全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Focal Adhesion-Chromatin Linkage Controls Tumor Cell Resistance to Radio- and Chemotherapy

DOI: 10.1155/2012/319287

Full-Text   Cite this paper   Add to My Lib

Abstract:

Cancer resistance to therapy presents an ongoing and unsolved obstacle, which has clear impact on patient's survival. In order to address this problem, novel in vitro models have been established and are currently developed that enable data generation in a more physiological context. For example, extracellular-matrix- (ECM-) based scaffolds lead to the identification of integrins and integrin-associated signaling molecules as key promoters of cancer cell resistance to radio- and chemotherapy as well as modern molecular agents. In this paper, we discuss the dynamic nature of the interplay between ECM, integrins, cytoskeleton, nuclear matrix, and chromatin organization and how this affects the response of tumor cells to various kinds of cytotoxic anticancer agents. 1. Introduction Resistance to radiotherapy, chemotherapy, and novel molecular drugs still represents one of the major obstacles in cancer therapy [1–3]. Limited effectiveness of therapy inevitably results in progressive disease or recurrence, thereby reducing the chance of cure for the patients. Phenotypically, two types can be distinguished: pretherapeutically existing and acquired resistances [4, 5]. Acquired resistance to irradiation is not known, but anticancer drugs, both conventional and molecular, frequently induce defense mechanisms [6–8]. To optimize the efficacy of cytotoxic agents, it is necessary to ameliorate drug delivery to the tumor and to better understand the underlying molecular mechanisms causing the resistance or evolving the defense process [5, 9]. In order to address the latter, we and others focused on a particular cellular substructure called focal adhesion (FA) [10–17]. FAs are membrane areas, which cells employ to interact with the surrounding extracellular matrix (ECM) via integrin adhesion receptors [10, 17–22]. Due to their multiprotein composition including growth factor receptors, signaling, and adapter proteins, FAs are huge hubs for signaling downstream to control critical cell functions such as cell survival, proliferation, differentiation, and invasion [11, 13, 14, 18, 20, 22–30]. The highly complex interplay between all of these signaling molecules secures homeostasis of single cells as well as of tissues in the context of responses to external signals from the microenvironment. In tumor cells, according to the hallmarks of cancer [31], the proper physiological communication with the extracellular space is massively disturbed as a consequence of gene mutations and epigenetic modifications. Despite tumor growth-driving gene mutations, malignant cells often

References

[1]  J. Overgaard, “Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck—a systematic review and meta-analysis,” Radiotherapy and Oncology, vol. 100, no. 1, pp. 22–32, 2011.
[2]  I. K. Mellinghoff and C. L. Sawyers, “The emergence of resistance to targeted cancer therapeutics,” Pharmacogenomics, vol. 3, no. 5, pp. 603–623, 2002.
[3]  C. L. Sawyers, “Making progress through molecular attacks on cancer,” Cold Spring Harbor Symposia on Quantitative Biology, vol. 70, pp. 479–482, 2005.
[4]  M. M. Gottesman, “Mechanisms of cancer drug resistance,” Annual Review of Medicine, vol. 53, pp. 615–627, 2002.
[5]  D. B. Longley and P. G. Johnston, “Molecular mechanisms of drug resistance,” Journal of Pathology, vol. 205, no. 2, pp. 275–292, 2005.
[6]  B. Desoize and J. C. Jardillier, “Multicellular resistance: a paradigm for clinical resistance?” Critical Reviews in Oncology/Hematology, vol. 36, no. 2-3, pp. 193–207, 2000.
[7]  B. Goldman, “Multidrug resistance: can new drugs help chemotherapy score against cancer?” Journal of the National Cancer Institute, vol. 95, no. 4, pp. 255–257, 2003.
[8]  J. Mendelsohn and J. Baselga, “Epidermal growth factor receptor targeting in cancer,” Seminars in Oncology, vol. 33, no. 4, pp. 369–385, 2006.
[9]  M. Baumann, M. Krause, and R. Hill, “Exploring the role of cancer stem cells in radioresistance,” Nature Reviews Cancer, vol. 8, no. 7, pp. 545–554, 2008.
[10]  A. L. Berrier and K. M. Yamada, “Cell-matrix adhesion,” Journal of Cellular Physiology, vol. 213, no. 3, pp. 565–573, 2007.
[11]  I. Eke, Y. Deuse, S. Hehlgans et al., “β1 Integrin/FAK/cortactin signaling is essential for human head and neck cancer resistance to radiotherapy,” Journal of Clinical Investigation, vol. 122, no. 4, pp. 1529–1540, 2012.
