Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

RETRACTED ARTICLE: MNAT1 is overexpressed in colorectal cancer and mediates p53 ubiquitin-degradation to promote colorectal cancer malignance

Authors: Shan Zhou, Jinping Lu, Yuejin Li, Chan Chen, Yongqiang Cai, Gongjun Tan, Zhengke Peng, Zhenlin Zhang, Zigang Dong, Tiebang Kang, Faqing Tang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

MNAT1 (menage a trois 1, MAT1), a cyclin-dependent kinase-activating kinase (CAK) complex, high expresses in various cancers and is involved in cancer pathogenesis. However, mechanisms underlying its regulation in carcinogenesis are unclear.

Methods

The tissue microarray of colorectal cancer (CRC) was used to evaluate MNAT1 expressions in CRC tissues using immunohistochemistry, CRC cell lines were also detected MNAT1 expression using Western-blotting. MNAT1 and shMNAT1 vectors were constructed, and transfected into CRC cells. Cell growths of the transfected cells were observed using MTT and colony formation. The affects of MNAT1 on p53 expression were analyzed using Western-blotting and Real-time PCR. Immunoprecipitation assay was used to analyze the interaction p53 and MNAT1, and Western-blotting was used to test the effects of MNAT1 on p53 downstream molecules. The apoptosis of CRC cells with MNAT1 or shMNAT1 were analyzed using flow cytometry. BABL/c athymic nude mice were used to observe the effect of MNAT1 on CRC cell growth in vivo.

Results

MNAT1 was found to be overexpressed in CRC tissues and cells, and MNAT1 expressions in CRC tissue samples were associated with CRC carcinogenesis and poor patient outcomes. MNAT1-knockin increased CRC cell growth and colony formation, and MNAT1-knockdown dramatically decreased cell motility and invasion. MNAT1 physically interacted with p53, MNAT1 also increased the interaction of MDM2 with p53. Strikingly, MNAT1 mediated p53 ubiquitin-degradation. MNAT1 shortened p53 half-life, and ectopic MNAT1 expression decreased p53 protein stability. Moreover, MNAT1 induced RAD51 and reduced p21, cleaved-caspase3, cleaved-PARP and BAX expression. MNAT1 inhibited CRC cell apoptosis. shMANT1 decreased tumor growths in nude mice following p53 increase.

Conclusion

MNAT1 binds to p53, mediates p53 ubiquitin-degradation through MDM2, increases cell growth and decreases cell apoptosis, and finally promotes CRC malignance. MNAT1 binding to p53 and mediating p53 ubiquitin-degradation axis represents a novel molecular joint in the p53 pathway.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jemal A, et al. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.PubMed Jemal A, et al. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.PubMed
2.
go back to reference Pancione M, Remo A, Colantuoni V. Genetic and epigenetic events generate multiple pathways in colorectal cancer progression. Patholog Res Int. 2012;509348(10):24. Pancione M, Remo A, Colantuoni V. Genetic and epigenetic events generate multiple pathways in colorectal cancer progression. Patholog Res Int. 2012;509348(10):24.
3.
go back to reference Ewing I, et al. The molecular genetics of colorectal cancer. Frontline Gastroenterol. 2014;5(1):26–30.PubMed Ewing I, et al. The molecular genetics of colorectal cancer. Frontline Gastroenterol. 2014;5(1):26–30.PubMed
4.
go back to reference Vogelstein B, et al. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319(9):525–32.PubMed Vogelstein B, et al. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319(9):525–32.PubMed
5.
go back to reference Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474(7351):318–26.PubMed Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474(7351):318–26.PubMed
6.
go back to reference Markowitz SD, Bertagnolli MM. Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med. 2009;361(25):2449–60.PubMedPubMedCentral Markowitz SD, Bertagnolli MM. Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med. 2009;361(25):2449–60.PubMedPubMedCentral
7.
go back to reference Najdi R, Holcombe RF, Waterman ML. Wnt signaling and colon carcinogenesis: beyond APC. J Carcinog. 2011;10(5):1477–3163. Najdi R, Holcombe RF, Waterman ML. Wnt signaling and colon carcinogenesis: beyond APC. J Carcinog. 2011;10(5):1477–3163.
