РОЛЬ БЕЛКОВ NOTCH В ПРОЦЕССАХ КАНЦЕРОГЕНЕЗА

Обложка

Цитировать

Полный текст

Об авторах

М. В. Новикова

Российский онкологический научный центр имени Н.Н. Блохина, Москва

Email: pbkopnin@mail.ru
Россия

В. А. Рыбко

Российский онкологический научный центр имени Н.Н. Блохина, Москва

Email: pbkopnin@mail.ru
Россия

Н. В. Хромова

Российский онкологический научный центр имени Н.Н. Блохина, Москва

Email: pbkopnin@mail.ru
Россия

М. Д. Фармаковская

Российский онкологический научный центр имени Н.Н. Блохина, Москва

Email: pbkopnin@mail.ru
Россия

П. Б. Копнин

Российский онкологический научный центр имени Н.Н. Блохина, Москва

Автор, ответственный за переписку.
Email: pbkopnin@mail.ru
Копнин Павел Борисович Россия

Список литературы

  1. Рыбко В.А., Копнин Б.П., Хромова Н.В. и др. Роль изменений активности белков Notch в онкогенезе: множественные механизмы и возможности прикладного использования. Клиническая онкогема- тология 2011;4(2):103–10. [Rybko V.A., Kopnin B.P., Khromova N.V. et al. Role of Notch signaling in tumorigenesis: multiple mechanisms and therapeutic potential. Klinicheskaya onkogematologiya = Clinical Oncohematology 2011;4(2):103–10. (In Russ.)].
  2. Kidd S., Kelley M.R., Young M.W. et al. Sequence of the notch locus of Drosophila melanogaster: relationship of the encoded protein to mammalian clotting and growth factors. Mol Cell Biol 1986;6(9):3094–108.
  3. Greenwald I. The lin-12 locus of Caenorhabditis elegans. Bioessays 1987;6(2):70–3.
  4. Ellisen L.W., Bird J., West D.C. et al. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell 1991;66(4):649–61.
  5. Francis R., McGrath R., Zhang J. et al. Aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 2002;3(1):85–97.
  6. Schroeter E.H., Kisslinger J.A., Kopan R. et al. Notch-1 signalling requires ligand- induced proteolytic release of intracellular domain. Nature 1998;393(6683):382–6.
  7. Strooper B.D., Annaert W., Cupers P. et al. A presenilin-1-dependent gamma-secretase- like protease mediates release of Notch intracellular domain. Nature 1999;398(6727):518–22.
  8. Louvi A., Artavanis-Tsakonas S. Notch and disease: a growing field. Semin Cell Dev Biol 2012;23(4):473–80.
  9. Demehri S., Turkoz A., Kopan R. Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell 2009;16(1):55–66.
  10. Wang N.J., Sanborn Z., Arnett K.L. et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci USA 2011;108(43):17761–6.
  11. Koch U., Lehal R., Radtke F. Stem cells living with a Notch. Development 2013;140(4):689–704.
  12. Kopan R., Ilagan M.X. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell 2009; 137(2):216–33.
  13. Bray S.J. Notch signalling: a simple pathway becomes complex. Nat Rev 2006;7(9):678–89.
  14. Lieber T., Kidd S., Struhl G. DSL-Notch signaling in the Drosophila brain in response to olfactory stimulation. Neuron 2011;69(3):468–81.
  15. Alberi L., Lui S., Wang Y. et al. Activity- induced Notch signaling in neurons requires Arc/Arg3.1 and is essential for synaptic plasticity in hippocampal networks. Neuron 2011;69(3):437–44.
  16. Andersson E.R., Sandberg R., Lendahl U. Notch signaling: simplicity in design, versatility in function. Development 2011;138(17):3593–612.
  17. Rana N.A., Haltiwanger R.S. Fringe benefits: functional and structural impacts of O-glycosylation on the extracellular domain of Notch receptors. Curr Opin Struct Biol 2011;21(5):583–9.
  18. Acar M., Jafar-Nejad H., Takeuchi H. et al. Rumi is a CAP10 domain glycosyl- transferase that modifies Notch and is required for Notch signaling. Cell 2008;132(2):247–58.
  19. Lee T.V., Sethi M.K., Leonardi J. et al. Negative regulation of notch signaling by xylose. PLoS Genet 2013;9(6):35–47.
  20. Logeat F., Bessia C., Brou C. et al. The Notch1 receptor is cleaved constitutively by a furin-like convertase. Proc Natl Acad Sci USA 1998;95(14):8108–12.
  21. Weinmaster G., Fischer J.A. Notch ligand ubiquitylation: what is it good for? Dev Cell 2011;21(1):134–44.
  22. Jorissen E., De Strooper B. Gamma- secretase and the intramembrane proteolysis of Notch. Curr Top Dev Biol 2010;92:201–30.
  23. Borggrefe T., Liefke R. Fine-tuning of the intracellular canonical Notch signaling pathway. Cell Cycle 2012;11(2):264–76.
