DOI QR코드

DOI QR Code

Targeted Silencing of Inhibitors of Apoptosis Proteins with siRNAs: A Potential Anti-cancer Strategy for Hepatocellular Carcinoma

  • Li, Gang (Department of General Surgery, Provincial Hospital Afficiated to Shandong University, Shandong University) ;
  • Chang, Hong (Department of General Surgery, Provincial Hospital Afficiated to Shandong University, Shandong University) ;
  • Zhai, Yun-Peng (Department of General Surgery, Provincial Hospital Afficiated to Shandong University, Shandong University) ;
  • Xu, Wei (Department of General Surgery, Provincial Hospital Afficiated to Shandong University, Shandong University)
  • Published : 2013.09.30

Abstract

Hepatocellular carcinoma (HCC) is one of the most common malignancies, with a very poor prognosis. Despite significant improvements in diagnosis and treatment in recent years, the long-term therapeutic efficacy is poor, partially due to tumor metastasis, tecurrence, and resistance to chemo-or radio-therapy. Recently, it was found that a major feature of tumors is a combination of unrestrained cell proliferation and impaired apoptosis. There are now 8 recogized members of the IAP-family: NAIP, c-IAP1, c-IAP2, XIAP, Survivin, Bruce, Livin and ILP-2. There proteins all contribute to ingibition of apoptosis, and provide new potential avenues of cancer treatment. As a powerful tool to suppress gene expression in mammalian cells, RNAi species for inhibiting IAP genes cab be directed against cancers. This review will provide a brief introduction to recent developments of the application IAP-siRNA in tumor studies, with the aim of inspiring future treatment of HCC.

Keywords

Acknowledgement

Supported by : National Science Foundation

References

  1. Akyurek N, Ren Y, Rassidakis GZ, Schlette EJ, Medeiros LJ (2006). Expression of inhibitor of apoptosis proteins in B-cell non-Hodgkin and Hodgkin lymphomas. Cancer, 107, 1844-51. https://doi.org/10.1002/cncr.22219
  2. Altieri DC (2003). Survivin in apoptosis control and cell cycle regulation in cancer. Prog Cell Cycle Res, 5, 447-52.
  3. Ashhab Y, AlianA, PolliackA, PanetA, Ben Yehuda D (2001). Two splicing variants of a new inhibitor of apoptosis gene with different biological properties and tissue distribution pattern. FEBS Lett, 495, 56-60. https://doi.org/10.1016/S0014-5793(01)02366-3
  4. Augello C, Caruso L, Maggioni M, et al (2009). Inhibitors of apoptosis proteins (IAPs) expression and their prognostic significance in hepatocellular carcinoma. BMC Cancer, 9, 125. https://doi.org/10.1186/1471-2407-9-125
  5. Bernstein E, Caudy AA, Hammond SM, Hannon GJ (2001). Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature, 409, 363-6. https://doi.org/10.1038/35053110
  6. Bhattacharyya M, Lemoine NR (2006). Gene therapy developments for pancreatic cancer. Best Pract Res Clin Gastroenterol, 20, 285-98.
  7. Bilim V, Yuuki K, Itoi T, et al (2008). Double inhibition of XIAP and Bcl-2 axis is beneficial for retrieving sensitivity of renal cell cancer to apoptosis. Br J Cancer, 98, 941-9. https://doi.org/10.1038/sj.bjc.6604268
  8. Bosch FX, Ribes J, Diaz M, Cleries R (2004). Primary liver cancer: worldwide incidence and trends. Gastroenterology, 127, S5-16. https://doi.org/10.1053/j.gastro.2004.09.011
  9. Buneker CK, Yu R, Deedigan L, Mohr A, Zwacka RM (2012). IFN-gamma combined with targeting of XIAP leads to increased apoptosis-sensitisation of TRAIL resistant pancreatic carcinoma cells. Cancer Lett, 316, 168-77. https://doi.org/10.1016/j.canlet.2011.10.035
  10. Caldas H, Holloway MP, Hall BM, Qualman SJ, Altura RA (2006). Survivin-directed RNA interference cocktail is a potent suppressor of tumour growth in vivo. J Med Genet, 43, 119-28.