8.
9.
go back to reference Devault A, et al. MAT1 (‘menage a trois’) a new RING finger protein subunit stabilizing cyclin H-cdk7 complexes in starfish and Xenopus CAK. EMBO J. 1995;14(20):5027–36.PubMedPubMedCentral Devault A, et al. MAT1 (‘menage a trois’) a new RING finger protein subunit stabilizing cyclin H-cdk7 complexes in starfish and Xenopus CAK. EMBO J. 1995;14(20):5027–36.PubMedPubMedCentral
10.
go back to reference Shiekhattar R, et al. Cdk-activating kinase complex is a component of human transcription factor TFIIH. Nature. 1995;374(6519):283–7.PubMed Shiekhattar R, et al. Cdk-activating kinase complex is a component of human transcription factor TFIIH. Nature. 1995;374(6519):283–7.PubMed
11.
go back to reference Tassan JP, et al. In vitro assembly of a functional human CDK7-cyclin H complex requires MAT1, a novel 36 kDa RING finger protein. EMBO J. 1995;14(22):5608–17.PubMedPubMedCentral Tassan JP, et al. In vitro assembly of a functional human CDK7-cyclin H complex requires MAT1, a novel 36 kDa RING finger protein. EMBO J. 1995;14(22):5608–17.PubMedPubMedCentral
12.
go back to reference Yee A, et al. Molecular cloning of CDK7-associated human MAT1, a cyclin-dependent kinase-activating kinase (CAK) assembly factor. Cancer Res. 1995;55(24):6058–62.PubMed Yee A, et al. Molecular cloning of CDK7-associated human MAT1, a cyclin-dependent kinase-activating kinase (CAK) assembly factor. Cancer Res. 1995;55(24):6058–62.PubMed
13.
14.
go back to reference Ko LJ, et al. p53 is phosphorylated by CDK7-cyclin H in a p36MAT1-dependent manner. Mol Cell Biol. 1997;17(12):7220–9.PubMedPubMedCentral Ko LJ, et al. p53 is phosphorylated by CDK7-cyclin H in a p36MAT1-dependent manner. Mol Cell Biol. 1997;17(12):7220–9.PubMedPubMedCentral
15.
go back to reference Inamoto S, et al. The cyclin-dependent kinase-activating kinase (CAK) assembly factor, MAT1, targets and enhances CAK activity on the POU domains of octamer transcription factors. J Biol Chem. 1997;272(47):29852–8.PubMed Inamoto S, et al. The cyclin-dependent kinase-activating kinase (CAK) assembly factor, MAT1, targets and enhances CAK activity on the POU domains of octamer transcription factors. J Biol Chem. 1997;272(47):29852–8.PubMed
16.
go back to reference Morgan DO. Principles of CDK regulation. Nature. 1995;374(6518):131–4.PubMed Morgan DO. Principles of CDK regulation. Nature. 1995;374(6518):131–4.PubMed
17.
go back to reference Wu L, et al. RNA antisense abrogation of MAT1 induces G1 phase arrest and triggers apoptosis in aortic smooth muscle cells. J Biol Chem. 1999;274(9):5564–72.PubMed Wu L, et al. RNA antisense abrogation of MAT1 induces G1 phase arrest and triggers apoptosis in aortic smooth muscle cells. J Biol Chem. 1999;274(9):5564–72.PubMed
18.
19.
go back to reference Zhang S, et al. MAT1-modulated cyclin-dependent kinase-activating kinase activity cross-regulates neuroblastoma cell G1 arrest and neurite outgrowth. Cancer Res. 2004;64(9):2977–83.PubMed Zhang S, et al. MAT1-modulated cyclin-dependent kinase-activating kinase activity cross-regulates neuroblastoma cell G1 arrest and neurite outgrowth. Cancer Res. 2004;64(9):2977–83.PubMed
20.
go back to reference Busso D, et al. Distinct regions of MAT1 regulate cdk7 kinase and TFIIH transcription activities. J Biol Chem. 2000;275(30):22815–23.PubMed Busso D, et al. Distinct regions of MAT1 regulate cdk7 kinase and TFIIH transcription activities. J Biol Chem. 2000;275(30):22815–23.PubMed
21.