  24. Nagel A.C., Krejci A., Tenin G. et al. Hairless-mediated repression of notch target genes requires the combined activity of Groucho and CtBP corepressors. Mol Cell Biol 2005;25(23):10433–41.
  25. Oswald F., Winkler M., Cao Y. et al. RBP-Jkappa/SHARP recruits CtIP/CtBP corepressors to silence Notch target genes. Mol Cell Biol 2005;25(23):10379–90.
  26. Wallberg A.E., Pedersen K., Lendahl U. et al. P300 and PCAF act cooperatively to mediate transcriptional activation from chromatin templates by notch intracellular domains in vitro. Mol Cell Biol 2002;22(22):7812–9.
  27. Hubbard E.J., Wu G., Kitajewski J. et al. Sel-10, a negative regulator of lin-12 activity in Caenorhabditis elegans, encodes a member of the CDC4 family of proteins. Genes Dev 1997;11(23):3182–93.
  28. Fryer C.J., White J.B., Jones K.A. Mastermind recruits CycC: CDK8 to phosphorylate the Notch ICD and coordinate activation with turnover. Mol Cell 2004;16(4):509–20.
  29. Capaccione K.M., Pine S.R. The Notch signaling pathway as a mediator of tumor survival. Carcinogenesis 2013;34(7):1420–30.
  30. Greenwald I. LIN-12/Notch signaling: lessons from worms and flies. Genes Dev 1998;12(12):1751–62.
  31. Ohlstein B., Spradling A. Multipotent Drosophila intestinal stem cells specify daughter cell fates by differential notch signaling. Science 2007;315(5814):988–92.
  32. Bicker F., Schmidt M.H. EGFL7: a new player in homeostasis of the nervous system. Cell Cycle 2010;9(7):1263–9.
  33. Pompa J.L., Wakeham A., Correia K.M. et al. Conservation of the Notch signalling pathway in mammalian neurogenesis. Development 1997;124(6):1139–48.
  34. Mani S.A., Guo W., Liao M.J. et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133(4):704–15.
  35. Quaegebeur A., Lange C., Carmeliet P. The neurovascular link in health and disease: molecular mechanisms and therapeutic implications. Neuron 2011;71(3):406–24.
  36. Perdigoto C.N., Bardin A.J. Sending the right signal: Notch and stem cells. Biochim Biophys Acta 2013;1830(2):2307–22.
  37. Carlson M.E., Hsu M., Conboy I.M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 2008;454(7203):528–32.
  38. Ohlstein B., Spradling A. The adult Drosophila posterior midgut is maintained by pluripotent stem cells. Nature 2006;439(7075):470–4.
  39. Bigas A., Robert-Moreno A., Espinosa L. The Notch pathway in the developing hematopoietic system. Int J Dev Biol 2010;54(6):1175–88.
  40. Fernández-Sánchez V., Pelayo R., Flores-Guzmán P. et al. In vitro effects of stromal cells expressing different levels of Jagged-1 and Delta-1 on the growth of primitive and intermediate CD34(+) cell subsets from human cord blood. Blood Cells Mol Dis 2011;47(4):205–13.
  41. Pooter R.F., Schmitt T.M., Pompa J.L. et al. Notch signaling requires GATA-2 to inhibit myelopoiesis from embryonic stem cells and primary hemopoietic progenitors. J Immunol 2006;176(9):5267–75.
  42. Varnum-Finney B., Brashem-Stein C., Bernstein I.D. Combined effects of Notch signaling and cytokines induce a multiple log increase in precursors with lymphoid and myeloid reconstituting ability. Blood 2003;101(5):1784–9.
  43. Ohishi K., Katayama N., Shiku H. et al. Notch signalling in hematopoiesis. Semin Cell Dev Biol 2003;14(2):143–50.
  44. Weng A.P., Ferrando A.A., Lee W. et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 2004;306(5694):269–71.
  45. Thompson B.J., Buonamici S., Sulis M.L. et al. The SCFFBW7 ubiquitin ligase complex as a tumor suppressor in T cell leukemia. J Exp Med 2007;204(8):1825–35.
  46. Chiang M.Y., Xu L., Shestova O. et al. Leukemia-associated NOTCH1 alleles are weak tumor initiators but accelerate K-ras-initiated leukemia. J Clin Invest 2008;118(9):3181–94.
  47. Fabbri G., Rasi S., Rossi D. et al. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation. J Exp Med 2011;208(7):1389–401.
  48. Ranganathan P., Weaver K.L., Capobianco A.J. Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 2011;11(5):338–51.
  49. Wang Z., Li Y., Banerjee S., Sarkar F.H. Emerging role of Notch in stem cells and cancer. Cancer Lett 2009;279(1):8–12.
  50. Lu J., Ye X., Fan F. et al. Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of Jagged-1. Cancer Cell 2013;23(2):171–85.
  51. Reedijk M., Odorcic S., Chang L. et al. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 2005;65(18):8530–7.
