Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2016

Open Access 01-12-2016 | Review

Signalling pathways in UHRF1-dependent regulation of tumor suppressor genes in cancer

Authors: Mahmoud Alhosin, Ziad Omran, Mazin A. Zamzami, Abdulrahman L. Al-Malki, Hani Choudhry, Marc Mousli, Christian Bronner

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2016

Login to get access

Abstract

Epigenetic silencing of tumor suppressor genes (TSGs) through DNA methylation and histone changes is a main hallmark of cancer. Ubiquitin-like with PHD and RING Finger domains 1 (UHRF1) is a potent oncogene overexpressed in various solid and haematological tumors and its high expression levels are associated with decreased expression of several TSGs including p16 INK4A , BRCA1, PPARG and KiSS1. Using its several functional domains, UHRF1 creates a strong coordinated dialogue between DNA methylation and histone post-translation modification changes causing the epigenetic silencing of TSGs which allows cancer cells to escape apoptosis. To ensure the silencing of TSGs during cell division, UHRF1 recruits several enzymes including histone deacetylase 1 (HDAC1), DNA methyltransferase 1 (DNMT1) and histone lysine methyltransferases G9a and Suv39H1 to the right place at the right moment. Several in vitro and in vivo works have reported the direct implication of the epigenetic player UHRF1 in tumorigenesis through the repression of TSGs expression and suggested UHRF1 as a promising target for cancer treatment. This review describes the molecular mechanisms underlying UHRF1 regulation in cancer and discusses its importance as a therapeutic target to induce the reactivation of TSGs and subsequent apoptosis.
Literature
1.
go back to reference Venza M, Visalli M, Biondo C, Oteri R, Agliano F, Morabito S, Teti D, Venza I. Epigenetic marks responsible for cadmium-induced melanoma cell overgrowth. Toxicol In Vitro. 2015;29(1):242–50.PubMedCrossRef Venza M, Visalli M, Biondo C, Oteri R, Agliano F, Morabito S, Teti D, Venza I. Epigenetic marks responsible for cadmium-induced melanoma cell overgrowth. Toxicol In Vitro. 2015;29(1):242–50.PubMedCrossRef
4.
5.
go back to reference Bronner C, Achour M, Arima Y, Chataigneau T, Saya H, Schini-Kerth VB. The UHRF family: oncogenes that are drugable targets for cancer therapy in the near future? Pharmacol Ther. 2007;115(3):419–34.PubMedCrossRef Bronner C, Achour M, Arima Y, Chataigneau T, Saya H, Schini-Kerth VB. The UHRF family: oncogenes that are drugable targets for cancer therapy in the near future? Pharmacol Ther. 2007;115(3):419–34.PubMedCrossRef
6.
go back to reference Bronner C, Krifa M, Mousli M. Increasing role of UHRF1 in the reading and inheritance of the epigenetic code as well as in tumorogenesis. Biochem Pharmacol. 2013;86(12):1643–9.PubMedCrossRef Bronner C, Krifa M, Mousli M. Increasing role of UHRF1 in the reading and inheritance of the epigenetic code as well as in tumorogenesis. Biochem Pharmacol. 2013;86(12):1643–9.PubMedCrossRef
7.
go back to reference Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23(46):7601–10.PubMedCrossRef Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23(46):7601–10.PubMedCrossRef
8.
go back to reference Unoki M. Current and potential anticancer drugs targeting members of the UHRF1 complex including epigenetic modifiers. Recent Pat Anticancer Drug Discov. 2011;6(1):116–30.PubMedCrossRef Unoki M. Current and potential anticancer drugs targeting members of the UHRF1 complex including epigenetic modifiers. Recent Pat Anticancer Drug Discov. 2011;6(1):116–30.PubMedCrossRef
9.
go back to reference Hashimoto H, Horton JR, Zhang X, Bostick M, Jacobsen SE, Cheng X. The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix. Nature. 2008;455(7214):826–9.PubMedPubMedCentralCrossRef Hashimoto H, Horton JR, Zhang X, Bostick M, Jacobsen SE, Cheng X. The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix. Nature. 2008;455(7214):826–9.PubMedPubMedCentralCrossRef
10.