go back to reference Wang J, et al. Retinoid-induced G1 arrest and differentiation activation are associated with a switch to cyclin-dependent kinase-activating kinase hypophosphorylation of retinoic acid receptor alpha. J Biol Chem. 2002;277(45):43369–76.PubMed Wang J, et al. Retinoid-induced G1 arrest and differentiation activation are associated with a switch to cyclin-dependent kinase-activating kinase hypophosphorylation of retinoic acid receptor alpha. J Biol Chem. 2002;277(45):43369–76.PubMed
22.
go back to reference Wang JG, et al. Retinoic acid induces leukemia cell G1 arrest and transition into differentiation by inhibiting cyclin-dependent kinase-activating kinase binding and phosphorylation of PML/RARalpha. FASEB J. 2006;20(12):2142–4.PubMed Wang JG, et al. Retinoic acid induces leukemia cell G1 arrest and transition into differentiation by inhibiting cyclin-dependent kinase-activating kinase binding and phosphorylation of PML/RARalpha. FASEB J. 2006;20(12):2142–4.PubMed
23.
go back to reference Luo P, et al. Intrinsic retinoic acid receptor alpha-cyclin-dependent kinase-activating kinase signaling involves coordination of the restricted proliferation and granulocytic differentiation of human hematopoietic stem cells. Stem Cells. 2007;25(10):2628–37.PubMed Luo P, et al. Intrinsic retinoic acid receptor alpha-cyclin-dependent kinase-activating kinase signaling involves coordination of the restricted proliferation and granulocytic differentiation of human hematopoietic stem cells. Stem Cells. 2007;25(10):2628–37.PubMed
24.
go back to reference Lou S, et al. The lost intrinsic fragmentation of MAT1 protein during granulopoiesis promotes the growth and metastasis of leukemic myeloblasts. Stem Cells. 2013;31(9):1942–53.PubMedPubMedCentral Lou S, et al. The lost intrinsic fragmentation of MAT1 protein during granulopoiesis promotes the growth and metastasis of leukemic myeloblasts. Stem Cells. 2013;31(9):1942–53.PubMedPubMedCentral
25.
go back to reference Patel H, et al. Expression of CDK7, cyclin H, and MAT1 is elevated in breast Cancer and is prognostic in estrogen receptor-positive breast Cancer. Clin Cancer Res. 2016;22(23):5929–38.PubMedPubMedCentral Patel H, et al. Expression of CDK7, cyclin H, and MAT1 is elevated in breast Cancer and is prognostic in estrogen receptor-positive breast Cancer. Clin Cancer Res. 2016;22(23):5929–38.PubMedPubMedCentral
26.
go back to reference Hong J, et al. CHK1 targets spleen tyrosine kinase (L) for proteolysis in hepatocellular carcinoma. J Clin Invest. 2012;122(6):2165–75.PubMedPubMedCentral Hong J, et al. CHK1 targets spleen tyrosine kinase (L) for proteolysis in hepatocellular carcinoma. J Clin Invest. 2012;122(6):2165–75.PubMedPubMedCentral
27.
go back to reference Zhao SH, et al. Basigin-2 is the predominant basigin isoform that promotes tumor cell migration and invasion and correlates with poor prognosis in epithelial ovarian cancer. J Transl Med. 2013;11:92.PubMedPubMedCentral Zhao SH, et al. Basigin-2 is the predominant basigin isoform that promotes tumor cell migration and invasion and correlates with poor prognosis in epithelial ovarian cancer. J Transl Med. 2013;11:92.PubMedPubMedCentral
28.
go back to reference Klopfleisch R. Multiparametric and semiquantitative scoring systems for the evaluation of mouse model histopathology--a systematic review. BMC Vet Res. 2013;9:123.PubMedPubMedCentral Klopfleisch R. Multiparametric and semiquantitative scoring systems for the evaluation of mouse model histopathology--a systematic review. BMC Vet Res. 2013;9:123.PubMedPubMedCentral
29.