[12]  B. Geiger, J. P. Spatz, and A. D. Bershadsky, “Environmental sensing through focal adhesions,” Nature Reviews Molecular Cell Biology, vol. 10, no. 1, pp. 21–33, 2009.
[13]  S. Hehlgans, M. Haase, and N. Cordes, “Signalling via integrins: implications for cell survival and anticancer strategies,” Biochimica et Biophysica Acta, vol. 1775, no. 1, pp. 163–180, 2007.
[14]  K. R. Legate, E. Monta?ez, O. Kudlacek, and R. F?ssler, “ILK, PINCH and parvin: the tIPP of integrin signalling,” Nature Reviews Molecular Cell Biology, vol. 7, no. 1, pp. 20–31, 2006.
[15]  S. H. Lo, “Focal adhesions: what's new inside,” Developmental Biology, vol. 294, no. 2, pp. 280–291, 2006.
[16]  S. K. Mitra, D. A. Hanson, and D. D. Schlaepfer, “Focal adhesion kinase: in command and control of cell motility,” Nature Reviews Molecular Cell Biology, vol. 6, no. 1, pp. 56–68, 2005.
[17]  M. Moser, K. R. Legate, R. Zent, and R. F?ssler, “The tail of integrins, talin, and kindlins,” Science, vol. 324, no. 5929, pp. 895–899, 2009.
[18]  E. H. J. Danen, “Integrins: regulators of tissue function and cancer progression,” Current Pharmaceutical Design, vol. 11, no. 7, pp. 881–891, 2005.
[19]  F. G. Giancotti and E. Ruoslahti, “Integrin signaling,” Science, vol. 285, no. 5430, pp. 1028–1032, 1999.
[20]  R. O. Hynes, “Integrins: bidirectional, allosteric signaling machines,” Cell, vol. 110, no. 6, pp. 673–687, 2002.
[21]  M. A. Schwartz and D. E. Ingber, “Integrating with integrins,” Molecular Biology of the Cell, vol. 5, no. 4, pp. 389–393, 1994.
[22]  M. M. Zutter, “Integrin-mediated adhesion: tipping the balance between chemosensitivity and chemoresistance,” Advances in Experimental Medicine and Biology, vol. 608, pp. 87–100, 2007.
[23]  M. J. Bissell, H. G. Hall, and G. Parry, “How does the extracellular matrix direct gene expression?” Journal of Theoretical Biology, vol. 99, no. 1, pp. 31–68, 1982.
[24]  C. Brakebusch and R. F?ssler, “β 1 integrin function in vivo: adhesion, migration and more,” Cancer and Metastasis Reviews, vol. 24, no. 3, pp. 403–411, 2005.
[25]  J. Chung and H. K. Tae, “Integrin-dependent translational control: implication in cancer progression,” Microscopy Research and Technique, vol. 71, no. 5, pp. 380–386, 2008.
[26]  N. Cordes and C. C. Park, “β1 integrin as a molecular therapeutic target,” International Journal of Radiation Biology, vol. 83, no. 11-12, pp. 753–760, 2007.
[27]  V. Morello, S. Cabodi, S. Sigismund et al., “β1 integrin controls EGFR signaling and tumorigenic properties of lung cancer cells,” Oncogene, vol. 30, pp. 4087–4096, 2011.
[28]  A. G. Ramsay, J. F. Marshall, and I. R. Hart, “Integrin trafficking and its role in cancer metastasis,” Cancer and Metastasis Reviews, vol. 26, no. 3-4, pp. 567–578, 2007.
[29]  M. A. Schwartz and R. K. Assoian, “Integrins and cell proliferation: regulation of cyclin-dependent kinases via cytoplasmic signaling pathways,” Journal of Cell Science, vol. 114, no. 14, pp. 2553–2560, 2001.
[30]  K. M. Yamada, R. Pankov, and E. Cukierman, “Dimensions and dynamics in integrin function,” Brazilian Journal of Medical and Biological Research, vol. 36, no. 8, pp. 959–966, 2003.
[31]  D. Hanahan and R. A. Weinberg, “Hallmarks of cancer: the next generation,” Cell, vol. 144, no. 5, pp. 646–674, 2011.
[32]  M. H. Barcellos-Hoff, C. Park, and E. G. Wright, “Radiation and the microenvironment—tumorigenesis and therapy,” Nature Reviews Cancer, vol. 5, no. 11, pp. 867–875, 2005.
[33]  I. Eke and N. Cordes, “Radiobiology goes 3D: how ECM and cell morphology impact on cell survival after irradiation,” Radiotherapy and Oncology, vol. 99, no. 3, pp. 271–278, 2011.
[34]  S. H. Kim, J. Turnbull, and S. Guimond, “Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor,” Journal of Endocrinology, vol. 209, no. 2, pp. 139–151, 2011.