  52. Nguyen B., Lefort K., Mandinova A. et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes Dev 2006;20(8):1028–42.
  53. Rangarajan A., Talora C., Okuyama R. et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J. 2001;20(13):3427–36.
  54. Nicolas M., Wolfer A., Raj K. et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet 2003;33(3):416–21.
  55. Viatour P., Ehmer U., Saddic L.A. et al. Notch signaling inhibits hepatocellular carcinoma following inactivation of the RB pathway. J Exp Med 2011;208(10):1963–76.
  56. Klinakis A., Lobry C., Abdel-Wahab O. et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature 2011;473(7346):230–3.
  57. Agrawal N., Frederick M.J., Pickering C.R. et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 2011;333(6046):1154–7.
  58. Lobry C., Oh P., Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it,s NOTCH what you think. J Exp Med 2011;208(10):1931–5.
  59. Wang Z., Li Y., Kong D. et al. Cross-talk between miRNA and Notch signaling pathways in tumor development and progression. Cancer Lett 2010; 292(2):141–8.
  60. Leong K.G., Niessen K., Kulic I. et al. Jagged1-mediated Notch activation induces epithelial-to-mesenchymal transition through Slug-induced repression of E-cadherin. J Exp Med 2007;204(12):2935–48.
  61. Wang Z., Li Y., Kong D. et al. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res 2009;69(6):2400–7.
  62. Sahlgren C., Gustafsson M.V., Jin S. et al. Notch signaling mediates hypoxia-induced tumor cell migration and invasion. Proc Natl Acad Sci USA 2008;105(17):6392–7.
  63. Jakowlew S.B. Transforming growth factor-beta in cancer and metastasis. Cancer Metastasis Rev 2006;25(3):435–57.
  64. Serini G., Gabbiani G. Mechanisms of myofibroblast activity and phenotypic modulation. Exp Cell Res 1999;250(2): 273–83.
  65. Studebaker A.W., Storci G., Werbeck J.L. et al. Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res 2008;68(21):9087–95.
  66. Minter L.M., Osborne B.A. Canonical and non-canonical Notch signaling in CD4+ T cells. Curr Top Microbiol Immunol 2012;360:99–114.
  67. Gentle M.E., Rose A., Bugeon L. et al. Noncanonical Notch signaling modulates cytokine responses of dendritic cells to inflammatory stimuli. J Immunol 2012;189(3):1274–84.
  68. Vacca A., Felli M.P., Palermo R. et al. Notch3 and pre-TCR interaction unveils distinct NF-kappaB pathways in T-cell development and leukemia. EMBO J 2006;25(5):1000–8.
  69. Jin S., Mutvei A.P., Chivukula I.V. et al. Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/ IKKβ. Oncogene 2013;32(41):4892–902.
  70. Ayaz F., Osborne B.A. Non-canonical notch signaling in cancer and immunity. Front Oncol 2014;4:345.
  71. Garzia L., Andolfo I., Cusanelli E. et al. MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma. PloS One 2009; 4(3):4998.
  72. Wu Z., Wu Y., Tian Y. et al. Differential effects of miR-34c-3p and miR-34c-5p on the proliferation, apoptosis and invasion of glioma cells. Oncol Lett 2013;6(5):1447–52.
  73. Wang Z., Banerjee S., Ahmad A. et al. Activated K-ras and INK4a/Arf deficiency cooperate during the development of pancreatic cancer by activation of Notch and NF-κB signaling pathways. PloS One 2011;6(6):20537.
  74. Ji Q., Hao X., Zhang M. et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PloS One 2009;4(8):6816.
  75. Pang R.T., Leung C.O., Ye T.M. et al. MicroRNA-34a suppresses invasion through downregulation of Notch1 and Jagged1 in cervical carcinoma and choriocarcinoma cells. Carcinogenesis 2010;31(6):1037–44.
  76. Bu P., Chen K.Y., Chen J.H. et al. A microRNA miR-34a-regulated bimodal switch targets Notch in colon cancer stem cells. Cell Stem Cell 2013; 12(5):602–15.
  77. Ohashi S., Natsuizaka M., Naganuma S. et al. A NOTCH3-mediated squamous cell differentiation program limits expansion of EMT-competent cells that express the ZEB transcription factors. Cancer Res 2011;71(21):6836–47.
  78. Brabletz S., Bajdak K., Meidhof S. et al. The ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells. EMBO J 2011;30(4):770–82.
  79. Wang X., Lu H., Li T. et al. Krüppel-like factor 8 promotes tumorigenic mammary stem cell induction by targeting miR-146a. Am J Cancer Res 2013;3(4):356–73.

Дополнительные файлы

Доп. файлы
Действие
1. JATS XML

© ,



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 57560 от  08.04.2014.