  11. Capalbo G, Rodel C, Stauber RH, et al (2007). The role of survivin for radiation therapy. Prognostic and predictive factor and therapeutic target. Strahlenther Onkol, 183, 593-9. https://doi.org/10.1007/s00066-007-1800-4
  12. Chai J, Du C, Wu JW, et al (2000). Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature, 406, 855-62. https://doi.org/10.1038/35022514
  13. Chai J, Shiozaki E, Srinivasula SM, et al (2001). Structural basis of caspase-7 inhibition by XIAP Cell, 104, 769-80. https://doi.org/10.1016/S0092-8674(01)00272-0
  14. Chang H (2007). RNAi-mediated knockdown of target genes: a promising strategy for pancreatic cancer research. Cancer Gene Ther, 14, 677-85. https://doi.org/10.1038/sj.cgt.7701063
  15. Che Y, Ye F, Xu R, et al (2012). Co-expression of XIAP and cyclin Dl complex correlates with a poor prognosis in patients with hepatocellular carcinoma. Am J Pathol, 180, 1798-807. https://doi.org/10.1016/j.ajpath.2012.01.016
  16. Chen J,XiaoXQ, DengCM, SuXS, Li GY (2006). Downregulation of xIAP expression by small interfering RNA inhibits cellular viability and increases chemosensitivity to methotrexate in human hepatoma cell line HepG2. J Chemother, 18,525-31. https://doi.org/10.1179/joc.2006.18.5.525
  17. Chen Z, Naito M, Hori S, et al (1999). A human IAP-family gene, apollon, expressed in human brain cancer cells. Biochem Biophys Res Commun, 264, 847-54. https://doi.org/10.1006/bbrc.1999.1585
  18. Choi J, Hwang YK, Choi YJ, et al (2007). Neuronal apoptosis inhibitory protein is overexpressed in patients with unfavorable prognostic factors in breast cancer. J Korean Med Sci, 22, S17-23. https://doi.org/10.3346/jkms.2007.22.S.S17
  19. Chu L, Gu J, Sun L, et al (2008). Oncolytic adenovirus-mediated shRNA against Apollon inhibits tumor cell growth and enhances antitumor effect of 5-fluorouracil. Gene Ther, 15, 484-94. https://doi.org/10.1038/gt.2008.6
  20. Cillessen SA, Reed JC, Welsh K, et al (2008). Small-molecule XIAP antagonist restores caspase-9 mediated apoptosis in XIAP-positive diffuse large B-cell lymphoma cells. Blood, 111, 369-75. https://doi.org/10.1182/blood-2007-04-085480
  21. Cotman CW (1998). Apoptosis decision cascades and neuronal degeneration in Alzheimer's disease. Neurobiol Aging, 19, S29-32. https://doi.org/10.1016/S0197-4580(98)00042-6
  22. Couzin J (2002). Breakthrough of the year. Small RNAs make big splash. Science, 298, 2296-7. https://doi.org/10.1126/science.298.5602.2296
  23. Crnkovic-Mertens I, Muley T, Meister M, et al (2006). The anti-apoptotic livin gene is an important determinant for the apoptotic resistance of non-small cell lung cancer cells. Lung Cancer, 54, 135-42. https://doi.org/10.1016/j.lungcan.2006.07.018
  24. Crook NE, Clem RJ, Miller LK (1993). An apoptosis-inhibiting baculovirus gene with a zinc finger-like motif. J Virol, 67, 2168-74
  25. Cryns V, Yuan J (1998). Proteases to die for. Genes Dev, 12, 1551-70. https://doi.org/10.1101/gad.12.11.1551
  26. DasguptaA, Alvarado CS, Xu Z, Findley HW (2010). Expression and functional role of inhibitor-of-apoptosis protein livin (BIRC7) in neuroblastoma. Biochem Biophys Res Commun, 400, 53-9. https://doi.org/10.1016/j.bbrc.2010.08.001
  27. Davoodi J, Lin L, Kelly J, Liston P, MacKenzie AE (2004). Neuronal apoptosis-inhibitory protein does not interact with Smac and requires ATP to bind caspase-9. J Biol Chem, 279, 40622-8. https://doi.org/10.1074/jbc.M405963200
  28. Dickins RA, McJunkin K, Hernando E, et al (2007). Tissue-specific and reversible RNA interference in transgenic mice. Nat Genet, 39, 914-21. https://doi.org/10.1038/ng2045
  29. Eckelman BP, Salvesen GS, Scott FL (2006). Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep, 7, 988-94. https://doi.org/10.1038/sj.embor.7400795
  30. El-Mesallamy HO, Hegab HM, Kamal AM (2011). Expression of inhibitor of apoptosis protein (IAP) livin/BIRC7 in acute leukemia in adults: correlation with prognostic factors and outcome. Leuk Res, 35, 1616-22. https://doi.org/10.1016/j.leukres.2011.05.026
  31. Endo T, Abe S, Seidlar HB, et al (2004). Expression of IAP family proteins in colon cancers from patients with different age groups. Cancer Immunol Immunother, 53, 770-6.