go back to reference Avvakumov GV, Walker JR, Xue S, Li Y, Duan S, Bronner C, Arrowsmith CH, Dhe-Paganon S. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature. 2008;455(7214):822–5.PubMedCrossRef Avvakumov GV, Walker JR, Xue S, Li Y, Duan S, Bronner C, Arrowsmith CH, Dhe-Paganon S. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature. 2008;455(7214):822–5.PubMedCrossRef
11.
go back to reference Arita K, Ariyoshi M, Tochio H, Nakamura Y, Shirakawa M. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature. 2008;455(7214):818–21.PubMedCrossRef Arita K, Ariyoshi M, Tochio H, Nakamura Y, Shirakawa M. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature. 2008;455(7214):818–21.PubMedCrossRef
12.
go back to reference Harrison JS, Cornett EM, Goldfarb D, DaRosa PA, Li ZM, Yan F, Dickson BM, Guo AH, Cantu DV, Kaustov L, et al. Hemi-methylated DNA regulates DNA methylation inheritance through allosteric activation of H3 ubiquitylation by UHRF1. Elife. 2016;5:e17101.PubMedPubMedCentralCrossRef Harrison JS, Cornett EM, Goldfarb D, DaRosa PA, Li ZM, Yan F, Dickson BM, Guo AH, Cantu DV, Kaustov L, et al. Hemi-methylated DNA regulates DNA methylation inheritance through allosteric activation of H3 ubiquitylation by UHRF1. Elife. 2016;5:e17101.PubMedPubMedCentralCrossRef
13.
go back to reference Nishiyama A, Yamaguchi L, Sharif J, Johmura Y, Kawamura T, Nakanishi K, Shimamura S, Arita K, Kodama T, Ishikawa F, et al. Uhrf1-dependent H3K23 ubiquitylation couples maintenance DNA methylation and replication. Nature. 2013;502(7470):249–53.PubMedCrossRef Nishiyama A, Yamaguchi L, Sharif J, Johmura Y, Kawamura T, Nakanishi K, Shimamura S, Arita K, Kodama T, Ishikawa F, et al. Uhrf1-dependent H3K23 ubiquitylation couples maintenance DNA methylation and replication. Nature. 2013;502(7470):249–53.PubMedCrossRef
14.
go back to reference Rothbart SB, Dickson BM, Ong MS, Krajewski K, Houliston S, Kireev DB, Arrowsmith CH, Strahl BD. Multivalent histone engagement by the linked tandem Tudor and PHD domains of UHRF1 is required for the epigenetic inheritance of DNA methylation. Genes Dev. 2013;27(11):1288–98.PubMedPubMedCentralCrossRef Rothbart SB, Dickson BM, Ong MS, Krajewski K, Houliston S, Kireev DB, Arrowsmith CH, Strahl BD. Multivalent histone engagement by the linked tandem Tudor and PHD domains of UHRF1 is required for the epigenetic inheritance of DNA methylation. Genes Dev. 2013;27(11):1288–98.PubMedPubMedCentralCrossRef
15.
go back to reference Tauber M, Fischle W. Conserved linker regions and their regulation determine multiple chromatin-binding modes of UHRF1. Nucleus (Austin, Tex). 2015;6(2):123–32. Tauber M, Fischle W. Conserved linker regions and their regulation determine multiple chromatin-binding modes of UHRF1. Nucleus (Austin, Tex). 2015;6(2):123–32.
16.
go back to reference Fang J, Cheng J, Wang J, Zhang Q, Liu M, Gong R, Wang P, Zhang X, Feng Y, Lan W, et al. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its histone recognition. Nat Commun. 2016;7:11197.PubMedPubMedCentralCrossRef Fang J, Cheng J, Wang J, Zhang Q, Liu M, Gong R, Wang P, Zhang X, Feng Y, Lan W, et al. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its histone recognition. Nat Commun. 2016;7:11197.PubMedPubMedCentralCrossRef
17.
go back to reference Gelato KA, Tauber M, Ong MS, Winter S, Hiragami-Hamada K, Sindlinger J, Lemak A, Bultsma Y, Houliston S, Schwarzer D, et al. Accessibility of different histone H3-binding domains of UHRF1 is allosterically regulated by phosphatidylinositol 5-phosphate. Mol Cell. 2014;54(6):905–19.PubMedCrossRef Gelato KA, Tauber M, Ong MS, Winter S, Hiragami-Hamada K, Sindlinger J, Lemak A, Bultsma Y, Houliston S, Schwarzer D, et al. Accessibility of different histone H3-binding domains of UHRF1 is allosterically regulated by phosphatidylinositol 5-phosphate. Mol Cell. 2014;54(6):905–19.PubMedCrossRef
19.
