リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Investigation of the mechanism for emergence of spatial heterogeneity of invading glioblastoma cells in the presence of perivascular factors (本文)」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Investigation of the mechanism for emergence of spatial heterogeneity of invading glioblastoma cells in the presence of perivascular factors (本文)

長南, 友太 慶應義塾大学

2022.03.23

概要

Glioblastoma (GBM) is the most lethal and common type of primary brain tumor in adults. The poor prognosis of GBM patients is attributed to tumor cell invasion into normal brain tissues along blood vessels, which limits efficacy of surgical resection. During invasion, GBM cells display spatial heterogeneity which comprises glioma stem cells at the invasion front and differentiated glioma cells (DGCs) in a tumor core. However, the mechanism for emergence of spatial heterogeneity of GBM cells during invasion toward blood vessels remains unknown. Furthermore, since perivascular factors, such as vascular endothelial cells (ECs) and interstitial flow, have been shown to regulate invasion and differentiation of GBM cells, it is speculated that these factors play a key role in generating spatial heterogeneity of GBM cells during invasion toward blood vessels. Therefore, this thesis aims to investigate the mechanism for emergence of the spatial heterogeneity of GBM cells in the presence of perivascular factors.

Chapter 1 summarizes the background and objectives of this study.

Chapter 2 summarizes the general materials and methods of this study.

Chapter 3 describes the effect of ECs on the spatial heterogeneity of GBM cells in a microfluidic device. Invasion of glioma initiating cells (GICs) into a collagen gel was promoted in the presence of human umbilical vein endothelial cells (HUVECs). During the invasion, GICs with stemness were observed at the invasion front, while DGCs trailed the preceding GICs. Furthermore, differentiation of GICs in a non-invasive region was promoted in the presence of HUVECs.

Chapter 4 describes the effect of intercellular communication among GICs on the spatial heterogeneity of GBM cells. GICs at the invasion front maintained stemness due to low cell density, while trailing cells at high cell density displayed differentiation via local accumulation of paracrine factors they produced. Furthermore, interstitial flow, a component of a perivascular space, suppressed the differentiation of trailing GICs due to the clearance of paracrine factors.

Chapter 5 describes the key signaling pathways which contribute to emergence of the spatial heterogeneity of GBM cells. Differentiation of trailing GICs was induced by paracrine signaling of TGF-β family, while GIC invasion was regulated by p38 and ERK signaling pathways. Furthermore, p38 activity was higher in GICs at the invasion front compared to trailing cells.

Chapter 6 summarizes the results of this study and future perspectives.

この論文で使われている画像

参考文献

Abbott, A. Biology’s new dimension. Nature, 424, 870–872, 2003

Aizawa, T. et al. Neural stem cell-like gene expression in a mouse ependymoma cell line transformed by human BK polyomavirus. Cancer Sci, 102, 122–129, 2011

Albro, M. B. et al. Accumulation of exogenous activated TGF-β in the superficial zoneof articular cartilage. Biophys J, 104, 1794–1804, 2013

Arima, Y. et al. Targeting of cancer stem cells by differentiation therapy. Cancer Sci, 111, 2689–2695, 2020

Arranz, L. et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature, 512, 78–81, 2014

Ayuso, J. M. et al. Glioblastoma on a microfluidic chip: Generating pseudopalisades and enhancing aggressiveness through blood vessel obstruction events. Neuro Oncol, 19, 503–513, 2017

Baker, G. J. et al. Mechanisms of Glioma Formation: Iterative Perivascular Glioma Growth and Invasion Leads to Tumor Progression, VEGF-Independent Vascularization, and Resistance to Antiangiogenic Therapy. Neoplasia (United States), 16, 543–561, 2014

Baker, G. J. et al. Mechanisms of glioma formation: iterative perivascular glioma growth and invasion leads to tumor progression, VEGF-independent vascularization, and resistance to antiangiogenic therapy. Neoplasia, 16, 543–61,2014

Bao, S. et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature, 444, 756–760, 2006