  32. Esposito I, Kleeff J, Abiatari I, et al (2007). Overexpression of cellular inhibitor of apoptosis protein 2 is an early event in the progression of pancreatic cancer. J Clin Pathol, 60, 885-95.
  33. Fabregat I (2009). Dysregulation of apoptosis in hepatocellular carcinoma cells. World J Gastroenterol, 15, 513-20. https://doi.org/10.3748/wjg.15.513
  34. Fuessel S, Herrmann J, Ning S, et al (2006). Chemosensitization of bladder cancer cells by survivin-directed antisense oligodeoxynucleotides and siRNA. Cancer Lett, 232, 243-54. https://doi.org/10.1016/j.canlet.2005.02.027
  35. Gazzaniga P, Gradilone A, Giuliani L, et al (2003). Expression and prognostic significance of LIVIN, SURVIVIN and other apoptosis-related genes in the progression of superficial bladder cancer. Ann Oncol, 14, 85-90. https://doi.org/10.1093/annonc/mdg002
  36. Ge QX, Li YY, Nie YQ, Zuo WG, Du YL (2013). Expression of survivin and its four splice variants in colorectal cancer and its clinical significances. Med Oncol, 30, 535. https://doi.org/10.1007/s12032-013-0535-6
  37. Giagkousiklidis S, Vellanki SH, Debatin KM, Fulda S (2007). Sensitization of pancreatic carcinoma cells for gamma-irradiation-induced apoptosis by XIAPinhibition. Oncogene, 26,7006-16. https://doi.org/10.1038/sj.onc.1210502
  38. Giering JC, Grimm D, Storm TA, Kay MA (2008). Expression of shRNA from a tissue-specific pol II promoter is an effective and safe RNAi therapeutic. Mol Ther, 16, 1630-6. https://doi.org/10.1038/mt.2008.144
  39. Hartmann A, Hunot S, Michel PR et al (2000). Caspase-3: A vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson's disease. Proc Natl AcadSci USA, 97, 2875-80. https://doi.org/10.1073/pnas.040556597
  40. Hatano M, Mizuno M, Yoshida J (2004). Enhancement of C2-ceramide antitumor activity by small interfering RNA on X chromosome-linked inhibitor of apoptosis protein in resistant human glioma cells. J Neurosurg, 101, 119-27. https://doi.org/10.3171/jns.2004.101.1.0119
  41. Hauser HP, Bardroff M, Pyrowolakis G, Jentsch S (1998). A giant ubiquitin-conjugating enzyme related to IAP apoptosis inhibitors. J Cell Biol, 141, 1415-22. https://doi.org/10.1083/jcb.141.6.1415
  42. Hiscutt EL, Hill DS, Martin S, et al (2010). Targeting X-linked inhibitor of apoptosis protein to increase the efficacy of endoplasmic reticulum stress-induced apoptosis for melanoma therapy. J Invest Dermatol, 130, 2250-8. https://doi.org/10.1038/jid.2010.146