go back to reference Zhao Q, Zhang J, Chen R, Wang L, Li B, Cheng H, Duan X, Zhu H, Wei W, Li J, et al. Dissecting the precise role of H3K9 methylation in crosstalk with DNA maintenance methylation in mammals. Nat Commun. 2016;7:12464.PubMedCrossRef Zhao Q, Zhang J, Chen R, Wang L, Li B, Cheng H, Duan X, Zhu H, Wei W, Li J, et al. Dissecting the precise role of H3K9 methylation in crosstalk with DNA maintenance methylation in mammals. Nat Commun. 2016;7:12464.PubMedCrossRef
20.
go back to reference Liang CC, Zhan B, Yoshikawa Y, Haas W, Gygi SP, Cohn MA. UHRF1 is a sensor for DNA interstrand crosslinks and recruits FANCD2 to initiate the Fanconi anemia pathway. Cell Rep. 2015;10(12):1947–56.PubMedPubMedCentralCrossRef Liang CC, Zhan B, Yoshikawa Y, Haas W, Gygi SP, Cohn MA. UHRF1 is a sensor for DNA interstrand crosslinks and recruits FANCD2 to initiate the Fanconi anemia pathway. Cell Rep. 2015;10(12):1947–56.PubMedPubMedCentralCrossRef
21.
go back to reference Liang CC, Cohn MA. UHRF1 is a sensor for DNA interstrand crosslinks. Oncotarget. 2016;7(1):3–4.PubMed Liang CC, Cohn MA. UHRF1 is a sensor for DNA interstrand crosslinks. Oncotarget. 2016;7(1):3–4.PubMed
23.
go back to reference von Meyenn F, Iurlaro M, Habibi E, Liu NQ, Salehzadeh-Yazdi A, Santos F, Petrini E, Milagre I, Yu M, Xie Z, et al. Impairment of DNA methylation maintenance is the main cause of global demethylation in naive embryonic stem cells. Mol Cell. 2016;62(6):848–61.CrossRef von Meyenn F, Iurlaro M, Habibi E, Liu NQ, Salehzadeh-Yazdi A, Santos F, Petrini E, Milagre I, Yu M, Xie Z, et al. Impairment of DNA methylation maintenance is the main cause of global demethylation in naive embryonic stem cells. Mol Cell. 2016;62(6):848–61.CrossRef
24.
go back to reference Myrianthopoulos V, Cartron PF, Liutkeviciute Z, Klimasauskas S, Matulis D, Bronner C, Martinet N, Mikros E. Tandem virtual screening targeting the SRA domain of UHRF1 identifies a novel chemical tool modulating DNA methylation. Eur J Med Chem. 2016;114:390–6.PubMedCrossRef Myrianthopoulos V, Cartron PF, Liutkeviciute Z, Klimasauskas S, Matulis D, Bronner C, Martinet N, Mikros E. Tandem virtual screening targeting the SRA domain of UHRF1 identifies a novel chemical tool modulating DNA methylation. Eur J Med Chem. 2016;114:390–6.PubMedCrossRef
25.
go back to reference Hopfner R, Mousli M, Garnier JM, Redon R, du Manoir S, Chatton B, Ghyselinck N, Oudet P, Bronner C. Genomic structure and chromosomal mapping of the gene coding for ICBP90, a protein involved in the regulation of the topoisomerase IIalpha gene expression. Gene. 2001;266(1–2):15–23.PubMedCrossRef Hopfner R, Mousli M, Garnier JM, Redon R, du Manoir S, Chatton B, Ghyselinck N, Oudet P, Bronner C. Genomic structure and chromosomal mapping of the gene coding for ICBP90, a protein involved in the regulation of the topoisomerase IIalpha gene expression. Gene. 2001;266(1–2):15–23.PubMedCrossRef
26.