Barczyk, M. et al. Integrins. Cell Tissue Res, 339, 269–280, 2010

Barnes, J. M. et al. A tension-mediated glycocalyx–integrin feedback loop promotes mesenchymal-like glioblastoma. Nat Cell Biol, 20, 1203–1214, 2018

Bastola, S. et al. Glioma-initiating cells at tumor edge gain signals from tumor core cells to promote their malignancy. Nat Commun, 11, 4660, 2020

Beadle, C. et al. The role of myosin II in glioma invasion of the brain. Mol Biol Cell, 19, 3357–68, 2008

Bi, J. et al. Altered cellular metabolism in gliomas — an emerging landscape of actionable co-dependency targets. Nat Rev Cancer, 20, 57–70, 2020

Bonnet, D. et al. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med, 3, 730–7, 1997

Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell, 155, 462, 2013

Cain, R. J. et al. Phosphoinositide 3-kinases in cell migration. Biol Cell, 101, 13–29, 2009

Calabrese, C. et al. A perivascular niche for brain tumor stem cells. Cancer Cell, 11, 69–82, 2007

Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature, 455, 1061–8, 2008

Chen, J. et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature, 488, 522–526, 2012

Cheng, L. et al. Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell, 153, 139–152, 2013

Cheng, L. et al. Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun, 406, 643–648, 2011

Ciasca, G. et al. Nano-mechanical signature of brain tumours. Nanoscale, 8, 19629– 19643, 2016

Clarke, M. F. et al. Cancer stem cells - Perspectives on current status and future directions: AACR workshop on cancer stem cells. in Cancer Research vol. 66 9339–9344, (2006).

Crampton, S. J. et al. Exposure of foetal neural progenitor cells to IL-1β impairs their proliferation and alters their differentiation - a role for maternal inflammation? J Neurochem, 120, 964–73, 2012

Cuddapah, V. A. et al. A neurocentric perspective on glioma invasion. Nat Rev Neurosci, 15, 455–465, 2014

David, C. J. et al. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol, 19, 419–435, 2018

Denais, C. M. et al. Nuclear envelope rupture and repair during cancer cell migration. Science (80- ), 352, 353–358, 2016

Ebben, J. D. et al. The cancer stem cell paradigm: A new understanding of tumor development and treatment. Expert Opinion on Therapeutic Targets vol. 14 621–632, 2010

Ehtesham, M. et al. CXCR4 mediates the proliferation of glioblastoma progenitor cells. Cancer Lett, 274, 305–12, 2009

Falcicchia, C. et al. Involvement of p38 mapk in synaptic function and dysfunction. Int J Mol Sci, 21, 1–14, 2020

Farin, A. et al. Transplanted glioma cells migrate and proliferate on host brain vasculature: a dynamic analysis. Glia, 53, 799–808, 2006

Fearon, E. R. et al. A genetic model for colorectal tumorigenesis. Cell, 61, 759–67, 1990

Filatova, A. et al. The cancer stem cell niche(s): The crosstalk between glioma stem cells and their microenvironment. Biochim Biophys Acta - Gen Subj, 1830, 2496– 2508, 2013

Friedl, P. et al. New dimensions in cell migration. Nat Rev Mol Cell Biol, 13, 743–747,2012

G. Gritsenko, P. et al. Interstitial guidance of cancer invasion. J Pathol, 226, 185–199, 2012

Geer, C. P. et al. Interstitial fluid flow along white matter tracts: a potentially important mechanism for the dissemination of primary brain tumors. J Neurooncol, 32, 193–201, 1997

Giese, A. et al. Cost of Migration: Invasion of Malignant Gliomas and Implications for Treatment. J Clin Oncol, 21, 1624–1636, 2003

Gimple, R. C. et al. Glioblastoma stem cells: Lessons from the tumor hierarchy in a lethal cancer. Genes Dev, 33, 591–609, 2019

de Gooijer, M. C. et al. An Experimenter’s Guide to Glioblastoma Invasion Pathways. Trends in Molecular Medicine vol. 24 763–780, 2018