[35]  L. A. Hazlehurst, T. H. Landowski, and W. S. Dalton, “Role of the tumor microenvironment in mediating de novo resistance to drugs and physiological mediators of cell death,” Oncogene, vol. 22, no. 47, pp. 7396–7402, 2003.
[36]  T. Elsdale and J. Bard, “Collagen substrata for studies on cell behavior,” Journal of Cell Biology, vol. 54, no. 3, pp. 626–637, 1972.
[37]  C. M. Nelson and M. J. Bissell, “Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer,” Annual Review of Cell and Developmental Biology, vol. 22, pp. 287–309, 2006.
[38]  F. Pampaloni, E. G. Reynaud, and E. H. K. Stelzer, “The third dimension bridges the gap between cell culture and live tissue,” Nature Reviews Molecular Cell Biology, vol. 8, no. 10, pp. 839–845, 2007.
[39]  O. W. Petersen, L. Ronnov-Jessen, A. R. Howlett, and M. J. Bissell, “Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 89, no. 19, pp. 9064–9068, 1992.
[40]  K. Storch, I. Eke, K. Borgmann et al., “Three-dimensional cell growth confers radioresistance by chromatin density modification,” Cancer Research, vol. 70, no. 10, pp. 3925–3934, 2010.
[41]  A. Alavi and D. G. Stupack, “Cell survival in a three-dimensional matrix,” Methods in Enzymology, vol. 426, pp. 85–101, 2007.
[42]  M. J. Bissell, A. Rizki, and I. S. Mian, “Tissue architecture: the ultimate regulator of breast epithelial function,” Current Opinion in Cell Biology, vol. 15, no. 6, pp. 753–762, 2003.
[43]  I. Eke, U. Koch, S. Hehlgans et al., “PINCH1 regulates Akt1 activation and enhances radioresistance by inhibiting PP1α,” Journal of Clinical Investigation, vol. 120, no. 7, pp. 2516–2527, 2010.
[44]  S. Hehlgans, I. Eke, K. Storch, M. Haase, G. B. Baretton, and N. Cordes, “Caveolin-1 mediated radioresistance of 3D grown pancreatic cancer cells,” Radiotherapy and Oncology, vol. 92, no. 3, pp. 362–370, 2009.
[45]  S. Hehlgans, I. Lange, I. Eke, and N. Cordes, “3D cell cultures of human head and neck squamous cell carcinoma cells are radiosensitized by the focal adhesion kinase inhibitor TAE226,” Radiotherapy and Oncology, vol. 92, no. 3, pp. 371–378, 2009.
[46]  J. Le Beyec, R. Xu, S. Y. Lee et al., “Cell shape regulates global histone acetylation in human mammary epithelial cells,” Experimental Cell Research, vol. 313, no. 14, pp. 3066–3075, 2007.
[47]  C. C. Park, H. J. Zhang, E. S. Yao, C. J. Park, and M. J. Bissell, “β1 integrin inhibition dramatically enhances radiotherapy efficacy in human breast cancer xenografts,” Cancer Research, vol. 68, no. 11, pp. 4398–4405, 2008.
[48]  C. D. Roskelley, P. Y. Desprez, and M. J. Bissell, “Extracellular matrix-dependent tissue-specific gene expression in mammary epithelial cells requires both physical and biochemical signal transduction,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 26, pp. 12378–12382, 1994.
[49]  C. H. Streuli, C. Schmidhauser, N. Bailey et al., “Laminin mediates tissue-specific gene expression in mammary epithelia,” Journal of Cell Biology, vol. 129, no. 3, pp. 591–603, 1995.
[50]  K. M. Yamada and E. Cukierman, “Modeling tissue morphogenesis and cancer in 3D,” Cell, vol. 130, no. 4, pp. 601–610, 2007.
[51]  I. Eke, F. Leonhardt, K. Storch, S. Hehlgans, and N. Cordes, “The small molecule inhibitor QLT0267 radiosensitizes squamous cell carcinoma cells of the head and neck,” PLoS ONE, vol. 4, no. 7, Article ID e6434, 2009.
[52]  V. M. Weaver, O. W. Petersen, F. Wang et al., “Reversion of the malignant phenotype of human breast cells in three- dimensional culture and in vivo by integrin blocking antibodies,” Journal of Cell Biology, vol. 137, no. 1, pp. 231–245, 1997.
[53]  R. Xu, C. M. Nelson, J. L. Muschler, M. Veiseh, B. K. Vonderhaar, and M. J. Bissell, “Sustained activation of STAT5 is essential for chromatin remodeling and maintenance of mammary-specifi c function,” Journal of Cell Biology, vol. 184, no. 1, pp. 57–66, 2009.