go back to reference Qu X, Davison J, Du L, Storer B, Stirewalt DL, Heimfeld S, Estey E, Appelbaum FR, Fang M. Identification of differentially methylated markers among cytogenetic risk groups of acute myeloid leukemia. Epigenetics. 2015;10(6):526–35.PubMedPubMedCentralCrossRef Qu X, Davison J, Du L, Storer B, Stirewalt DL, Heimfeld S, Estey E, Appelbaum FR, Fang M. Identification of differentially methylated markers among cytogenetic risk groups of acute myeloid leukemia. Epigenetics. 2015;10(6):526–35.PubMedPubMedCentralCrossRef
27.
go back to reference Unoki M, Brunet J, Mousli M. Drug discovery targeting epigenetic codes: the great potential of UHRF1, which links DNA methylation and histone modifications, as a drug target in cancers and toxoplasmosis. Biochem Pharmacol. 2009;78(10):1279–88.PubMedCrossRef Unoki M, Brunet J, Mousli M. Drug discovery targeting epigenetic codes: the great potential of UHRF1, which links DNA methylation and histone modifications, as a drug target in cancers and toxoplasmosis. Biochem Pharmacol. 2009;78(10):1279–88.PubMedCrossRef
29.
go back to reference Alhosin M, Sharif T, Mousli M, Etienne-Selloum N, Fuhrmann G, Schini-Kerth VB, Bronner C. Down-regulation of UHRF1, associated with re-expression of tumor suppressor genes, is a common feature of natural compounds exhibiting anti-cancer properties. J Exp Clin Cancer Res. 2011;30:41.PubMedPubMedCentralCrossRef Alhosin M, Sharif T, Mousli M, Etienne-Selloum N, Fuhrmann G, Schini-Kerth VB, Bronner C. Down-regulation of UHRF1, associated with re-expression of tumor suppressor genes, is a common feature of natural compounds exhibiting anti-cancer properties. J Exp Clin Cancer Res. 2011;30:41.PubMedPubMedCentralCrossRef
30.
go back to reference Bostick M, Kim JK, Esteve PO, Clark A, Pradhan S, Jacobsen SE. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science. 2007;317(5845):1760–4.PubMedCrossRef Bostick M, Kim JK, Esteve PO, Clark A, Pradhan S, Jacobsen SE. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science. 2007;317(5845):1760–4.PubMedCrossRef
31.
go back to reference Achour M, Jacq X, Ronde P, Alhosin M, Charlot C, Chataigneau T, Jeanblanc M, Macaluso M, Giordano A, Hughes AD, et al. The interaction of the SRA domain of ICBP90 with a novel domain of DNMT1 is involved in the regulation of VEGF gene expression. Oncogene. 2008;27(15):2187–97.PubMedCrossRef Achour M, Jacq X, Ronde P, Alhosin M, Charlot C, Chataigneau T, Jeanblanc M, Macaluso M, Giordano A, Hughes AD, et al. The interaction of the SRA domain of ICBP90 with a novel domain of DNMT1 is involved in the regulation of VEGF gene expression. Oncogene. 2008;27(15):2187–97.PubMedCrossRef
32.
go back to reference Achour M, Mousli M, Alhosin M, Ibrahim A, Peluso J, Muller CD, Schini-Kerth VB, Hamiche A, Dhe-Paganon S, Bronner C. Epigallocatechin-3-gallate up-regulates tumor suppressor gene expression via a reactive oxygen species-dependent down-regulation of UHRF1. Biochem Biophys Res Commun. 2013;430(1):208–12.PubMedCrossRef Achour M, Mousli M, Alhosin M, Ibrahim A, Peluso J, Muller CD, Schini-Kerth VB, Hamiche A, Dhe-Paganon S, Bronner C. Epigallocatechin-3-gallate up-regulates tumor suppressor gene expression via a reactive oxygen species-dependent down-regulation of UHRF1. Biochem Biophys Res Commun. 2013;430(1):208–12.PubMedCrossRef
33.
go back to reference Bashtrykov P, Jankevicius G, Jurkowska RZ, Ragozin S, Jeltsch A. The UHRF1 protein stimulates the activity and specificity of the maintenance DNA methyltransferase DNMT1 by an allosteric mechanism. J Biol Chem. 2014;289(7):4106–15.PubMedCrossRef Bashtrykov P, Jankevicius G, Jurkowska RZ, Ragozin S, Jeltsch A. The UHRF1 protein stimulates the activity and specificity of the maintenance DNA methyltransferase DNMT1 by an allosteric mechanism. J Biol Chem. 2014;289(7):4106–15.PubMedCrossRef
34.