  43. Huang Y, Park YC, Rich RL, et al (2001). Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell, 104, 781-90.
  44. Hung CS, Lin SF, Liu HH, et al (2012). Survivin-mediated therapeutic efficacy of gemcitabine through glucose-regulated protein 78 in hepatocellular carcinoma. Ann Surg Oncol, 19, 2744-52. https://doi.org/10.1245/s10434-011-2188-z
  45. Izquierdo M (2005). Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Ther, 12, 217-27. https://doi.org/10.1038/sj.cgt.7700791
  46. Karagiannis TC, El-Osta A (2005). RNA interference and potential therapeutic applications of short interfering RNAs. Cancer Gene Ther, 12, 787-95. https://doi.org/10.1038/sj.cgt.7700857
  47. Kasof GM, Gomes BC (2001). Livin, a novel inhibitor of apoptosis protein family member. J Biol Chem, 276, 3238-46. https://doi.org/10.1074/jbc.M003670200
  48. Kempkensteffen C, Fritzsche FR, Johannsen M, et al (2009). Down-regulation of the pro-apoptotic XIAP associated factor-1 (XAF1) during progression of clear-cell renal cancer. BMC Cancer, 9, 276. https://doi.org/10.1186/1471-2407-9-276
  49. Kesari A, Misra UK, Kalita J, et al (2005). Study of survival of motor neuron (SMN) and neuronal apoptosis inhibitory protein (NAIP) gene deletions in SMA patients. J Neurol, 252, 667-71. https://doi.org/10.1007/s00415-005-0714-2
  50. Kroemer G, Reed JC (2000). Mitochondrial control of cell death. Nat Med, 6, 513-9. https://doi.org/10.1038/74994
  51. LaCasse EC, Mahoney DJ, Cheung HH, et al (2008). IAP-targeted therapies for cancer. Oncogene, 27, 6252-75. https://doi.org/10.1038/onc.2008.302
  52. Lazar I, Perlman R, Lotem M, et al (2012). The clinical effect of the inhibitor of apopotosis protein livin in melanoma. Oncology, 82, 197-204. https://doi.org/10.1159/000334234
  53. Li F (2003). Survivin study: what is the next wave? J Cell Physiol, 197, 8-29. https://doi.org/10.1002/jcp.10327
  54. Li F (2005). Role of survivin and its splice variants in tumorigenesis. Br J Cancer, 92, 212-6. https://doi.org/10.1038/sj.bjc.6602340
  55. Li SX, Chai L, Cai ZG, et al (2012). Expression of survivin and caspase 3 in oral squamous cell carcinoma and peritumoral tissue. Asian Pac J Cancer Prev, 13, 5027-31. https://doi.org/10.7314/APJCP.2012.13.10.5027
  56. Lima RT, Martins LM, Guimaraes JE, Sambade C, Vasconcelos MH (2004). Specific downregulation of bcl-2 and xIAP by RNAi enhances the effects of chemotherapeutic agents in MCF-7 human breast cancer cells. Cancer Gene Ther, 11, 309-16. https://doi.org/10.1038/sj.cgt.7700706
  57. Lin JH, Deng G, Huang Q, Morser J (2000). KIAP, a novel member of the inhibitor of apoptosis protein family. Biochem Biophys Res Commun, 279, 820-31. https://doi.org/10.1006/bbrc.2000.4027
  58. Liu B, Han M, Wen JK, Wang L (2007). Livin/ML-IAP as a new target for cancer treatment. Cancer Lett, 250, 168-76. https://doi.org/10.1016/j.canlet.2006.09.024
  59. Liu H, Wang S, Sun H, et al (2010). Inhibition of tumorigenesis and invasion of hepatocellular carcinoma by siRNA-mediated silencing of the livin gene. Mol Med Rep, 3, 903-7.
  60. Liu HB, Kong CZ, Zeng Y, et al (2009a). Livin may serve as a marker for prognosis of bladder cancer relapse and a target of bladder cancer treatment. Urol Oncol, 27, 277-83. https://doi.org/10.1016/j.urolonc.2008.03.015
  61. Liu W, Zhu F, Jiang Y, et al (2013). siRNA targeting survivin inhibits the growth and enhances the ch,mosensitivity of hepatocellular carcinoma cells. Oncol Rep, 29, 1183-8. https://doi.org/10.3892/or.2012.2196
  62. Liu WS, Yan HJ, Qin RY, et al (2009b). siRNA directed against survivin enhances pancreatic cancer cell gemcitabine chemosensitivity. Dig Dis Sci, 54, 89-96. https://doi.org/10.1007/s10620-008-0329-4
  63. Liu Z, Sun C, Olejniczak ET, et al (2000). Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature, 408, 1004-8. https://doi.org/10.1038/35050006
  64. Llovet JM, Bruix J (2008). Novel advancements in the management of hepatocellular carcinoma in 2008. J Hepatol, 48, S20-37.
  65. Lopergolo A, Pennati M, Gandellini P, et al (2009). Apollon gene silencing induces apoptosis in breast cancer cells through p53 stabilisation and caspase-3 activation. Br J Cancer, 100, 739-46.
  66. Ma JJ, Chen BL, Xin XY (2009). XIAP gene downregulation by small interfering RNA inhibits proliferation, induces apoptosis, reverses the cisplatin resistance of ovarian carcinoma. Eur J Obstet Gynecol Reprod Biol, 146, 222-6. https://doi.org/10.1016/j.ejogrb.2009.06.011
  67. Maier JK, Lahoua Z, Gendron NH, et al (2002). The neuronal apoptosis inhibitory protein is a direct inhibitor of caspases 3 and 7. J Neurosci, 22, 2035-43.