[54]  R. E. Durand, “The influence of microenvironmental factors during cancer therapy,” In Vivo, vol. 8, no. 5, pp. 691–702, 1994.
[55]  I. Eke and N. Cordes, “Dual targeting of EGFR and focal adhesion kinase in 3D grown HNSCC cell cultures,” Radiotherapy and Oncology, vol. 99, no. 3, pp. 279–286, 2011.
[56]  L. A. Kunz-Schughart, J. P. Freyer, F. Hofstaedter, and R. Ebner, “The use of 3-D cultures for high-throughput screening: the multicellular spheroid model,” Journal of Biomolecular Screening, vol. 9, no. 4, pp. 273–285, 2004.
[57]  I. Dufau, C. Frongia, F. Sicard et al., “Multicellular tumor spheroid model to evaluate spatio-temporal dynamics effect of chemotherapeutics: application to the gemcitabine/CHK1 inhibitor combination in pancreatic cancer,” BMC Cancer, vol. 12, article 15, 2012.
[58]  J. S. Damiano, “Integrins as novel drug targets for overcoming innate drug resistance,” Current Cancer Drug Targets, vol. 2, no. 1, pp. 37–43, 2002.
[59]  J. S. Damiano, A. E. Cress, L. A. Hazlehurst, A. A. Shtil, and W. S. Dalton, “Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines,” Blood, vol. 93, no. 5, pp. 1658–1667, 1999.
[60]  M. B. Meads, R. A. Gatenby, and W. S. Dalton, “Environment-mediated drug resistance: a major contributor to minimal residual disease,” Nature Reviews Cancer, vol. 9, no. 9, pp. 665–674, 2009.
[61]  V. Sandfort, U. Koch, and N. Cordes, “Cell adhesion-mediated radioresistance revisited,” International Journal of Radiation Biology, vol. 83, no. 11-12, pp. 727–732, 2007.
[62]  V. M. Weaver and C. D. Roskelley, “Extracellular matrix: the central regulator of cell and tissue homeostasis,” Trends in Cell Biology, vol. 7, no. 1, pp. 40–42, 1997.
[63]  B. Weigelt, A. T. Lo, C. C. Park, J. W. Gray, and M. J. Bissell, “HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment,” Breast Cancer Research and Treatment, vol. 122, no. 1, pp. 35–43, 2010.
[64]  D. E. Ingber, “Tensegrity: the architectural basis of cellular mechanotransduction,” Annual Review of Physiology, vol. 59, pp. 575–599, 1997.
[65]  M. A. Schwartz, “Integrin signaling revisited,” Trends in Cell Biology, vol. 11, no. 12, pp. 466–470, 2001.
[66]  A. Van der Flier and A. Sonnenberg, “Function and interactions of integrins,” Cell and Tissue Research, vol. 305, no. 3, pp. 285–298, 2001.
[67]  N. Wang and D. E. Ingber, “Control of cytoskeletal mechanics by extracellular matrix, cell shape, and mechanical tension,” Biophysical Journal, vol. 66, no. 6, pp. 2181–2189, 1994.
[68]  N. Wang, J. D. Tytell, and D. E. Ingber, “Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus,” Nature Reviews Molecular Cell Biology, vol. 10, no. 1, pp. 75–82, 2009.
[69]  M. J. Bissell, D. C. Radisky, A. Rizki, V. M. Weaver, and O. W. Petersen, “The organizing principle: microenvironmental influences in the normal and malignant breast,” Differentiation, vol. 70, no. 9-10, pp. 537–546, 2002.
[70]  M. Larsen, V. V. Artym, J. A. Green, and K. M. Yamada, “The matrix reorganized: extracellular matrix remodeling and integrin signaling,” Current Opinion in Cell Biology, vol. 18, no. 5, pp. 463–471, 2006.
[71]  R. Xu, A. Boudreau, and M. J. Bissell, “Tissue architecture and function: dynamic reciprocity via extra- and intra-cellular matrices,” Cancer and Metastasis Reviews, vol. 28, no. 1-2, pp. 167–176, 2009.
[72]  N. Cordes and V. Meineke, “Cell adhesion-mediated radioresistance (CAM-RR): extracellular matrix-dependent improvement of cell survival in human tumor and normal cells in vitro,” Strahlentherapie und Onkologie, vol. 179, no. 5, pp. 337–344, 2003.
[73]  W. S. Dalton, “The tumor microenvironment: focus on myeloma,” Cancer Treatment Reviews, vol. 29, supplement 1, pp. 11–19, 2003.
