DOI QR코드

DOI QR Code

The role of NUMB/NUMB isoforms in cancer stem cells

  • Choi, Hye Yeon (Department of Molecular Microbiology and Immunology, University of Southern California) ;
  • Seok, Jaekwon (Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University) ;
  • Kang, Geun-Ho (Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University) ;
  • Lim, Kyung Min (Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University) ;
  • Cho, Ssang-Goo (Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University)
  • Received : 2021.03.18
  • Accepted : 2021.05.17
  • Published : 2021.07.31

Abstract

Cancer stem cells (CSCs) are a subpopulation of cancer that can self-renew and differentiate into large tumor masses. Evidence accumulated to date shows that CSCs affect tumor proliferation, recurrence, and resistance to chemotherapy. Recent studies have shown that, like stem cells, CSCs maintain cells with self-renewal capacity by means of asymmetric division and promote cell proliferation by means of symmetric division. This cell division is regulated by fate determinants, such as the NUMB protein, which recently has also been confirmed as a tumor suppressor. Loss of NUMB expression leads to uncontrolled proliferation and amplification of the CSC pool, which promotes the Notch signaling pathway and reduces the expression of the p53 protein. NUMB genes are alternatively spliced to produce six functionally distinct isoforms. An interesting recent discovery is that the protein NUMB isoform produced by alternative splicing of NUMB plays an important role in promoting carcinogenesis. In this review, we summarize the known functions of NUMB and NUMB isoforms related to the proliferation and generation of CSCs.

Keywords

Acknowledgement

This paper was supported by Konkuk University Researcher Fund in 2019. This study was supported by grants from the National Research Foundation (NRF) funded by the Korean government (grant no. 2019M3A9H1030682 and NRF-2015R1A5A1009701). This paper was supported by Konkuk University Researcher Fund in 2019.