  68. Mansouri A, Zhang Q, Ridgway LD, Tian L, Claret FX (2003). Cisplatin resistance in an ovarian carcinoma is associated with a defect in programmed cell death control through XIAP regulation. Oncol Res, 13, 399-404. https://doi.org/10.3727/096504003108748410
  69. Merry DE, Korsmeyer SJ (1997). Bcl-2 gene family in the nervous system. Annu Rev Neurosci, 20, 245-67. https://doi.org/10.1146/annurev.neuro.20.1.245
  70. Notarbartolo M, Cervello M, Dusonchet L, D'Alessandro N (2002). NAIP-deltaEx10-11: a novel splice variant of the apoptosis inhibitor NAIP differently expressed in drug-sensitive and multidrug-resistant HL60 leukemia cells. Leuk Res, 26, 857-62. https://doi.org/10.1016/S0145-2126(02)00016-4
  71. Notarbartolo M, Cervello M, Giannitrapani L, et al (2004). Expression of IAPs and alternative splice variants in hepatocellular carcinoma tissues and cells. Ann N Y Acad Sci, 1028, 289-93. https://doi.org/10.1196/annals.1322.033
  72. Oh BY, Lee RA, Kim KH (2011). siRNA targeting Livin decreases tumor in a xenograft model for colon cancer. World J Gastroenterol, 17, 2563-71. https://doi.org/10.3748/wjg.v17.i20.2563
  73. Qiu XB, Markant SL, Yuan J, Goldberg AL (2004). Nrdpl-mediated degradation of the gigantic IAP, BRUCE, is a novel pathway for triggering apoptosis. EMBO J, 23,800-10. https://doi.org/10.1038/sj.emboj.7600075
  74. Ramp U, Krieg T, Caliskan E, et al (2004). XIAP expression is an independent prognostic marker in clear-cell renal carcinomas. Hum Pathol, 35, 1022-8. https://doi.org/10.1016/j.humpath.2004.03.011
  75. Ren J, Shi M, Liu R, et al (2005). The Birc6 (Bruce) gene regulates p53 and the mitochondrial pathway of apoptosis and is essential for mouse embryonic development. Proc Natl Acad Sci USA, 102, 565-70. https://doi.org/10.1073/pnas.0408744102
  76. Richter BW, Mir SS, Eiben LJ, et al (2001). Molecular cloning of ILP-2, a novel member of the inhibitor of apoptosis protein family. Mol Cell Biol, 21, 4292-301. https://doi.org/10.1128/MCB.21.13.4292-4301.2001
  77. Rothe M, Pan MG, Henzel WJ, Ayres TM, Goeddel DV (1995). The TNFR2-TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins. Cell, 83, 1243-52. https://doi.org/10.1016/0092-8674(95)90149-3
  78. Ruckert F, Samm N, Lehner AK, et al (2010). Simultaneous gene silencing of Bcl-2, XIAP and Survivin re-sensitizes pancreatic cancer cells towards apoptosis. BMC Cancer, 10, 379. https://doi.org/10.1186/1471-2407-10-379
  79. Ryan BM, Konecny GE, Kahlert S, et al (2006). Survivin expression in breast cancer predicts clinical outcome and is associated with HER2, VEGF, urokinase plasminogen activator and PAI-l. Ann Oncol, 17, 597-604. https://doi.org/10.1093/annonc/mdj121
  80. Salvesen GS, Duckett CS (2002). IAP proteins: blocking the road to death's door. Nat Rev Mol Cell Biol, 3, 401-10. https://doi.org/10.1038/nrm830
  81. Shankar P, Manjunath N, Lieberman J (2005). The prospect of silencing disease using RNA interference. JAMA, 293, 1367-73. https://doi.org/10.1001/jama.293.11.1367
  82. Shi Y (2002). Mechanisms of caspase activation and inhibition during apoptosis. Mol Cell, 9, 459-70. https://doi.org/10.1016/S1097-2765(02)00482-3
  83. Shi YH, Ding WX, Zhou J, et al (2008). Expression of X-linked inhibitor-of-apoptosis protein in hepatocellular carcinoma promotes metastasis and tumor recurrence. Hepatology, 48, 497-507. https://doi.org/10.1002/hep.22393
  84. Shiozaki EN, Chai J, Rigotti DJ, et al (2003). Mechanism of XIAP-mediated inhibition of caspase-9. Mol Cell, 11, 519-27. https://doi.org/10.1016/S1097-2765(03)00054-6
  85. Shiraki K, Sugimoto K, Yamanaka Y, et al (2003). Overexpression of X-linked inhibitor of apoptosis in human hepatocellular carcinoma. Int J Mol Med, 12, 705-8.
