67
Views
2
CrossRef citations to date
0
Altmetric
Original Research

AATF is Overexpressed in Human Bladder Cancer and Regulates Chemo-Sensitivity Through Survivin

, &
Pages 5493-5505 | Published online: 29 Dec 2021

References

  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.
  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30.
  • Fujita N, Hatakeyama S, Momota M, et al. Preoperative chronic kidney disease predicts poor prognosis in patients with primary non-muscle-invasive bladder cancer who underwent transurethral resection of bladder tumor. Urol Oncol. 2020;38:684e1- e8.
  • Yin M, Joshi M, Meijer RP, et al. Neoadjuvant chemotherapy for muscle-invasive bladder cancer: a systematic review and two-step meta-analysis. Oncologist. 2016;21:708–715.
  • Dong Q, Fu L, Zhao Y, Tan S, Wang E. Derlin-1 overexpression confers poor prognosis in muscle invasive bladder cancer and contributes to chemoresistance and invasion through PI3K/AKT and ERK/MMP signaling. Oncotarget. 2017;8:17059–17069.
  • Srinivas AN, Suresh D, Mirshahi F, Santhekadur PK, Sanyal AJ, Kumar DP. Emerging roles of AATF: checkpoint signaling and beyond. J Cell Physiol. 2021;236:3383–3395.
  • Bruno T, Iezzi S, De Nicola F, et al. Che-1 activates XIAP expression in response to DNA damage. Cell Death Differ. 2008;15:515–520.
  • Kumar DP, Santhekadur PK, Seneshaw M, et al. Role of apoptosis antagonizing transcription factor in the pathogenesis of nonalcoholic fatty liver disease and hepatocellular carcinoma. Hepatology. 2019;69:1520–1534.
  • Iezzi S, Fanciulli M. Discovering Che-1/AATF: a new attractive target for cancer therapy. Front Genet. 2015;6:141.
  • Bruno T, Iezzi S, Fanciulli M. Che-1/AATF: a critical cofactor for both wild-type- and Mutant-p53 proteins. Front Oncol. 2016;6:34.
  • Hopker K, Hagmann H, Khurshid S, et al. AATF/Che-1 acts as a phosphorylation-dependent molecular modulator to repress p53-driven apoptosis. EMBO J. 2012;31:3961–3975.
  • Bruno T, De Nicola F, Corleone G, et al. Che-1/AATF-induced transcriptionally active chromatin promotes cell proliferation in multiple myeloma. Blood Adv. 2020;4:5616–5630.
  • Remmele W, Stegner HE. [Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue]. Der Pathologe. 1987;8:138–140. German.
  • Fedchenko N, Reifenrath J. Different approaches for interpretation and reporting of immunohistochemistry analysis results in the bone tissue - a review. Diagn Pathol. 2014;9:221.
  • Liu J, Lu J, Ma Z, Li W. A nomogram based on a three-gene signature derived from AATF coexpressed genes predicts overall survival of hepatocellular carcinoma patients. Biomed Res Int. 2020;2020:7310768.
  • Pang W, Li Y, Guo W, Shen H. Cyclin E: a potential treatment target to reverse cancer chemoresistance by regulating the cell cycle. Am J Transl Res. 2020;12:5170–5187.
  • Guerra F, Arbini AA, Moro L. Mitochondria and cancer chemoresistance. Biochim Biophys Acta. 2017;1858:686–699.
  • Kim JS, Lee JM, Chwae YJ, et al. Cisplatin-induced apoptosis in Hep3B cells: mitochondria-dependent and -independent pathways. Biochem Pharmacol. 2004;67:1459–1468.
  • Zhao W, You CC, Zhuang JP, et al. Viability inhibition effect of gambogic acid combined with cisplatin on osteosarcoma cells via mitochondria-independent apoptotic pathway. Mol Cell Biochem. 2013;382:243–252.
  • Li H, Fu L, Liu B, Lin X, Dong Q, Wang E. Ajuba overexpression regulates mitochondrial potential and glucose uptake through YAP/Bcl-xL/GLUT1 in human gastric cancer. Gene. 2019;693:16–24.
  • Ow YP, Green DR, Hao Z, Mak TW. Cytochrome c: functions beyond respiration. Nat Rev Mol Cell Biol. 2008;9:532–542.
  • Chen X, Wong JY, Wong P, Radany EH. Low-dose valproic acid enhances radiosensitivity of prostate cancer through acetylated p53-dependent modulation of mitochondrial membrane potential and apoptosis. Mol Cancer Res. 2011;9:448–461.
  • Waligorska-Stachura J, Jankowska A, Wasko R, et al. Survivin–prognostic tumor biomarker in human neoplasms–review. Ginekol Pol. 2012;83:537–540.
  • Liu JL, Gao W, Kang QM, Zhang XJ, Yang SG. Prognostic value of survivin in patients with gastric cancer: a systematic review with meta-analysis. PLoS One. 2013;8:e71930.
  • Sattari M, Pazhang Y, Imani M. Calprotectin induces cell death in human prostate cancer cell (LNCaP) through survivin protein alteration. Cell Biol Int. 2014;38:1311–1320.
  • Akhtar M, Gallagher L, Rohan S. Survivin: role in diagnosis, prognosis, and treatment of bladder cancer. Adv Anat Pathol. 2006;13:122–126.
  • Krafft U, Tschirdewahn S, Hess J, et al. Validation of survivin and HMGA2 as biomarkers for cisplatin resistance in bladder cancer. Urol Oncol. 2019;37:810e7- e15.