[74]  F. M. Watt, “Role of integrins in regulating epidermal adhesion, growth and differentiation,” The EMBO Journal, vol. 21, no. 15, pp. 3919–3926, 2002.
[75]  R. J. Blaschke, A. R. Howlett, P. Y. Desprez, O. W. Petersen, and M. J. Bissell, “Cell differentiation by extracellular matrix components,” Methods in Enzymology, vol. 245, pp. 535–556, 1995.
[76]  M. A. LaBarge, O. W. Petersen, and M. J. Bissell, “Of microenvironments and mammary stem cells,” Stem Cell Reviews, vol. 3, no. 2, pp. 137–146, 2007.
[77]  O. W. Petersen, L. Ronnov-Jessen, V. M. Weaver, and M. J. Bissell, “Differentiation and cancer in the mammary gland: shedding light on an old dichotomy,” Advances in Cancer Research, vol. 75, pp. 135–161, 1998.
[78]  N. Cordes, M. A. Blaese, V. Meineke, and D. Van Beuningen, “Ionizing radiation induces up-regulation of functional β1-integrin in human lung tumour cell lines in vitro,” International Journal of Radiation Biology, vol. 78, no. 5, pp. 347–357, 2002.
[79]  N. Cordes, B. Hansmeier, C. Beinke, V. Meineke, and D. Van Beuningen, “Irradiation differentially affects substratum-dependent survival, adhesion, and invasion of glioblastoma cell lines,” British Journal of Cancer, vol. 89, no. 11, pp. 2122–2132, 2003.
[80]  N. Cordes, J. Seidler, R. Durzok, H. Geinitz, and C. Brakebusch, “β1-integrin-mediated signaling essentially contributes to cell survival after radiation-induced genotoxic injury,” Oncogene, vol. 25, no. 9, pp. 1378–1390, 2006.
[81]  F. Hess, D. Estrugo, A. Fischer, C. Belka, and N. Cordes, “Integrin-linked kinase interacts with caspase-9 and -8 in an adhesion-dependent manner for promoting radiation-induced apoptosis in human leukemia cells,” Oncogene, vol. 26, no. 10, pp. 1372–1384, 2007.
[82]  V. Meineke, K. P. Gilbertz, K. Schilperoort et al., “Ionizing radiation modulates cell surface integrin expression and adhesion of COLO-320 cells to collagen and fibronectin in vitro,” Strahlentherapie und Onkologie, vol. 178, no. 12, pp. 709–714, 2002.
[83]  T. Sethi, R. C. Rintoul, S. M. Moore et al., “Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo,” Nature Medicine, vol. 5, no. 6, pp. 662–668, 1999.
[84]  K. M. Yamada and S. Even-Ram, “Integrin regulation of growth factor receptors,” Nature Cell Biology, vol. 4, no. 4, pp. E75–E76, 2002.
[85]  D. E. Ingber, L. Dike, L. Hansen et al., “Cellular tensegrity: exploring how mechanical changes in the cytoskeleton regulate cell growth, migration, and tissue pattern during morphogenesis,” International Review of Cytology, vol. 150, pp. 173–224, 1994.
[86]  S. Lelièvre, V. M. Weaver, and M. J. Bissell, “Extracellular matrix signaling from the cellular membrane skeleton to the nuclear skeleton: a model of gene regulation,” Recent Progress in Hormone Research, vol. 51, pp. 417–432, 1996.
[87]  J. G. Eriksen, T. Steiniche, H. S?gaard, and J. Overgaard, “Expression of integrins and E-cadherin in squamous cell carcinomas of the head and neck,” APMIS, vol. 112, no. 9, pp. 560–568, 2004.
[88]  A. C. Dingemans, V. van den Boogaart, B. A. Vosse, R. J. van Suylen, A. W. Griffioen, and V. L. Thijssen, “Integrin expression profiling identifies integrin alpha5 and beta1 as prognostic factors in early stage non-small cell lung cancer,” Molecular Cancer, vol. 9, article 152, 2010.
[89]  S. Dedhar, R. Saulnier, R. Nagel, and C. M. Overall, “Specific alterations in the expression of α3β1 and α6β4 integrins in highly invasive and metastatic variants of human prostate carcinoma cells selected by in vitro invasion through reconstituted basement membrane,” Clinical and Experimental Metastasis, vol. 11, no. 5, pp. 391–400, 1993.
[90]  N. Ahmed, F. Pansino, R. Clyde et al., “Overexpression of αvβ6 integrin in serous epithelial ovarian cancer regulates extracellular matrix degradation via the plasminogen activation cascade,” Carcinogenesis, vol. 23, no. 2, pp. 237–244, 2002.