Gritsenko, P. G. et al. Adaptive adhesion systems mediate glioma cell invasion in complex environments. J Cell Sci, 131, 2018

Guha, A. et al. Proliferation of human malignant astrocytomas is dependent on Ras activation. Oncogene, 15, 2755–65, 1997

Hannon, G. J. et al. Pl5INK4B is a potentia| effector of TGF-β-induced cell cycle arrest. Nature, 371, 257–261, 1994

Hompland, T. et al. Interstitial fluid pressure and associated lymph node metastasis revealed in tumors by dynamic contrast-enhanced MRI. Cancer Res, 72, 4899– 4908, 2012

Huang, C. et al. MAP kinases and cell migration. J Cell Sci, 117, 4619–4628, 2004

Huang, D. W. et al. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, 4, 44–57, 2009

Huang, Y. C. et al. Stromal cell-derived factor-1 enhances motility and integrin up-regulation through CXCR4, ERK and NF-κB-dependent pathway in human lung cancer cells. Biochem Pharmacol, 74, 1702–1712, 2007

Huijbers, I. J. et al. A role for fibrillar collagen deposition and the collagen internalization receptor endo180 in glioma invasion. PLoS One, 5, 1–12, 2010

Hutmacher, D. W. et al. Can tissue engineering concepts advance tumor biology research? Trends Biotechnol, 28, 125–133, 2010

Iliff, J. J. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med, 4, 147ra111, 2012

Infanger, D. W. et al. Glioblastoma stem cells are regulated by interleukin-8 signaling in a tumoral perivascular niche. Cancer Res, 73, 7079–7089, 2013

Inoue, A. et al. Cancer stem-like cells of glioblastoma characteristically express MMP-13 and display highly invasive activity. Int J Oncol, 37, 1121–31, 2010

Iosif, R. E. et al. Tumor necrosis factor receptor 1 is a negative regulator of progenitor proliferation in adult hippocampal neurogenesis. J Neurosci, 26, 9703–9712, 2006

Kingsmore, K. M. et al. MRI analysis to map interstitial flow in the brain tumor microenvironment. APL Bioeng, 2, 031905, 2018

Kingsmore, K. M. et al. Interstitial flow differentially increases patient-derived glioblastoma stem cell invasion via CXCR4, CXCL12, and CD44-mediated mechanisms. Integr Biol (Camb), 8, 1246–1260, 2016

Lathia, J. et al. Cancer stem cells in glioblastoma. Genes Dev, 29, 1203–1217, 2015

Lathia, J. D. et al. Laminin alpha 2 enables glioblastoma stem cell growth. Ann Neurol, 72, 766–778, 2012

Lavoie, H. et al. ERK signalling: a master regulator of cell behaviour, life and fate. Nat Rev Mol Cell Biol, 21, 607–632, 2020

Li, T.-T. T. et al. Genetic variation responsible for mouse strain differences in integrin alpha 2 expression is associated with altered platelet responses to collagen. Blood, 103, 3396–3402, 2004

Louis, D. N. et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol, 131, 803–820, 2016

McCoy, M. G. et al. Endothelial cells promote 3D invasion of GBM by IL-8-dependent induction of cancer stem cell properties. Sci Rep, 9, 1–14, 2019

Miroshnikova, Y. A. et al. Tissue mechanics promote IDH1-dependent HIF1α-tenascin C feedback to regulate glioblastoma aggression. Nat Cell Biol, 18, 1336–1345, 2016

Munson, J. M. et al. Interstitial flow in a 3D microenvironment increases glioma invasion by a CXCR4-dependent mechanism. Cancer Res, 73, 1536–46, 2013

Namba, N. et al. Heterogeneous Glioma Cell Invasion under Interstitial Flow Depending on Their Differentiation Status. Tissue Eng - Part A, 27, 467–478, 2021

Neftel, C. et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell, 178, 835-849.e21, 2019