go back to reference Fu D, et al. Mdm2 promotes myogenesis through the ubiquitination and degradation of CCAAT/enhancer-binding protein beta. J Biol Chem. 2015;290(16):10200–7.PubMedPubMedCentral Fu D, et al. Mdm2 promotes myogenesis through the ubiquitination and degradation of CCAAT/enhancer-binding protein beta. J Biol Chem. 2015;290(16):10200–7.PubMedPubMedCentral
30.
go back to reference Xu S, et al. hSSB1 regulates both the stability and the transcriptional activity of p53. Cell Res. 2013;23(3):423–35.PubMed Xu S, et al. hSSB1 regulates both the stability and the transcriptional activity of p53. Cell Res. 2013;23(3):423–35.PubMed
31.
go back to reference Kang T, et al. GSK-3 beta targets Cdc25A for ubiquitin-mediated proteolysis, and GSK-3 beta inactivation correlates with Cdc25A overproduction in human cancers. Cancer Cell. 2008;13(1):36–47.PubMedPubMedCentral Kang T, et al. GSK-3 beta targets Cdc25A for ubiquitin-mediated proteolysis, and GSK-3 beta inactivation correlates with Cdc25A overproduction in human cancers. Cancer Cell. 2008;13(1):36–47.PubMedPubMedCentral
32.
go back to reference Xu M, et al. Reversal effect of adenovirus-mediated human interleukin 24 transfection on the cisplatin resistance of A549/DDP lung cancer cells. Oncol Rep. 2017;38(5):2843–51.PubMedPubMedCentral Xu M, et al. Reversal effect of adenovirus-mediated human interleukin 24 transfection on the cisplatin resistance of A549/DDP lung cancer cells. Oncol Rep. 2017;38(5):2843–51.PubMedPubMedCentral
33.
go back to reference Tang J, et al. Paradoxical role of CBX8 in proliferation and metastasis of colorectal cancer. Oncotarget. 2014;5(21):10778–90.PubMedPubMedCentral Tang J, et al. Paradoxical role of CBX8 in proliferation and metastasis of colorectal cancer. Oncotarget. 2014;5(21):10778–90.PubMedPubMedCentral
34.
go back to reference He Z, et al. Fyn is a novel target of (−)-epigallocatechin gallate in the inhibition of JB6 Cl41 cell transformation. Mol Carcinog. 2008;47(3):172–83.PubMedPubMedCentral He Z, et al. Fyn is a novel target of (−)-epigallocatechin gallate in the inhibition of JB6 Cl41 cell transformation. Mol Carcinog. 2008;47(3):172–83.PubMedPubMedCentral
35.
go back to reference Tang FQ, et al. HSP70 and mucin 5B: novel protein targets of N,N'-dinitrosopiperazine-induced nasopharyngeal tumorigenesis. Cancer Sci. 2009;100(2):216–24.PubMed Tang FQ, et al. HSP70 and mucin 5B: novel protein targets of N,N'-dinitrosopiperazine-induced nasopharyngeal tumorigenesis. Cancer Sci. 2009;100(2):216–24.PubMed
36.
go back to reference Tang F, et al. N,N'-dinitrosopiperazine-mediated ezrin protein phosphorylation via activation of rho kinase and protein kinase C is involved in metastasis of nasopharyngeal carcinoma 6-10B cells. J Biol Chem. 2011;286(42):36956–67.PubMedPubMedCentral Tang F, et al. N,N'-dinitrosopiperazine-mediated ezrin protein phosphorylation via activation of rho kinase and protein kinase C is involved in metastasis of nasopharyngeal carcinoma 6-10B cells. J Biol Chem. 2011;286(42):36956–67.PubMedPubMedCentral
37.
go back to reference Xu S, et al. hSSB1 binds and protects p21 from ubiquitin-mediated degradation and positively correlates with p21 in human hepatocellular carcinomas. Oncogene. 2011;30(19):2219–29.PubMed Xu S, et al. hSSB1 binds and protects p21 from ubiquitin-mediated degradation and positively correlates with p21 in human hepatocellular carcinomas. Oncogene. 2011;30(19):2219–29.PubMed
38.