go back to reference Al-Kaabi A, van Bockel LW, Pothen AJ, Willems SM. p16INK4A and p14ARF gene promoter hypermethylation as prognostic biomarker in oral and oropharyngeal squamous cell carcinoma: a review. Dis Markers. 2014;2014:260549.PubMedPubMedCentralCrossRef Al-Kaabi A, van Bockel LW, Pothen AJ, Willems SM. p16INK4A and p14ARF gene promoter hypermethylation as prognostic biomarker in oral and oropharyngeal squamous cell carcinoma: a review. Dis Markers. 2014;2014:260549.PubMedPubMedCentralCrossRef
35.
go back to reference Wang F, Yang YZ, Shi CZ, Zhang P, Moyer MP, Zhang HZ, Zou Y, Qin HL. UHRF1 promotes cell growth and metastasis through repression of p16(ink(4)a) in colorectal cancer. Ann Surg Oncol. 2012;19(8):2753–62.PubMedCrossRef Wang F, Yang YZ, Shi CZ, Zhang P, Moyer MP, Zhang HZ, Zou Y, Qin HL. UHRF1 promotes cell growth and metastasis through repression of p16(ink(4)a) in colorectal cancer. Ann Surg Oncol. 2012;19(8):2753–62.PubMedCrossRef
36.
go back to reference Daskalos A, Oleksiewicz U, Filia A, Nikolaidis G, Xinarianos G, Gosney JR, Malliri A, Field JK, Liloglou T. UHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancer. Cancer. 2011;117(5):1027–37.PubMedCrossRef Daskalos A, Oleksiewicz U, Filia A, Nikolaidis G, Xinarianos G, Gosney JR, Malliri A, Field JK, Liloglou T. UHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancer. Cancer. 2011;117(5):1027–37.PubMedCrossRef
37.
go back to reference Zhou L, Shang Y, Jin Z, Zhang W, Lv C, Zhao X, Liu Y, Li N, Liang J. UHRF1 promotes proliferation of gastric cancer via mediating tumor suppressor gene hypermethylation. Cancer Biol Ther. 2015;16(8):1241–51.PubMedPubMedCentralCrossRef Zhou L, Shang Y, Jin Z, Zhang W, Lv C, Zhao X, Liu Y, Li N, Liang J. UHRF1 promotes proliferation of gastric cancer via mediating tumor suppressor gene hypermethylation. Cancer Biol Ther. 2015;16(8):1241–51.PubMedPubMedCentralCrossRef
38.
go back to reference Boukhari A, Alhosin M, Bronner C, Sagini K, Truchot C, Sick E, Schini-Kerth VB, Andre P, Mely Y, Mousli M, et al. CD47 activation-induced UHRF1 over-expression is associated with silencing of tumor suppressor gene p16INK4A in glioblastoma cells. Anticancer Res. 2015;35(1):149–57.PubMed Boukhari A, Alhosin M, Bronner C, Sagini K, Truchot C, Sick E, Schini-Kerth VB, Andre P, Mely Y, Mousli M, et al. CD47 activation-induced UHRF1 over-expression is associated with silencing of tumor suppressor gene p16INK4A in glioblastoma cells. Anticancer Res. 2015;35(1):149–57.PubMed
39.
go back to reference Jeanblanc M, Mousli M, Hopfner R, Bathami K, Martinet N, Abbady AQ, Siffert JC, Mathieu E, Muller CD, Bronner C. The retinoblastoma gene and its product are targeted by ICBP90: a key mechanism in the G1/S transition during the cell cycle. Oncogene. 2005;24(49):7337–45.PubMedCrossRef Jeanblanc M, Mousli M, Hopfner R, Bathami K, Martinet N, Abbady AQ, Siffert JC, Mathieu E, Muller CD, Bronner C. The retinoblastoma gene and its product are targeted by ICBP90: a key mechanism in the G1/S transition during the cell cycle. Oncogene. 2005;24(49):7337–45.PubMedCrossRef
40.