References

  1. Yoo YD and Kwon YT (2015) Molecular mechanisms controlling asymmetric and symmetric self-renewal of cancer stem cells. J Anal Sci Technol 6, 28 https://doi.org/10.1186/s40543-015-0071-4
  2. Budillon A, Curley S, Fusco R and Mancini R (2019) Identification and targeting of stem cell-activated pathways in cancer therapy. Stem Cells Int 2019, 8549020
  3. Yang L, Shi P, Zhao G et al (2020) Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 5, 8 https://doi.org/10.1038/s41392-020-0110-5
  4. Clevers H (2005) Stem cells, asymmetric division and cancer. Nat Genet 37, 1027-1028 https://doi.org/10.1038/ng1005-1027
  5. Knoblich JA (2008) Mechanisms of asymmetric stem cell division. Cell 132, 583-597 https://doi.org/10.1016/j.cell.2008.02.007
  6. Knoblich JA (2010) Asymmetric cell division: recent developments and their implications for tumour biology. Nat Rev Mol Cell Biol 11, 849-860 https://doi.org/10.1038/nrm3010
  7. Majumdar S and Liu ST (2020) Cell division symmetry control and cancer stem cells. AIMS Mol Sci 7, 82-98 https://doi.org/10.3934/molsci.2020006
  8. Badve S and Nakshatri H (2012) Breast-cancer stem cells-beyond semantics. Lancet Oncol 13, e43-48 https://doi.org/10.1016/S1470-2045(11)70191-7
  9. Najafi M, Mortezaee K and Ahadi R (2019) Cancer stem cell (a)symmetry & plasticity: tumorigenesis and therapy relevance. Life Sci 231, 116520 https://doi.org/10.1016/j.lfs.2019.05.076
  10. Brooks MD, Burness ML and Wicha MS (2015) Therapeutic implications of cellular heterogeneity and plasticity in breast cancer. Cell Stem Cell 17, 260-271 https://doi.org/10.1016/j.stem.2015.08.014
  11. Plaks V, Kong N and Werb Z (2015) The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16, 225-238 https://doi.org/10.1016/j.stem.2015.02.015
  12. Morrison SJ and Kimble J (2006) Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441, 1068-1074 https://doi.org/10.1038/nature04956
  13. Losick VP, Morris LX, Fox DT and Spradling A (2011) Drosophila stem cell niches: a decade of discovery suggests a unified view of stem cell regulation. Dev Cell 21, 159-171 https://doi.org/10.1016/j.devcel.2011.06.018
  14. Santoro A, Vlachou T, Carminati M, Pelicci PG and Mapelli M (2016) Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 17, 1700-1720 https://doi.org/10.15252/embr.201643021
  15. Venkei ZG and Yamashita YM (2018) Emerging mechanisms of asymmetric stem cell division. J Cell Biol 217, 3785-3795 https://doi.org/10.1083/jcb.201807037
  16. Knoblich JA (2001) Asymmetric cell division during animal development. Nat Rev Mol Cell Biol 2, 11-20 https://doi.org/10.1038/35048085
  17. Gehring WJ (2004) Precis of Edwin G. Conklin's JEZ article, "Mosaic Development in Ascidian Eggs". J Exp Zool A Comp Exp Biol 301, 461-463 https://doi.org/10.1002/jez.a.87
  18. Sunchu B and Cabernard C (2020) Principles and mechanisms of asymmetric cell division. Development 147, dev167650 https://doi.org/10.1242/dev.167650
  19. Uemura T, Shepherd S, Ackerman L, Jan LY and Jan YN (1989) Numb, a gene required in determination of cell fate during sensory organ formation in Drosophila embryos. Cell 58, 349-360 https://doi.org/10.1016/0092-8674(89)90849-0
  20. Cayouette M and Raff M (2002) Asymmetric segregation of Numb: a mechanism for neural specification from Drosophila to mammals. Nat Neurosci 5, 1265-1269 https://doi.org/10.1038/nn1202-1265
  21. Dho SE, Jacob S, Wolting CD, French MB, Rohrschneider LR and McGlade CJ (1998) The mammalian numb phosphotyrosine-binding domain. Characterization of binding specificity and identification of a novel PDZ domaincontaining numb binding protein, LNX. J Biol Chem 273, 9179-9187 https://doi.org/10.1074/jbc.273.15.9179
  22. Gulino A, Di Marcotullio L and Screpanti I (2010) The multiple functions of Numb. Exp Cell Res 316, 900-906 https://doi.org/10.1016/j.yexcr.2009.11.017