  86. Shrikhande SV, Kleeff J, Kayed H, et al (2006). Silencing of X-linked inhibitor of apoptosis (XIAP) decreases gemcitabine resistance of pancreatic cancer cells. Anticancer Res, 26, 3265-73.
  87. Smith CA, Farrah T, Goodwin RG (1994). The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, death. Cell, 76, 959-62. https://doi.org/10.1016/0092-8674(94)90372-7
  88. Song X, Wang JB, Yin DL, et al (2009). Down-regulation of lung resistance related protein by RNA interference targeting survivin induces the reversal of chemoresistances in hepatocellular carcinoma. Chin Med J (Engl), 122, 2636-42.
  89. Sprenger T, Rodel F, Beissbarth T, et al (2011). Failure of downregulation of survivin following neoadjuvant radiochemotherapy in rectal cancer is associated with distant metastases and shortened survival. Clin Cancer Res, 17, 1623-31. https://doi.org/10.1158/1078-0432.CCR-10-2592
  90. Srinivasula SM, Hegde R, Saleh A, et al (2001). A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature, 410, 112-6. https://doi.org/10.1038/35065125
  91. Sung KW, Choi J, Hwang YK, et al (2007). Overexpression of Apollon, an antiapoptotic protein, is associated with poor prognosis in childhood de novo acute myeloid leukemia. Clin Cancer Res, 13, 5109-14. https://doi.org/10.1158/1078-0432.CCR-07-0693
  92. Takashima H, Nakajima T, Moriguchi M, et al (2005). In vivo expression patterns of survivin and its splicing variants in chronic liver disease and hepatocellular carcinoma. Liver Int, 25, 77-84. https://doi.org/10.1111/j.1478-3231.2004.0979.x
  93. Takeuchi H, Kim J, Fujimoto A, et al (2005a). X-Linked inhibitor of apoptosis protein expression level in colorectal cancer is regulated by hepatocyte growth factor/C-met pathway via Akt signaling. Clin Cancer Res, 11, 7621-8.
  94. Takeuchi H, Morton DL, Elashoff D, Hoon DS (2005b). Survivin expression by metastatic melanoma predicts poor disease outcome in patients receiving adjuvant polyvalent vaccine. Int J Cancer, 117, 1032-8. https://doi.org/10.1002/ijc.21267
  95. Tanabe H, Yagihashi A, Tsuji N, et al (2004). Expression of survivin mRNA and livin mRNA in non-small-cell lung cancer. Lung Cancer, 46, 299-304 https://doi.org/10.1016/j.lungcan.2004.05.002
  96. Tartaglia LA, Goeddel DV (1992a). Tumor necrosis factor receptor signaling. A dominant negative mutation suppresses the activation of the 55-kDa tumor necrosis factor receptor. J Biol Chem, 267, 4304-7.