go back to reference Johmura Y, et al. SCF(Fbxo22)-KDM4A targets methylated p53 for degradation and regulates senescence. Nat Commun. 2016;7:10574.PubMedPubMedCentral Johmura Y, et al. SCF(Fbxo22)-KDM4A targets methylated p53 for degradation and regulates senescence. Nat Commun. 2016;7:10574.PubMedPubMedCentral
39.
go back to reference Shamseddine AA, et al. P53-dependent upregulation of neutral sphingomyelinase-2: role in doxorubicin-induced growth arrest. Cell Death Dis. 2015;6:e1947.PubMedPubMedCentral Shamseddine AA, et al. P53-dependent upregulation of neutral sphingomyelinase-2: role in doxorubicin-induced growth arrest. Cell Death Dis. 2015;6:e1947.PubMedPubMedCentral
40.
go back to reference Hochstrasser M. Ubiquitin, proteasomes, and the regulation of intracellular protein degradation. Curr Opin Cell Biol. 1995;7(2):215–23.PubMed Hochstrasser M. Ubiquitin, proteasomes, and the regulation of intracellular protein degradation. Curr Opin Cell Biol. 1995;7(2):215–23.PubMed
41.
go back to reference Ciechanover A. The ubiquitin-proteasome proteolytic pathway. Cell. 1994;79(1):13–21.PubMed Ciechanover A. The ubiquitin-proteasome proteolytic pathway. Cell. 1994;79(1):13–21.PubMed
42.
go back to reference Orlowski RZ, Dees EC. The role of the ubiquitination-proteasome pathway in breast cancer: applying drugs that affect the ubiquitin-proteasome pathway to the therapy of breast cancer. Breast Cancer Res. 2003;5(1):1–7.PubMed Orlowski RZ, Dees EC. The role of the ubiquitination-proteasome pathway in breast cancer: applying drugs that affect the ubiquitin-proteasome pathway to the therapy of breast cancer. Breast Cancer Res. 2003;5(1):1–7.PubMed
43.
44.
go back to reference Momand J, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69(7):1237–45.PubMed Momand J, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69(7):1237–45.PubMed
45.
go back to reference Oliner JD, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362(6423):857–60.PubMed Oliner JD, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362(6423):857–60.PubMed
46.
go back to reference Tacar O, Sriamornsak P, Dass CR. Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. J Pharm Pharmacol. 2013;65(2):157–70.PubMed Tacar O, Sriamornsak P, Dass CR. Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. J Pharm Pharmacol. 2013;65(2):157–70.PubMed
47.
go back to reference Wang X, Simpson ER, Brown KA. p53: protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer Res. 2015;75(23):5001–7.PubMed Wang X, Simpson ER, Brown KA. p53: protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer Res. 2015;75(23):5001–7.PubMed
48.
go back to reference Efeyan A, Serrano M. p53: guardian of the genome and policeman of the oncogenes. Cell Cycle. 2007;6(9):1006–10.PubMed Efeyan A, Serrano M. p53: guardian of the genome and policeman of the oncogenes. Cell Cycle. 2007;6(9):1006–10.PubMed
49.
go back to reference Meulmeester E, Jochemsen AG. p53: a guide to apoptosis. Curr Cancer Drug Targets. 2008;8(2):87–97.PubMed Meulmeester E, Jochemsen AG. p53: a guide to apoptosis. Curr Cancer Drug Targets. 2008;8(2):87–97.PubMed
50.
go back to reference Bartek J, et al. Aberrant expression of the p53 oncoprotein is a common feature of a wide spectrum of human malignancies. Oncogene. 1991;6(9):1699–703.PubMed Bartek J, et al. Aberrant expression of the p53 oncoprotein is a common feature of a wide spectrum of human malignancies. Oncogene. 1991;6(9):1699–703.PubMed
51.
go back to reference Wen S, et al. p53 increase mitochondrial copy number via up-regulation of mitochondrial transcription factor A in colorectal cancer. Oncotarget. 2016;7(10):12514. Wen S, et al. p53 increase mitochondrial copy number via up-regulation of mitochondrial transcription factor A in colorectal cancer. Oncotarget. 2016;7(10):12514.