go back to reference Liu W, Qiao RH, Wang DM, Huang XW, Li B, Wang D. UHRF1 promotes human osteosarcoma cell invasion by downregulating the expression of Ecadherin in an Rb1dependent manner. Mol Med Rep. 2016;13(1):315–20.PubMed Liu W, Qiao RH, Wang DM, Huang XW, Li B, Wang D. UHRF1 promotes human osteosarcoma cell invasion by downregulating the expression of Ecadherin in an Rb1dependent manner. Mol Med Rep. 2016;13(1):315–20.PubMed
41.
go back to reference Chen H, Zhang C, Sheng Y, Yao S, Liu Z, Zhang C, Zhang T. Frequent SOCS3 and 3OST2 promoter methylation and their epigenetic regulation in endometrial carcinoma. Am J Cancer Res. 2015;5(1):180–90.PubMed Chen H, Zhang C, Sheng Y, Yao S, Liu Z, Zhang C, Zhang T. Frequent SOCS3 and 3OST2 promoter methylation and their epigenetic regulation in endometrial carcinoma. Am J Cancer Res. 2015;5(1):180–90.PubMed
42.
go back to reference Jin W, Chen L, Chen Y, Xu SG, Di GH, Yin WJ, Wu J, Shao ZM. UHRF1 is associated with epigenetic silencing of BRCA1 in sporadic breast cancer. Breast Cancer Res Treat. 2010;123(2):359–73.PubMedCrossRef Jin W, Chen L, Chen Y, Xu SG, Di GH, Yin WJ, Wu J, Shao ZM. UHRF1 is associated with epigenetic silencing of BRCA1 in sporadic breast cancer. Breast Cancer Res Treat. 2010;123(2):359–73.PubMedCrossRef
43.
go back to reference Zhang Y, Huang Z, Zhu Z, Zheng X, Liu J, Han Z, Ma X, Zhang Y. Upregulated UHRF1 promotes bladder cancer cell invasion by epigenetic silencing of KiSS1. PLoS One. 2014;9(10):e104252.PubMedPubMedCentralCrossRef Zhang Y, Huang Z, Zhu Z, Zheng X, Liu J, Han Z, Ma X, Zhang Y. Upregulated UHRF1 promotes bladder cancer cell invasion by epigenetic silencing of KiSS1. PLoS One. 2014;9(10):e104252.PubMedPubMedCentralCrossRef
44.
go back to reference Sheng Y, Wang H, Liu D, Zhang C, Deng Y, Yang F, Zhang T, Zhang C. Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma. Gynecol Oncol. 2016;140(1):145–51.PubMedCrossRef Sheng Y, Wang H, Liu D, Zhang C, Deng Y, Yang F, Zhang T, Zhang C. Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma. Gynecol Oncol. 2016;140(1):145–51.PubMedCrossRef
45.
go back to reference Tae S, Karkhanis V, Velasco K, Yaneva M, Erdjument-Bromage H, Tempst P, Sif S. Bromodomain protein 7 interacts with PRMT5 and PRC2, and is involved in transcriptional repression of their target genes. Nucleic Acids Res. 2011;39(13):5424–38.PubMedPubMedCentralCrossRef Tae S, Karkhanis V, Velasco K, Yaneva M, Erdjument-Bromage H, Tempst P, Sif S. Bromodomain protein 7 interacts with PRMT5 and PRC2, and is involved in transcriptional repression of their target genes. Nucleic Acids Res. 2011;39(13):5424–38.PubMedPubMedCentralCrossRef
46.
go back to reference Li M, Chen D, Shiloh A, Luo J, Nikolaev AY, Qin J, Gu W. Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature. 2002;416(6881):648–53.PubMedCrossRef Li M, Chen D, Shiloh A, Luo J, Nikolaev AY, Qin J, Gu W. Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature. 2002;416(6881):648–53.PubMedCrossRef
47.
go back to reference Qin W, Leonhardt H, Spada F. Usp7 and Uhrf1 control ubiquitination and stability of the maintenance DNA methyltransferase Dnmt1. J Cell Biochem. 2011;112(2):439–44.PubMedCrossRef Qin W, Leonhardt H, Spada F. Usp7 and Uhrf1 control ubiquitination and stability of the maintenance DNA methyltransferase Dnmt1. J Cell Biochem. 2011;112(2):439–44.PubMedCrossRef
48.