  97. Tartaglia LA, Goeddel DV (1992b). Two TNF receptors. Immunol Today, 13, 151-3.
  98. Thornberry NA, Lazebnik Y (1998). Caspases: enemies within. Science, 281, 1312-6. https://doi.org/10.1126/science.281.5381.1312
  99. Varfolomeev E, Goncharov T, Fedorova AV, et al (2008). c-IAPl and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. J Biol Chem, 283, 24295-9. https://doi.org/10.1074/jbc.C800128200
  100. Vucic D, Deshayes K, Ackerly H, et al (2002). SMAC negatively regulates the anti-apoptotic activity of melanoma inhibitor of apoptosis (ML-IAP). J Biol Chem, 277, 12275-9. https://doi.org/10.1074/jbc.M112045200
  101. Vucic D, Fairbrother WJ (2007). The inhibitor of apoptosis proteins as therapeutic targets in cancer. Clin Cancer Res, 13, 5995-6000. https://doi.org/10.1158/1078-0432.CCR-07-0729
  102. Vucic D, Franklin MC, Wallweber HJ, et al (2005). Engineering ML-IAP to produce an extraordinarily potent caspase 9 inhibitor: implications for Smac-dependent anti-apoptotic activity of ML-IAP Biochem J, 385, 11-20. https://doi.org/10.1042/BJ20041108
  103. Vucic D, Stennicke HR, Pisabarro MT, Salvesen GS, Dixit VM (2000). ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed in human melanomas. Curr Biol, 10, 1359-66. https://doi.org/10.1016/S0960-9822(00)00781-8
  104. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS, Jr. (1998). NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAPl and C-IAP2 to suppress caspase-8 activation. Science, 281, 1680-3. https://doi.org/10.1126/science.281.5383.1680
  105. Wang H, Tan SS, Wang XY, et al (2007). Silencing livin gene by siRNA leads to apoptosis induction, cell cycle arrest, proliferation inhibition in malignant melanoma LiBr cells. Acta Pharmacol Sin, 28, 1968-74. https://doi.org/10.1111/j.1745-7254.2007.00724.x
  106. Wang S, Bai L, Lu J, et al (2012). Targeting inhibitors of apoptosis proteins (IAPs) for new breast cancer therapeutics. J Mammary Gland Biol Neoplasia, 17, 217-28. https://doi.org/10.1007/s10911-012-9265-1
  107. Wang TS, Ding QQ, Guo RH, et al (2010). Expression of livin in gastric cancer and induction of apoptosis in SGC-7901 cells by shRNA-mediated silencing of livin gene. Biomed Pharmacother, 64, 333-8. https://doi.org/10.1016/j.biopha.2009.06.002
  108. Wei W, Wanjun L, Hui S, et al (2013). miR-203 inhibits proliferation of HCC cells by targeting survivin. Cell Biochem Fund, 31, 82-5. https://doi.org/10.1002/cbf.2863
  109. Wei Y, Fan T, Yu M (2008). Inhibitor of apoptosis proteins and apoptosis. Acta Biochim Biophys Sin (Shanghai), 40,278-88. https://doi.org/10.1111/j.1745-7270.2008.00407.x
  110. Wong LL (2002). Current status of liver transplantation for hepatocellular cancer. Am J Surg, 183, 309-16. https://doi.org/10.1016/S0002-9610(02)00785-7
  111. Wu G, Chai J, Suber TL, et al (2000). Structural basis of IAP recognition by Smac/DIABLO. Nature, 408, 1008-12. https://doi.org/10.1038/35050012
  112. Wu L, Wang Y, Tian D (2007). Knockdown of survivin expression by siRNA induces apoptosis of hepatocellular carcinoma cells. J Huazhong Unix Sci Technolog Med Sci, 27, 403-6. https://doi.org/10.1007/s11596-007-0413-6
  113. Xiang Y, Yao H, Wang S, et al (2006). Prognostic value of Survivin and Livin in nasopharyngeal carcinoma. Laryngoscope, 116, 126-30. https://doi.org/10.1097/01.mlg.0000187392.87904.35
  114. Yahya RS, Fouda MI, El-Baz HA, Mosa TE, Elmaksoud MD (2012). Serum Survivin and TP53 Gene Expression in Children with Acute Lymphoblastic Leukemia. Iran J Public Health, 41, 37-44.
  115. Yang D, Song X, Zhang J, et al (2010). Suppression of livin gene expression by siRNA leads to growth inhibition and apoptosis induction in human bladder cancer T24 cells. Biosci Biotechnol Biochem, 74, 1039-44. https://doi.org/10.1271/bbb.90934
  116. Yang H, Fu JH, Hu Y, et al (2008). Influence of SiRNA targeting survivin on chemosensitivity of H460/cDDP lung cancer cells. J Int Med Res, 36, 734-47. https://doi.org/10.1177/147323000803600416
  117. Yang M, Liu Y, Lu S, et al (2013). Analysis of the expression levels of survivin and VEGF in patients with acute lymphoblastic leukemia. Exp Ther Med, 5, 305-7. https://doi.org/10.3892/etm.2012.769
  118. Yuan B, Ran B, Wang S, et al (2012). siRNA directed against Livin inhibits tumor growth and induces apoptosis in human glioma cells. J Neurooncol, 107, 81-7. https://doi.org/10.1007/s11060-011-0728-9
  119. Yuan D, Liu L, Xu H, Gu D (2009). The effects on cell growth and chemosensitivity by livin RNAi in non-small cell lung cancer. Mol Cell Biochem, 320, 133-40. https://doi.org/10.1007/s11010-008-9915-1
  120. Zaffaroni N, Pennati M, Daidone MG (2005). Survivin as a target for new anticancer interventions. J Cell Mol Med, 9, 360-72. https://doi.org/10.1111/j.1582-4934.2005.tb00361.x
  121. Zender L, Spector MS, Xue W, et al (2006). Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell, 125, 1253-67. https://doi.org/10.1016/j.cell.2006.05.030
  122. Zhang M, Yang J, Li F (2006). Transcriptional and post- transcriptional controls of survivin in cancer cells: novel approaches for cancer treatment. J Exp Clin Cancer Res, 25, 391-402.