[91]  E. S. Yao, H. Zhang, Y. Y. Chen et al., “Increased β1 integrin is associated with decreased survival in invasive breast cancer,” Cancer Research, vol. 67, no. 2, pp. 659–664, 2007.
[92]  T. M. desRochers, Y. Shamis, A. Alt-Holland et al., “The 3D tissue microenvironment modulates DNA methylation and E-cadherin expression in squamous cell carcinoma,” Epigenetics, vol. 7, no. 1, pp. 34–46, 2012.
[93]  P. Jayaraj, S. Sen, A. Sharma, et al., “Epigenetic inactivation of E-cadherin gene in Eyelid Sebaceous gland carcinoma,” British Journal of Dermatology. In press.
[94]  A. J. Maniotis, K. Valyi-Nagy, J. Karavitis et al., “Chromatin organization measured by AluI restriction enzyme changes with malignancy and is regulated by the extracellular matrix and the cytoskeleton,” American Journal of Pathology, vol. 166, no. 4, pp. 1187–1203, 2005.
[95]  M. J. Bissell, V. M. Weaver, S. A. Lelièvre, F. Wang, O. W. Petersen, and K. L. Schmeichel, “Tissue structure, nuclear organization, and gene expression in normal and malignant breast,” Cancer Research, vol. 59, no. 7, supplement, pp. 1757s–1764s, 1999.
[96]  E. Cukierman, R. Pankov, and K. M. Yamada, “Cell interactions with three-dimensional matrices,” Current Opinion in Cell Biology, vol. 14, no. 5, pp. 633–639, 2002.
[97]  J. Alcaraz, R. Xu, H. Mori et al., “Laminin and biomimetic extracellular elasticity enhance functional differentiation in mammary epithelia,” The EMBO Journal, vol. 27, no. 21, pp. 2829–2838, 2008.
[98]  J. M. Onoda, M. P. Piechocki, and K. V. Honn, “Radiation-induced increase in expression of the α(IIb)β3 integrin in melanoma cells: effects on metastatic potential,” Radiation Research, vol. 130, no. 3, pp. 281–288, 1992.
[99]  Z. Fuks, I. Vlodavsky, M. Andreeff, M. McLoughlin, and A. Haimovitz-Friedman, “Effects of extracellular matrix on the response of endothelial cells to radiation in vitro,” European Journal of Cancer Part A, vol. 28, no. 4-5, pp. 725–731, 1992.
[100]  V. Meineke, K. Müller, R. Ridi et al., “Development and evaluation of a skin organ model for the analysis of radiation effects,” Strahlentherapie und Onkologie, vol. 180, no. 2, pp. 102–108, 2004.
[101]  I. Vlodavsky, Z. Fuks, R. Ishai-Michaeli et al., “Extracellular matrix-resident basic fibroblast growth factor: implication for the control of angiogenesis,” Journal of Cellular Biochemistry, vol. 45, no. 2, pp. 167–176, 1991.
[102]  N. Cordes, “Integrin-mediated cell-matrix interactions for prosurvivaland antiapoptotic signaling after genotoxic injury,” Cancer Letters, vol. 242, no. 1, pp. 11–19, 2006.
[103]  N. Cordes and D. Van Beuningen, “Cell adhesion to the extracellular matrix protein fibronectin modulates radiation-dependent G2 phase arrest involving integrin-linked kinase (ILK) and glycogen synthase kinase-3β (GSK-3β) in vitro,” British Journal of Cancer, vol. 88, no. 9, pp. 1470–1479, 2003.
[104]  I. Eke, V. Sandfort, A. Mischkus, M. Baumann, and N. Cordes, “Antiproliferative effects of EGFR tyrosine kinase inhibition and radiation-induced genotoxic injury are attenuated by adhesion to fibronectin,” Radiotherapy and Oncology, vol. 80, no. 2, pp. 178–184, 2006.
[105]  L. A. Hazlehurst, J. S. Damiano, I. Buyuksal, W. J. Pledger, and W. S. Dalton, “Adhesion to fibronectin via β1 integrins regulates p27(kip1) levels and contributes to cell adhesion mediated drug resistance (CAM-DR),” Oncogene, vol. 19, no. 38, pp. 4319–4327, 2000.
[106]  S. Mochizuki and Y. Okada, “ADAMs in cancer cell proliferation and progression,” Cancer Science, vol. 98, no. 5, pp. 621–628, 2007.
[107]  V. M. Weaver, S. Lelièvre, J. N. Lakins et al., “β4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium,” Cancer Cell, vol. 2, no. 3, pp. 205–216, 2002.
[108]  C. L. Kremer, M. Schmelz, and A. E. Cress, “Integrin-dependent amplification of the G2 arrest induced by ionizing radiation,” Prostate, vol. 66, no. 1, pp. 88–96, 2006.