Nowell, P. C. The clonal evolution of tumor cell populations. Science, 194, 23–8, 1976 Ostrom, Q. T. et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2011-2015. Neuro Oncol, 20,iv1–iv86, 2018

Osuka, S. et al. Overcoming therapeutic resistance in glioblastoma: the way forward. J Clin Invest, 127, 415–426, 2017

Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science, 344, 1396–401, 2014

Piccirillo, S. G. M. et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature, 444, 761–765, 2006

Pistollato, F. et al. Intratumoral hypoxic gradient drives stem cells distribution and MGMT expression in glioblastoma. Stem Cells, 28, 851–862, 2010

Polacheck, W. J. et al. Mechanotransduction of fluid stresses governs 3D cell migration.

Proc Natl Acad Sci U S A, 111, 2447–2452, 2014

Principe, D. R. et al. TGF-β: Duality of function between tumor prevention and carcinogenesis. Journal of the National Cancer Institute vol. 106 2014

Qiang, L. et al. HIF-1α is critical for hypoxia-mediated maintenance of glioblastoma stem cells by activating Notch signaling pathway. Cell Death Differ, 19, 284–294, 2012

Qiu, B. et al. Human brain glioma stem cells are more invasive than their differentiated progeny cells in vitro. J Clin Neurosci, 19, 130–134, 2012

Quail, D. F. et al. The Microenvironmental Landscape of Brain Tumors. Cancer Cell, 31, 326–341, 2017

Rao, S. et al. CXCL12 mediates trophic interactions between endothelial and tumor cells in glioblastoma. PLoS One, 7, e33005, 2012

Sadatsuki, R. et al. Perlecan is required for the chondrogenic differentiation of synovial mesenchymal cells through regulation of Sox9 gene expression. J Orthop Res, 1– 10, 2016 doi:10.1002/jor.23318

Sampetrean, O. et al. Invasion precedes tumor mass formation in a malignant brain tumor model of genetically modified neural stem cells. Neoplasia, 13, 784–91, 2011

Sampetrean, O. et al. Characteristics of glioma stem cells. Brain Tumor Pathol, 30, 209–14, 2013

Sampetrean, O. et al. Modeling phenotypes of malignant gliomas. Cancer Sci, 109, 6– 14, 2018

Sánchez-Martín, F. J. et al. Lead induces similar gene expression changes in brains of gestationally exposed adult mice and in neurons differentiated from mouse embryonic stem cells. PLoS One, 8, 1–13, 2013

Sano, H. et al. Control of vessel diameters mediated by flow-induced outward vascular remodeling in vitro. Biofabrication, 12, 2020

Scherer, H. J. Structural development in gliomas. Am J Cancer, 34, 333–351, 1938

Serrano, M. et al. Role of the INK4a locus in tumor suppression and cell mortality. Cell, 85, 27–37, 1996

Shannon, P. et al. Pathological and Molecular Progression of Astrocytomas in a GFAP:12V-Ha-Ras Mouse Astrocytoma Model. Am J Pathol, 167, 859–867, 2005

Shao, N. et al. Interleukin-8 upregulates integrin β3 expression and promotes estrogen receptor-negative breast cancer cell invasion by activating the PI3K/Akt/NF-κB pathway. Cancer Lett, 8, 1–8, 2015

Shibao, S. et al. Metabolic heterogeneity and plasticity of glioma stem cells in a mouse glioblastoma model. Neuro Oncol, 20, 343–354, 2018

Shin, Y. et al. Microfluidic assay for simultaneous culture of multiple cell types on surfaces or within hydrogels. Nat Protoc, 7, 1247–59, 2012

Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature, 432, 396–401, 2004

Snuderl, M. et al. Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell, 20, 810–817, 2011

Sottoriva, A. et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A, 110, 4009–4014, 2013

Strojnik, T. et al. Neural stem cell markers, nestin and musashi proteins, in the progression of human glioma: correlation of nestin with prognosis of patient survival. Surg Neurol, 68, 133–143, 2007