  23. Zhong W, Feder JN, Jiang MM, Jan LY and Jan YN (1996) Asymmetric localization of a mammalian numb homolog during mouse cortical neurogenesis. Neuron 17, 43-53 https://doi.org/10.1016/S0896-6273(00)80279-2
  24. Bani-Yaghoub M, Kubu CJ, Cowling R et al (2007) A switch in numb isoforms is a critical step in cortical development. Dev Dyn 236, 696-705 https://doi.org/10.1002/dvdy.21072
  25. Dho SE, French MB, Woods SA and McGlade CJ (1999) Characterization of four mammalian numb protein isoforms. Identification of cytoplasmic and membrane-associated variants of the phosphotyrosine binding domain. J Biol Chem 274, 33097-33104 https://doi.org/10.1074/jbc.274.46.33097
  26. Karaczyn A, Bani-Yaghoub M, Tremblay R et al (2010) Two novel human NUMB isoforms provide a potential link between development and cancer. Neural Dev 5, 31 https://doi.org/10.1186/1749-8104-5-31
  27. Karaczyn AA, Adams TL, Cheng RY, Matluk NN and Verdi JM (2017) Human NUMB6 induces epithelial-mesenchymal transition and enhances breast cancer cells migration and invasion. J Cell Biochem 118, 237-251 https://doi.org/10.1002/jcb.25628
  28. Li Y, Wang D, Wang H et al (2021) A splicing factor switch controls hematopoietic lineage specification of pluripotent stem cells. EMBO Rep 22, e50535
  29. Wei R, Liu X, Voss C et al (2019) NUMB regulates the endocytosis and activity of the anaplastic lymphoma kinase in an isoform-specific manner. J Mol Cell Biol 11, 994-1005 https://doi.org/10.1093/jmcb/mjz003
  30. Kavanaugh WM, Turck CW and Williams LT (1995) PTB domain binding to signaling proteins through a sequence motif containing phosphotyrosine. Science 268, 1177-1179 https://doi.org/10.1126/science.7539155
  31. Verdi JM, Schmandt R, Bashirullah A et al (1996) Mammalian NUMB is an evolutionarily conserved signaling adapter protein that specifies cell fate. Curr Biol 6, 1134-1145 https://doi.org/10.1016/S0960-9822(02)70680-5
  32. Salcini AE, Confalonieri S, Doria M et al (1997) Binding specificity and in vivo targets of the EH domain, a novel protein-protein interaction module. Genes Dev 11, 2239-2249 https://doi.org/10.1101/gad.11.17.2239
  33. Santolini E, Puri C, Salcini AE et al (2000) Numb is an endocytic protein. J Cell Biol 151, 1345-1352 https://doi.org/10.1083/jcb.151.6.1345
  34. Verdi JM, Bashirullah A, Goldhawk DE et al (1999) Distinct human NUMB isoforms regulate differentiation vs. proliferation in the neuronal lineage. Proc Natl Acad Sci U S A 96, 10472-10476 https://doi.org/10.1073/pnas.96.18.10472
  35. Revil T, Gaffney D, Dias C, Majewski J and Jerome-Majewska LA (2010) Alternative splicing is frequent during early embryonic development in mouse. BMC Genomics 11, 399 https://doi.org/10.1186/1471-2164-11-399
  36. Dooley CM, James J, Jane McGlade C and Ahmad I (2003) Involvement of numb in vertebrate retinal development: evidence for multiple roles of numb in neural differentiation and maturation. J Neurobiol 54, 313-325 https://doi.org/10.1002/neu.10176
  37. Nilsen TW and Graveley BR (2010) Expansion of the eukaryotic proteome by alternative splicing. Nature 463, 457-463 https://doi.org/10.1038/nature08909
  38. Climente-Gonzalez H, Porta-Pardo E, Godzik A and Eyras E (2017) The functional impact of alternative splicing in cancer. Cell Rep 20, 2215-2226 https://doi.org/10.1016/j.celrep.2017.08.012
  39. Kahles A, Lehmann KV, Toussaint NC et al (2018) Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer Cell 34, 211-224 e216 https://doi.org/10.1016/j.ccell.2018.07.001
  40. Sciarrillo R, Wojtuszkiewicz A, Assaraf YG et al (2020) The role of alternative splicing in cancer: from oncogenesis to drug resistance. Drug Resist Updat 53, 100728 https://doi.org/10.1016/j.drup.2020.100728