[109]  D. Estrugo, A. Fischer, F. Hess, H. Scherthan, C. Belka, and N. Cordes, “Ligand bound β1 integrins inhibit procaspase-8 for mediating cell adhesion-mediated drug and radiation resistance in human leukemia cells,” PLoS ONE, vol. 2, no. 3, article e269, 2007.
[110]  I. Eke, S. Hehlgans, and N. Cordes, “There's something about ILK,” International Journal of Radiation Biology, vol. 85, no. 11, pp. 929–936, 2009.
[111]  S. A. Wickstr?m, A. Lange, E. Montanez, and R. F?ssler, “The ILK/PINCH/parvin complex: the kinase is dead, long live the pseudokinase!,” The EMBO Journal, vol. 29, no. 2, pp. 281–291, 2010.
[112]  N. Cordes, S. Frick, T. B. Brunner et al., “Human pancreatic tumor cells are sensitized to ionizing radiation by knockdown of caveolin-1,” Oncogene, vol. 26, no. 48, pp. 6851–6862, 2007.
[113]  I. Eke, V. Sandfort, K. Storch, M. Baumann, B. R?per, and N. Cordes, “Pharmacological inhibition of EGFR tyrosine kinase affects ILK-mediated cellular radiosensitization in vitro,” International Journal of Radiation Biology, vol. 83, no. 11-12, pp. 793–802, 2007.
[114]  S. Hehlgans and N. Cordes, “Caveolin-1: an essential modulator of cancer cell radio-and chemoresistance,” American Journal of Cancer Research, vol. 1, pp. 521–530, 2011.
[115]  S. Hehlgans, I. Eke, and N. Cordes, “An essential role of integrin-linked kinase in the cellular radiosensitivity of normal fibroblasts during the process of cell adhesion and spreading,” International Journal of Radiation Biology, vol. 83, no. 11-12, pp. 769–779, 2007.
[116]  S. Hehlgans, I. Eke, Y. Deuse, and N. Cordes, “Integrin-linked kinase: dispensable for radiation survival of three-dimensionally cultured fibroblasts,” Radiotherapy and Oncology, vol. 86, no. 3, pp. 329–335, 2008.
[117]  T. Kasahara, E. Koguchi, M. Funakoshi, E. Aizu-yokota, and Y. Sonoda, “Antiapoptotic action of focal adhesion kinase (FAK) against ionizing radiation,” Antioxidants and Redox Signaling, vol. 4, no. 3, pp. 491–499, 2002.
[118]  S. Hehlgans, I. Eke, and N. Cordes, “Targeting FAK radiosensitizes 3-dimensional grown human HNSCC cells through reduced Akt1 and MEK1/2 signaling,” International Journal of Radiation Oncology, Biology, Physics. In press.
[119]  G. W. McLean, N. O. Carragher, E. Avizienyte, J. Evans, V. G. Brunton, and M. C. Frame, “The role of focal-adhesion kinase in cancer—a new therapeutic opportunity,” Nature Reviews Cancer, vol. 5, no. 7, pp. 505–515, 2005.
[120]  C. S. Smith, V. M. Golubovskaya, E. Peck et al., “Effect of focal adhesion kinase (FAK) downregulation with FAK antisense oligonucleotides and 5-fluorouracil on the viability of melanoma cell lines,” Melanoma Research, vol. 15, no. 5, pp. 357–362, 2005.
[121]  W. Huanwen, L. Zhiyong, S. Xiaohua, R. Xinyu, W. Kai, and L. Tonghua, “Intrinsic chemoresistance to gemcitabine is associated with constitutive and laminin-induced phosphorylation of FAK in pancreatic cancer cell lines,” Molecular Cancer, vol. 8, article 125, 2009.
[122]  V. Sandfort, I. Eke, and N. Cordes, “The role of the focal adhesion protein PINCH1 for the radiosensitivity of adhesion and suspension cell cultures,” PLoS ONE, vol. 5, no. 9, Article ID e13056, 2010.
[123]  D. A. Reardon, B. Neyns, M. Weller, J. C. Tonn, L. B. Nabors, and R. Stupp, “Cilengitide: an RGD pentapeptide ανβ3 and ανβ5 integrin inhibitor in development for glioblastoma and other malignancies,” Future Oncology, vol. 7, no. 3, pp. 339–354, 2011.
[124]  M. R. Gilbert, J. Kuhn, K. R. Lamborn et al., “Cilengitide in patients with recurrent glioblastoma: the results of NABTC 03-02, a phase II trial with measures of treatment delivery,” Journal of Neuro-Oncology, vol. 106, no. 1, pp. 147–153, 2012.