  123. Zhang R, Ma L, Zheng M, et al (2010). Survivin knockdown by short hairpin RNA abrogates the growth of human hepatocellular carcinoma xenografts in nude mice. Cancer Gene Ther, 17, 275-88. https://doi.org/10.1038/cgt.2009.68
  124. Zhang S, Ding F, Luo A, et al (2007). XIAP is highly expressed in esophageal cancer and its downregulation by RNAi sensitizes esophageal carcinoma cell lines to chemotherapeutics. Cancer Biol Ther, 6, 973-80.
  125. Zhu H, Chen XP, Zhang WG, Luo SF, Zhang BX (2005). Expression and significance of new inhibitor of apoptosis protein survivin in hepatocellular carcinoma. World J Gastroenterol, 11, 3855-9. https://doi.org/10.3748/wjg.v11.i25.3855

Cited by

  1. Rutin mediated targeting of signaling machinery in cancer cells vol.14, pp.1, 2014, https://doi.org/10.1186/s12935-014-0124-6
  2. BCR/ABL mRNA Targeting Small Interfering RNA Effects on Proliferation and Apoptosis in Chronic Myeloid Leukemia vol.15, pp.12, 2014, https://doi.org/10.7314/APJCP.2014.15.12.4773
  3. Down-Regulation of Mcl-1 by Small Interference RNA Induces Apoptosis and Sensitizes HL-60 Leukemia Cells to Etoposide vol.15, pp.2, 2014, https://doi.org/10.7314/APJCP.2014.15.2.629
  4. Prognostic Significance of Survivin in Breast Cancer: Meta-analysis vol.20, pp.5, 2014, https://doi.org/10.1111/tbj.12303
  5. Theranostic Properties of a Survivin-Directed Molecular Beacon in Human Melanoma Cells vol.9, pp.12, 2014, https://doi.org/10.1371/journal.pone.0114588
  6. Inflammatory microenvironment and expression of chemokines in hepatocellular carcinoma vol.21, pp.16, 2015, https://doi.org/10.3748/wjg.v21.i16.4864
  7. Targeted silencing of CXCL1 by siRNA inhibits tumor growth and apoptosis in hepatocellular carcinoma vol.47, pp.6, 2015, https://doi.org/10.3892/ijo.2015.3203
  8. Peridinin, a carotenoid, inhibits proliferation and survival of HTLV-1-infected T-cell lines vol.49, pp.4, 2016, https://doi.org/10.3892/ijo.2016.3648
  9. Chemokine CXCL1 may serve as a potential molecular target for hepatocellular carcinoma vol.5, pp.10, 2016, https://doi.org/10.1002/cam4.843
  10. Inducing death in tumor cells: roles of the inhibitor of apoptosis proteins vol.6, pp.2046-1402, 2017, https://doi.org/10.12688/f1000research.10625.1
  11. Evaluation of Antimetastatic Effect of lncRNA-ATB siRNA Delivered Using Ultrasound-Targeted Microbubble Destruction vol.35, pp.8, 2016, https://doi.org/10.1089/dna.2016.3254
  12. Decreased expression of the augmenter of liver regeneration results in growth inhibition and increased chemosensitivity of acute T lymphoblastic leukemia cells vol.38, pp.5, 2017, https://doi.org/10.3892/or.2017.5984
  13. The SMAC Mimetic APG-1387 Sensitizes Immune-Mediated Cell Apoptosis in Hepatocellular Carcinoma vol.9, pp.1663-9812, 2018, https://doi.org/10.3389/fphar.2018.01298
  14. Small interfering RNA-mediated gene suppression as a therapeutic intervention in hepatocellular carcinoma pp.00219541, 2018, https://doi.org/10.1002/jcp.27015