  41. Lu Y, Xu W, Ji J et al (2015) Alternative splicing of the cell fate determinant Numb in hepatocellular carcinoma. Hepatology 62, 1122-1131 https://doi.org/10.1002/hep.27923
  42. Misquitta-Ali CM, Cheng E, O'Hanlon D et al (2011) Global profiling and molecular characterization of alternative splicing events misregulated in lung cancer. Mol Cell Biol 31, 138-150 https://doi.org/10.1128/MCB.00709-10
  43. Zhao YJ, Han HZ, Liang Y, Shi CZ, Zhu QC and Yang J (2015) Alternative splicing of VEGFA, APP and NUMB genes in colorectal cancer. World J Gastroenterol 21, 6550-6560 https://doi.org/10.3748/wjg.v21.i21.6550
  44. Kim KK, Nam J, Mukouyama YS and Kawamoto S (2013) Rbfox3-regulated alternative splicing of Numb promotes neuronal differentiation during development. J Cell Biol 200, 443-458 https://doi.org/10.1083/jcb.201206146
  45. Tarn WY, Kuo HC, Yu HI et al (2016) RBM4 promotes neuronal differentiation and neurite outgrowth by modulating Numb isoform expression. Mol Biol Cell 27, 1676-1683 https://doi.org/10.1091/mbc.E15-11-0798
  46. Kyriazis GA, Wei Z, Vandermey M et al (2008) Numb endocytic adapter proteins regulate the transport and processing of the amyloid precursor protein in an isoform-dependent manner: implications for Alzheimer disease pathogenesis. J Biol Chem 283, 25492-25502 https://doi.org/10.1074/jbc.M802072200
  47. Ntelios D, Berninger B and Tzimagiorgis G (2012) Numb and Alzheimer's disease: the current picture. Front Neurosci 6, 145 https://doi.org/10.3389/fnins.2012.00145
  48. Dho SE, Trejo J, Siderovski DP and McGlade CJ (2006) Dynamic regulation of mammalian numb by G protein-coupled receptors and protein kinase C activation: structural determinants of numb association with the cortical membrane. Mol Biol Cell 17, 4142-4155 https://doi.org/10.1091/mbc.e06-02-0097
  49. Sato K, Watanabe T, Wang S et al (2011) Numb controls E-cadherin endocytosis through p120 catenin with aPKC. Mol Biol Cell 22, 3103-3119 https://doi.org/10.1091/mbc.e11-03-0274
  50. Pece S, Confalonieri S, Romano PR and Di Fiore PP (2011) NUMB-ing down cancer by more than just a NOTCH. Biochim Biophys Acta 1815, 26-43
  51. Colaluca IN, Tosoni D, Nuciforo P et al (2008) NUMB controls p53 tumour suppressor activity. Nature 451, 76-80 https://doi.org/10.1038/nature06412
  52. Westhoff B, Colaluca IN, D'Ario G et al (2009) Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci U S A 106, 22293-22298 https://doi.org/10.1073/pnas.0907781106
  53. Hong J, Liu Z, Zhu H et al (2014) The tumor suppressive role of NUMB isoform 1 in esophageal squamous cell carcinoma. Oncotarget 5, 5602-5614 https://doi.org/10.18632/oncotarget.2136
  54. Pece S, Serresi M, Santolini E et al (2004) Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J Cell Biol 167, 215-221 https://doi.org/10.1083/jcb.200406140
  55. Siddique HR, Feldman DE, Chen CL, Punj V, Tokumitsu H and Machida K (2015) NUMB phosphorylation destabilizes p53 and promotes self-renewal of tumor-initiating cells by a NANOG-dependent mechanism in liver cancer. Hepatology 62, 1466-1479 https://doi.org/10.1002/hep.27987
  56. Tosoni D, Zecchini S, Coazzoli M et al (2015) The Numb/p53 circuitry couples replicative self-renewal and tumor suppression in mammary epithelial cells. J Cell Biol 211, 845-862 https://doi.org/10.1083/jcb.201505037
  57. Rajendran D, Zhang Y, Berry DM and McGlade CJ (2016) Regulation of Numb isoform expression by activated ERK signaling. Oncogene 35, 5202-5213 https://doi.org/10.1038/onc.2016.69
  58. Langer W, Sohler F, Leder G et al (2010) Exon array analysis using re-defined probe sets results in reliable identification of alternatively spliced genes in non-small cell lung cancer. BMC Genomics 11, 676 https://doi.org/10.1186/1471-2164-11-676
  59. Artavanis-Tsakonas S, Rand MD and Lake RJ (1999) Notch signaling: cell fate control and signal integration in development. Science 284, 770-776 https://doi.org/10.1126/science.284.5415.770