[125]  R. Harisi, J. Dudas, J. Nagy-Olah, F. Timar, M. Szendroi, and A. Jeney, “Extracellular matrix induces doxorubicin-resistance in human osteosarcoma cells by suppression of p53 function,” Cancer Biology and Therapy, vol. 6, no. 8, pp. 1240–1246, 2007.
[126]  O. Zschenker, T. Streichert, S. Hehlgans, and N. Cordes, “Genome-wide gene expression analysis in cancer cells reveals 3D growth to affect ECM and processes associated with cell adhesion but not DNA repair,” PLoS ONE, vol. 7, no. 4, Article ID e34279, 2012.
[127]  A. J. Maniotis, C. S. Chen, and D. E. Ingber, “Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 3, pp. 849–854, 1997.
[128]  C. Schmidhauser, G. F. Casperson, C. A. Myers, K. T. Sanzo, S. Bolten, and M. J. Bissell, “A novel transcriptional enhancer is involved in the prolactin- and extracellular matrix-dependent regulation of β-casein gene expression,” Molecular Biology of the Cell, vol. 3, no. 6, pp. 699–709, 1992.
[129]  S. A. Lelièvre, “Contributions of extracellular matrix signaling and tissue architecture to nuclear mechanisms and spatial organization of gene expression control,” Biochimica et Biophysica Acta, vol. 1790, no. 9, pp. 925–935, 2009.
[130]  N. Boudreau, C. Myers, and M. J. Bissell, “From laminin to lamin: regulation of tissue-specific gene expression by the ECM,” Trends in Cell Biology, vol. 5, no. 1, pp. 1–4, 1995.
[131]  S. I. S. Grewal and S. Jia, “Heterochromatin revisited,” Nature Reviews Genetics, vol. 8, no. 1, pp. 35–46, 2007.
[132]  T. Kouzarides, “Chromatin Modifications and Their Function,” Cell, vol. 128, no. 4, pp. 693–705, 2007.
[133]  G. N. Li, L. L. Livi, C. M. Gourd, E. S. Deweerd, and D. Hoffman-Kim, “Genomic and morphological changes of neuroblastoma cells in response to three-dimensional matrices,” Tissue Engineering, vol. 13, no. 5, pp. 1035–1047, 2007.
[134]  S. Ghosh, G. C. Spagnoli, I. Martin et al., “Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study,” Journal of Cellular Physiology, vol. 204, no. 2, pp. 522–531, 2005.
[135]  C. Plachot and S. A. Lelièvre, “DNA methylation control of tissue polarity and cellular differentiation in the mammary epithelium,” Experimental Cell Research, vol. 298, no. 1, pp. 122–132, 2004.
[136]  T. Jenuwein and C. D. Allis, “Translating the histone code,” Science, vol. 293, no. 5532, pp. 1074–1080, 2001.
[137]  W. S. Xu, R. B. Parmigiani, and P. A. Marks, “Histone deacetylase inhibitors: molecular mechanisms of action,” Oncogene, vol. 26, no. 37, pp. 5541–5552, 2007.
[138]  E. I. Campos and D. Reinberg, “Histones: annotating chromatin,” Annual Review of Genetics, vol. 43, pp. 559–599, 2009.
[139]  C. C. Dufort, M. J. Paszek, and V. M. Weaver, “Balancing forces: architectural control of mechanotransduction,” Nature Reviews Molecular Cell Biology, vol. 12, no. 5, pp. 308–319, 2011.
[140]  K. J. Pienta and D. S. Coffey, “Nuclear-cytoskeletal interactions: evidence for physical connections between the nucleus and cell periphery and their alteration by transformation,” Journal of Cellular Biochemistry, vol. 49, no. 4, pp. 357–365, 1992.
[141]  M. Crisp, Q. Liu, K. Roux et al., “Coupling of the nucleus and cytoplasm: role of the LINC complex,” Journal of Cell Biology, vol. 172, no. 1, pp. 41–53, 2006.
[142]  J. A. Mellad, D. T. Warren, and C. M. Shanahan, “Nesprins LINC the nucleus and cytoskeleton,” Current Opinion in Cell Biology, vol. 23, no. 1, pp. 47–54, 2011.
[143]  H. J. Worman and G. G. Gundersen, “Here come the SUNs: a nucleocytoskeletal missing link,” Trends in Cell Biology, vol. 16, no. 2, pp. 67–69, 2006.
[144]  Q. Zhang, J. N. Skepper, F. Yang et al., “Nesprins: a novel family of spectrin-repeat-containing proteins that localize to the nuclear membrane in multiple tissues,” Journal of Cell Science, vol. 114, no. 24, pp. 4485–4498, 2001.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413