Stupp, R. et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III

study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol, 10, 459–466, 2009

Stupp, R. et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med, 352, 987–996, 2005

Sudo, R. et al. Transport-mediated angiogenesis in 3D epithelial coculture. FASEB J, 23, 2155–2164, 2009

Sun, X. et al. Mathematical modeling reveals a critical role for cyclin D1 dynamics in phenotype switching during glioma differentiation. FEBS Lett, 589, 2304–2311, 2015

Szerlip, N. J. et al. Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci U S A, 109, 3041–3046, 2012

Takanaga, H. et al. cAMP-induced Astrocytic Differentiation of C6 Glioma Cells Is Mediated by Autocrine Interleukin-6. J Biol Chem, 279, 15441–15447, 2004

Tamura, R. et al. Visualization of spatiotemporal dynamics of human glioma stem cell invasion. Mol Brain, 12, 45, 2019

De Thé, H. Differentiation therapy revisited. Nat Rev Cancer, 18, 117–127, 2018

Toole, B. P. Hyaluronan-CD44 interactions in cancer: Paradoxes and possibilities. Clinical Cancer Research vol. 15 7462–7468, 2009

Truong, D. et al. A three-dimensional (3D) organotypic microfluidic model for glioma stem cells – Vascular interactions. Biomaterials, 198, 63–77, 2019

Ulmasov, B. et al. Inhibitors of Arg-Gly-Asp-Binding Integrins Reduce Development of Pancreatic Fibrosis in Mice. C Cell Mol Gastroenterol Hepatol, 2, 499–518, 2016

Ulmasov, B. et al. Differences in the degree of cerulein-induced chronic pancreatitis in C57BL/6 mouse substrains lead to new insights in identification of potential risk factors in the development of chronic pancreatitis. Am J Pathol, 183, 692–708, 2013

Verhaak, R. G. W. et al. Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell, 17, 98–110, 2010

Wachs, F. P. et al. Transforming growth factor-β1 is a negative modulator of adult neurogenesis. J Neuropathol Exp Neurol, 65, 358–370, 2006

Watkins, S. et al. Disruption of astrocyte-vascular coupling and the blood-brain barrier by invading glioma cells. Nat Commun, 5, 4196, 2014

Wen, P. Y. et al. Malignant gliomas in adults. N Engl J Med, 359, 492–507, 2008

Wolf, K. et al. Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol, 201, 1069– 1084, 2013

Wolf, K. J. et al. A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles. Proc Natl Acad Sci U S A, 117, 2020

Wolf, K. J. et al. A 3D topographical model of parenchymal infiltration and perivascular invasion in glioblastoma. APL Bioeng, 2, 031903, 2018

Xie, Q. et al. Targeting adaptive glioblastoma: An overview of proliferation and invasion. Neuro Oncol, 16, 1575–1584, 2014

Xing, F. et al. The Anti-Warburg Effect Elicited by the cAMP-PGC1α Pathway Drives Differentiation of Glioblastoma Cells into Astrocytes. Cell Rep, 18, 468–481, 2017

Yadav, V. et al. CXCR4 increases in-vivo glioma perivascular invasion, and reduces radiation induced apoptosis: A genetic knockdown study. Oncotarget, 7, 83701– 83719, 2016

Ye, X. et al. Tumor-Associated Microglia/Macrophages Enhance the Invasion of Glioma Stem-like Cells via TGF-β1 Signaling Pathway. J Immunol, 189, 444–453, 2012

Young, M. E. et al. Estimation of diffusion coefficients of proteins. Biotechnol Bioeng, 22, 947–955, 1980

Zanca, C. et al. Glioblastoma cellular cross-talk converges on NF-κB to attenuate EGFR inhibitor sensitivity. Genes Dev, 31, 1212–1227, 2017

Zhou, Y. et al. Autocrine BMP4 Signaling Enhances Tumor Aggressiveness via Promoting Wnt/β-Catenin Signaling in IDH1-mutant Gliomas. Transl Oncol, 13, 125–134, 2020

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る