  60. Hori K, Sen A and Artavanis-Tsakonas S (2013) Notch signaling at a glance. J Cell Sci 126, 2135-2140 https://doi.org/10.1242/jcs.127308
  61. Guo M, Jan LY and Jan YN (1996) Control of daughter cell fates during asymmetric division: interaction of Numb and Notch. Neuron 17, 27-41 https://doi.org/10.1016/S0896-6273(00)80278-0
  62. Schweisguth F (2004) Regulation of notch signaling activity. Curr Biol 14, R129-138 https://doi.org/10.1016/j.cub.2004.01.023
  63. Wai P, Truong B and Bhat KM (1999) Cell division genes promote asymmetric interaction between Numb and Notch in the Drosophila CNS. Development 126, 2759-2770 https://doi.org/10.1242/dev.126.12.2759
  64. Shao X, Ding Z, Zhao M et al (2017) Mammalian Numb protein antagonizes Notch by controlling postendocytic trafficking of the Notch ligand Delta-like 4. J Biol Chem 292, 20628-20643 https://doi.org/10.1074/jbc.M117.800946
  65. Brou C (2009) Intracellular trafficking of Notch receptors and ligands. Exp Cell Res 315, 1549-1555 https://doi.org/10.1016/j.yexcr.2008.09.010
  66. Frise E, Knoblich JA, Younger-Shepherd S, Jan LY and Jan YN (1996) The Drosophila Numb protein inhibits signaling of the Notch receptor during cell-cell interaction in sensory organ lineage. Proc Natl Acad Sci U S A 93, 11925-11932 https://doi.org/10.1073/pnas.93.21.11925
  67. Flores AN, McDermott N, Meunier A and Marignol L (2014) NUMB inhibition of NOTCH signalling as a therapeutic target in prostate cancer. Nat Rev Urol 11, 499-507 https://doi.org/10.1038/nrurol.2014.195
  68. Crawford TQ and Roelink H (2007) The notch response inhibitor DAPT enhances neuronal differentiation in embryonic stem cell-derived embryoid bodies independently of sonic hedgehog signaling. Dev Dyn 236, 886-892 https://doi.org/10.1002/dvdy.21083
  69. Liu X, Xu QR, Xie WF and Wang MD (2014) DAPT suppresses the proliferation of human glioma cell line SHG-44. Asian Pac J Trop Med 7, 552-556 https://doi.org/10.1016/S1995-7645(14)60092-4
  70. Luo Z, Mu L, Zheng Y et al (2020) NUMB enhances Notch signaling by repressing ubiquitination of NOTCH1 intracellular domain. J Mol Cell Biol 12, 345-358 https://doi.org/10.1093/jmcb/mjz088
  71. Bolos V, Grego-Bessa J and de la Pompa JL (2007) Notch signaling in development and cancer. Endocr Rev 28, 339-363 https://doi.org/10.1210/er.2006-0046
  72. Leong KG and Karsan A (2006) Recent insights into the role of Notch signaling in tumorigenesis. Blood 107, 2223-2233 https://doi.org/10.1182/blood-2005-08-3329
  73. Nowell CS and Radtke F (2017) Notch as a tumour suppressor. Nat Rev Cancer 17, 145-159 https://doi.org/10.1038/nrc.2016.145
  74. Kruse JP and Gu W (2009) Modes of p53 regulation. Cell 137, 609-622 https://doi.org/10.1016/j.cell.2009.04.050
  75. Bieging KT, Mello SS and Attardi LD (2014) Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer 14, 359-370 https://doi.org/10.1038/nrc3711
  76. Nag S, Qin J, Srivenugopal KS, Wang M and Zhang R (2013) The MDM2-p53 pathway revisited. J Biomed Res 27, 254-271 https://doi.org/10.7555/JBR.27.20130030
  77. Wang Z, Sandiford S, Wu C and Li SS (2009) Numb regulates cell-cell adhesion and polarity in response to tyrosine kinase signalling. EMBO J 28, 2360-2373 https://doi.org/10.1038/emboj.2009.190
  78. Steed E, Balda MS and Matter K (2010) Dynamics and functions of tight junctions. Trends Cell Biol 20, 142-149 https://doi.org/10.1016/j.tcb.2009.12.002
  79. Wang Z, Li Y, Kong D and Sarkar FH (2010) The role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr Drug Targets 11, 745-751 https://doi.org/10.2174/138945010791170860
  80. Qi S, Zhao X, Li M et al (2015) Aberrant expression of Notch1/numb/snail signaling, an epithelial mesenchymal transition related pathway, in adenomyosis. Reprod Biol Endocrinol 13, 96 https://doi.org/10.1186/s12958-015-0084-2