52.
go back to reference Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997;88(3):323–31.PubMed Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997;88(3):323–31.PubMed
53.
go back to reference Sarasqueta AF, et al. Integral analysis of p53 and its value as prognostic factor in sporadic colon cancer. BMC Cancer. 2013;13(277):1471–2407. Sarasqueta AF, et al. Integral analysis of p53 and its value as prognostic factor in sporadic colon cancer. BMC Cancer. 2013;13(277):1471–2407.
54.
go back to reference Liu BW, et al. Prognostic effect of p53 expression in patients with completely resected colorectal cancer. Tumour Biol. 2014;35(10):9893–6.PubMed Liu BW, et al. Prognostic effect of p53 expression in patients with completely resected colorectal cancer. Tumour Biol. 2014;35(10):9893–6.PubMed
55.
go back to reference Stefancikova L, et al. Prognostic impact of p53 aberrations for R-CHOP-treated patients with diffuse large B-cell lymphoma. Int J Oncol. 2011;39(6):1413–20.PubMed Stefancikova L, et al. Prognostic impact of p53 aberrations for R-CHOP-treated patients with diffuse large B-cell lymphoma. Int J Oncol. 2011;39(6):1413–20.PubMed
56.
go back to reference Harvey M, et al. Genetic background alters the spectrum of tumors that develop in p53-deficient mice. FASEB J. 1993;7(10):938–43.PubMed Harvey M, et al. Genetic background alters the spectrum of tumors that develop in p53-deficient mice. FASEB J. 1993;7(10):938–43.PubMed
57.
go back to reference Jacks T, et al. Tumor spectrum analysis in p53-mutant mice. Curr Biol. 1994;4(1):1–7.PubMed Jacks T, et al. Tumor spectrum analysis in p53-mutant mice. Curr Biol. 1994;4(1):1–7.PubMed
58.
go back to reference Strong LC. General keynote: hereditary cancer: lessons from li-Fraumeni syndrome. Gynecol Oncol. 2003;88(1 Pt 2):S11–3. Strong LC. General keynote: hereditary cancer: lessons from li-Fraumeni syndrome. Gynecol Oncol. 2003;88(1 Pt 2):S11–3.
59.
60.
go back to reference Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001;7(3):683–94.PubMed Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001;7(3):683–94.PubMed
61.
go back to reference Bouvard V, et al. Tissue and cell-specific expression of the p53-target genes: bax, fas, mdm2 and waf1/p21, before and following ionising irradiation in mice. Oncogene. 2000;19(5):649–60.PubMed Bouvard V, et al. Tissue and cell-specific expression of the p53-target genes: bax, fas, mdm2 and waf1/p21, before and following ionising irradiation in mice. Oncogene. 2000;19(5):649–60.PubMed
62.
go back to reference Muller M, et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J Exp Med. 1998;188(11):2033–45.PubMedPubMedCentral Muller M, et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J Exp Med. 1998;188(11):2033–45.PubMedPubMedCentral
63.
go back to reference Aubrey BJ, et al. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–13.PubMed Aubrey BJ, et al. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–13.PubMed
64.
go back to reference Hock AK, Vousden KH. The role of ubiquitin modification in the regulation of p53. Biochim Biophys Acta. 2014;1:137–49. Hock AK, Vousden KH. The role of ubiquitin modification in the regulation of p53. Biochim Biophys Acta. 2014;1:137–49.
65.
go back to reference Pant V, Lozano G. Limiting the power of p53 through the ubiquitin proteasome pathway. Genes Dev. 2014;28(16):1739–51.PubMedPubMedCentral Pant V, Lozano G. Limiting the power of p53 through the ubiquitin proteasome pathway. Genes Dev. 2014;28(16):1739–51.PubMedPubMedCentral
Metadata
Title
RETRACTED ARTICLE: MNAT1 is overexpressed in colorectal cancer and mediates p53 ubiquitin-degradation to promote colorectal cancer malignance
Authors
Shan Zhou
Jinping Lu
Yuejin Li
Chan Chen
Yongqiang Cai
Gongjun Tan
Zhengke Peng
Zhenlin Zhang
Zigang Dong
Tiebang Kang
Faqing Tang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0956-3

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine