Protoc 13, 1091–1105, doi:10.1038/nprot.2018.020 (2018).
    1. Mazurowski MA, Desjardins A & Malof JM Imaging descriptors improve the predictive power of survival models for glioblastoma patients. Neuro Oncol 15, 1389–1394, doi:10.1093/neuonc/nos335 (2013).
    1. Chambless LB et al. The relative value of postoperative versus preoperative Karnofsky Performance Scale scores as a predictor of survival after surgical resection of glioblastoma multiforme. J Neurooncol 121, 359–364, doi:10.1007/s11060-014-1640-x (2015).
    1. Chauhan VP et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun 4, 2516, doi:10.1038/ncomms3516 (2013).
    1. Vakoc BJ et al. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat Med 15, 1219–1223, doi:10.1038/nm.1971 (2009).
    1. Bar-Kochba E, Scimone MT, Estrada JB & Franck C Strain and rate-dependent neuronal injury in a 3D in vitro compression model of traumatic brain injury. Sci Rep 6, 30550, doi:10.1038/srep30550 (2016).
    1. Arundine M, Aarts M, Lau A & Tymianski M Vulnerability of central neurons to secondary insults after in vitro mechanical stretch. J Neurosci 24, 8106–8123, doi:10.1523/JNEUROSCI.1362-04.2004 (2004).
    1. Field AS et al. Diffusion tensor eigenvector directional color imaging patterns in the evaluation of cerebral white matter tracts altered by tumor. J Magn Reson Imaging 20, 555–562, doi:10.1002/jmri.20169 (2004).
    1. Blanchet L et al. Discrimination between metastasis and glioblastoma multiforme based on morphometric analysis of MR images. AJNR Am J Neuroradiol 32, 67–73, doi:10.3174/ajnr.A2269 (2011).
    1. Takano T et al. Glutamate release promotes growth of malignant gliomas. Nat Med 7, 1010–1015, doi:10.1038/nm0901-1010 (2001).
    1. Balkaya M, Krober JM, Rex A & Endres M Assessing post-stroke behavior in mouse models of focal ischemia. J Cereb Blood Flow Metab 33, 330–338, doi:10.1038/jcbfm.2012.185 (2013).
    1. Roth L et al. Impaired gait pattern as a sensitive tool to assess hypoxic brain damage in a novel mouse model of atherosclerotic plaque rupture. Physiol Behav 139, 397–402, doi:10.1016/j.physbeh.2014.11.047 (2015).
    1. Budday S et al. Mechanical characterization of human brain tissue. Acta Biomater 48, 319–340, doi:10.1016/j.actbio.2016.10.036 (2017).
    1. Budday S et al. Mechanical properties of gray and white matter brain tissue by indentation. J Mech Behav Biomed Mater 46, 318–330, doi:10.1016/j.jmbbm.2015.02.024 (2015).
    1. Johnson CL et al. Local mechanical properties of white matter structures in the human brain. Neuroimage 79, 145–152, doi:10.1016/j.neuroimage.2013.04.089 (2013).
    1. Ohue S et al. Evaluation of intraoperative brain shift using an ultrasound-linked navigation system for brain tumor surgery. Neurol Med Chir (Tokyo) 50, 291–300 (2010).
    1. Paul DA et al. White matter changes linked to visual recovery after nerve decompression. Sci Transl Med 6, 266ra173, doi:10.1126/scitranslmed.3010798 (2014).
    1. Yang SH et al. Nec-1 alleviates cognitive impairment with reduction of Abeta and tau abnormalities in APP/PS1 mice. EMBO Mol Med 9, 61–77, doi:10.15252/emmm.201606566 (2017).
    1. Dash PK et al. Valproate administered after traumatic brain injury provides neuroprotection and improves cognitive function in rats. PLoS One 5, e11383, doi:10.1371/journal.pone.0011383 (2010).
    1. Makoukji J et al. Lithium enhances remyelination of peripheral nerves. Proc Natl Acad Sci U S A 109, 3973–3978, doi:10.1073/pnas.1121367109 (2012).
    1. Rocksen D, Lilliehook B, Larsson R, Johansson T & Bucht A Differential anti-inflammatory and anti-oxidative effects of dexamethasone and N-acetylcysteine in endotoxin-induced lung inflammation. Clin Exp Immunol 122, 249–256 (2000).
    1. Capasso A, Di Giannuario A, Loizzo A, Pieretti S & Sorrentino L Dexamethasone pretreatment reduces the psychomotor stimulant effects induced by cocaine and amphetamine in mice. Prog Neuropsychopharmacol Biol Psychiatry 19, 1063–1079 (1995).
    1. Forester BP et al. Brain lithium levels and effects on cognition and mood in geriatric bipolar disorder: a lithium-7 magnetic resonance spectroscopy study. Am J Geriatr Psychiatry 17, 13–23, doi:10.1097/JGP.0b013e318172b3d0 (2009).
    1. Phiel CJ et al. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 276, 36734–36741, doi:10.1074/jbc.M101287200 (2001).
    1. Jagtap PG et al. Structure-activity relationship study of tricyclic necroptosis inhibitors. J Med Chem 50, 1886–1895, doi:10.1021/jm061016o (2007).
    1. Huang da W, Sherman BT & Lempicki RA Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4, 44–57, doi:10.1038/nprot.2008.211 (2009).
    1. Can A et al. Antidepressant-like responses to lithium in genetically diverse mouse strains. Genes Brain Behav 10, 434–443, doi:10.1111/j.1601-183X.2011.00682.x (2011).
    1. Moore GJ, Bebchuk JM, Wilds IB, Chen G & Manji HK Lithium-induced increase in human brain grey matter. Lancet 356, 1241–1242 (2000).
    1. Foland LC et al. Increased volume of the amygdala and hippocampus in bipolar patients treated with lithium. Neuroreport 19, 221–224, doi:10.1097/WNR.0b013e3282f48108 (2008).
    1. Rowe MK & Chuang DM Lithium neuroprotection: molecular mechanisms and clinical implications. Expert Rev Mol Med 6, 1–18, doi:10.1017/S1462399404008385 (2004).
    1. Yazlovitskaya EM et al. Lithium treatment prevents neurocognitive deficit resulting from cranial irradiation. Cancer Res 66, 11179–11186, doi:10.1158/0008-5472.CAN-06-2740 (2006).
    1. Yu F et al. Lithium ameliorates neurodegeneration, suppresses neuroinflammation, and improves behavioral performance in a mouse model of traumatic brain injury. J Neurotrauma 29, 362–374, doi:10.1089/neu.2011.1942 (2012).
    1. Li Q et al. Lithium reduces apoptosis and autophagy after neonatal hypoxia-ischemia. Cell Death Dis 1, e56, doi:10.1038/cddis.2010.33 (2010).
    1. Han S et al. Lithium enhances the antitumour effect of temozolomide against TP53 wild-type glioblastoma cells via NFAT1/FasL signalling. Br J Cancer 116, 1302–1311, doi:10.1038/bjc.2017.89 (2017).
    1. Korur S et al. GSK3beta regulates differentiation and growth arrest in glioblastoma. PLoS One 4, e7443, doi:10.1371/journal.pone.0007443 (2009).
    1. Nowicki MO et al. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro Oncol 10, 690–699, doi:10.1215/15228517-2008-041 (2008).
    1. Meyers CA & Brown PD Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol 24, 1305–1309, doi:10.1200/JCO.2005.04.6086 (2006).
    1. Wakimoto H et al. Maintenance of primary tumor phenotype and genotype in glioblastoma stem cells. Neuro Oncol 14, 132–144, doi:10.1093/neuonc/nor195 (2012).
    1. Griveau A et al. A Glial Signature and Wnt7 Signaling Regulate Glioma-Vascular Interactions and Tumor Microenvironment. Cancer Cell 33, 874–889 e877, doi:10.1016/j.ccell.2018.03.020 (2018).
    1. Kloepper J et al. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival. Proc Natl Acad Sci U S A 113, 4476–4481, doi:10.1073/pnas.1525360113 (2016).
    1. Peterson TE et al. Dual inhibition of Ang-2 and VEGF receptors normalizes tumor vasculature and prolongs survival in glioblastoma by altering macrophages. Proc Natl Acad Sci U S A 113, 4470–4475, doi:10.1073/pnas.1525349113 (2016).
    1. Askoxylakis V et al. Preclinical Efficacy of Ado-trastuzumab Emtansine in the Brain Microenvironment. J Natl Cancer Inst 108, doi:10.1093/jnci/djv313 (2016).
    1. Nia HT, Han L, Li Y, Ortiz C & Grodzinsky A Poroelasticity of Cartilage at the Nanoscale. Biophys J 101, 2304–2313, doi:DOI10.1016/j.bpj.2011.09.011 (2011).
    1. Netti PA, Berk DA, Swartz MA, Grodzinsky AJ & Jain RK Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer research 60, 2497–2503 (2000).
    1. Kiviranta P et al. Collagen network primarily controls Poisson’s ratio of bovine articular cartilage in compression. J Orthop Res 24, 690–699, doi:10.1002/jor.20107 (2006).
    1. Buschmann MD et al. Stimulation of aggrecan synthesis in cartilage explants by cyclic loading is localized to regions of high interstitial fluid flow. Arch Biochem Biophys 366, 1–7 (1999).
    1. Jamin Y et al. Exploring the biomechanical properties of brain malignancies and their pathologic determinants in vivo with magnetic resonance elastography. Cancer Res 75, 1216–1224, doi:10.1158/0008-5472.CAN-14-1997 (2015).
    1. Arani A et al. Acute pressure changes in the brain are correlated with MR elastography stiffness measurements: initial feasibility in an in vivo large animal model. Magnetic resonance in medicine 79, 1043–1051 (2018).
    1. Weickenmeier J et al. Brain stiffens post mortem. Journal of the mechanical behavior of biomedical materials 84, 88–98 (2018).
    1. Pogoda K et al. Compression stiffening of brain and its effect on mechanosensing by glioma cells. New journal of physics 16, 075002 (2014).
    1. Storm C, Pastore JJ, MacKintosh FC, Lubensky TC & Janmey PA Nonlinear elasticity in biological gels. Nature 435, 191 (2005).
    1. Timoshenko S & Goodier J Theory of elasticity. 1951. New York 412, 108.
    1. Mihai LA, Chin L, Janmey PA & Goriely A A comparison of hyperelastic constitutive models applicable to brain and fat tissues. Journal of The Royal Society Interface 12, 20150486 (2015).
    1. Lang GE, Stewart PS, Vella D, Waters SL & Goriely A Is the Donnan effect sufficient to explain swelling in brain tissue slices? Journal of The Royal Society Interface 11, 20140123 (2014).
    1. Grodzinsky AJ Fields, Forces, and Flows in Biological Systems. (Garland Science, 2011).
    1. Ager EI et al. Blockade of MMP14 activity in murine breast carcinomas: implications for macrophages, vessels, and radiotherapy. J Natl Cancer Inst 107, doi:10.1093/jnci/djv017 (2015).
    1. Emblem KE et al. A generic support vector machine model for preoperative glioma survival associations. Radiology 275, 228–234, doi:10.1148/radiol.14140770 (2015).
    1. Pinho MC et al. Low incidence of pseudoprogression by imaging in newly diagnosed glioblastoma patients treated with cediranib in combination with chemoradiation. Oncologist 19, 75–81, doi:10.1634/theoncologist.2013-0101 (2014).
    1. Sorensen AG et al. Comparison of diameter and perimeter methods for tumor volume calculation. J Clin Oncol 19, 551–557, doi:10.1200/JCO.2001.19.2.551 (2001).
    1. Oh J et al. Quantitative apparent diffusion coefficients and T2 relaxation times in characterizing contrast enhancing brain tumors and regions of peritumoral edema. J Magn Reson Imaging 21, 701–708, doi:10.1002/jmri.20335 (2005).
    1. Basser PJ & Pierpaoli C Microstructural and physiological features of tissues elucidated by quantitative-diffusion-tensor MRI. J Magn Reson B 111, 209–219 (1996).
    1. Bjornerud A & Emblem KE A fully automated method for quantitative cerebral hemodynamic analysis using DSC-MRI. J Cereb Blood Flow Metab 30, 1066–1078, doi:10.1038/jcbfm.2010.4 (2010).
    1. Emblem KE & Bjornerud A An automatic procedure for normalization of cerebral blood volume maps in dynamic susceptibility contrast-based glioma imaging. AJNR Am J Neuroradiol 30, 1929–1932, doi:10.3174/ajnr.A1680 (2009).
    1. Emblem KE, Due-Tonnessen P, Hald JK & Bjornerud A Automatic vessel removal in gliomas from dynamic susceptibility contrast imaging. Magn Reson Med 61, 1210–1217, doi:10.1002/mrm.21944 (2009).
    1. Ren J et al. Protein kinase C-delta (PKCdelta) regulates proinflammatory chemokine expression through cytosolic interaction with the NF-kappaB subunit p65 in vascular smooth muscle cells. J Biol Chem 289, 9013–9026, doi:10.1074/jbc.M113.515957 (2014).
    1. Vitner EB et al. RIPK3 as a potential therapeutic target for Gaucher’s disease. Nat Med 20, 204–208, doi:10.1038/nm.3449 (2014).
    1. Gao X et al. Anti-VEGF treatment improves neurological function and augments radiation response in NF2 schwannoma model. Proc Natl Acad Sci U S A 112, 14676–14681, doi:10.1073/pnas.1512570112 (2015).
    1. Dobin A et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21, doi:10.1093/bioinformatics/bts635 (2013).
    1. Lawrence M et al. Software for computing and annotating genomic ranges. PLoS Comput Biol 9, e1003118, doi:10.1371/journal.pcbi.1003118 (2013).
    1. Love MI, Huber W & Anders S Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550, doi:10.1186/s13059-014-0550-8 (2014).

Source: PubMed

3
Sottoscrivi