go back to reference Felle M, Joppien S, Nemeth A, Diermeier S, Thalhammer V, Dobner T, Kremmer E, Kappler R, Langst G. The USP7/Dnmt1 complex stimulates the DNA methylation activity of Dnmt1 and regulates the stability of UHRF1. Nucleic Acids Res. 2011;39(19):8355–65.PubMedPubMedCentralCrossRef Felle M, Joppien S, Nemeth A, Diermeier S, Thalhammer V, Dobner T, Kremmer E, Kappler R, Langst G. The USP7/Dnmt1 complex stimulates the DNA methylation activity of Dnmt1 and regulates the stability of UHRF1. Nucleic Acids Res. 2011;39(19):8355–65.PubMedPubMedCentralCrossRef
49.
go back to reference Ma H, Chen H, Guo X, Wang Z, Sowa ME, Zheng L, Hu S, Zeng P, Guo R, Diao J, et al. M phase phosphorylation of the epigenetic regulator UHRF1 regulates its physical association with the deubiquitylase USP7 and stability. Proc Natl Acad Sci U S A. 2012;109(13):4828–33.PubMedPubMedCentralCrossRef Ma H, Chen H, Guo X, Wang Z, Sowa ME, Zheng L, Hu S, Zeng P, Guo R, Diao J, et al. M phase phosphorylation of the epigenetic regulator UHRF1 regulates its physical association with the deubiquitylase USP7 and stability. Proc Natl Acad Sci U S A. 2012;109(13):4828–33.PubMedPubMedCentralCrossRef
50.
go back to reference Zhang ZM, Rothbart SB, Allison DF, Cai Q, Harrison JS, Li L, Wang Y, Strahl BD, Wang GG, Song J. An allosteric interaction links USP7 to deubiquitination and chromatin targeting of UHRF1. Cell Rep. 2015;12(9):1400–6.PubMedPubMedCentralCrossRef Zhang ZM, Rothbart SB, Allison DF, Cai Q, Harrison JS, Li L, Wang Y, Strahl BD, Wang GG, Song J. An allosteric interaction links USP7 to deubiquitination and chromatin targeting of UHRF1. Cell Rep. 2015;12(9):1400–6.PubMedPubMedCentralCrossRef
51.
go back to reference Ramakrishna S, Suresh B, Baek KH. The role of deubiquitinating enzymes in apoptosis. Cell Mol Life Sci. 2011;68(1):15–26.PubMedCrossRef Ramakrishna S, Suresh B, Baek KH. The role of deubiquitinating enzymes in apoptosis. Cell Mol Life Sci. 2011;68(1):15–26.PubMedCrossRef
52.
go back to reference Dai C, Shi D, Gu W. Negative regulation of the acetyltransferase TIP60-p53 interplay by UHRF1 (ubiquitin-like with PHD and RING finger domains 1). J Biol Chem. 2013;288(27):19581–92.PubMedPubMedCentralCrossRef Dai C, Shi D, Gu W. Negative regulation of the acetyltransferase TIP60-p53 interplay by UHRF1 (ubiquitin-like with PHD and RING finger domains 1). J Biol Chem. 2013;288(27):19581–92.PubMedPubMedCentralCrossRef
53.
go back to reference Achour M, Fuhrmann G, Alhosin M, Ronde P, Chataigneau T, Mousli M, Schini-Kerth VB, Bronner C. UHRF1 recruits the histone acetyltransferase Tip60 and controls its expression and activity. Biochem Biophys Res Commun. 2009;390(3):523–8.PubMedCrossRef Achour M, Fuhrmann G, Alhosin M, Ronde P, Chataigneau T, Mousli M, Schini-Kerth VB, Bronner C. UHRF1 recruits the histone acetyltransferase Tip60 and controls its expression and activity. Biochem Biophys Res Commun. 2009;390(3):523–8.PubMedCrossRef
54.
go back to reference Tang Y, Luo J, Zhang W, Gu W. Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell. 2006;24(6):827–39.PubMedCrossRef Tang Y, Luo J, Zhang W, Gu W. Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell. 2006;24(6):827–39.PubMedCrossRef
55.
go back to reference Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent A, Minty A, Chalon P, Lelias JM, Dumont X, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90(4):809–19.PubMedCrossRef Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent A, Minty A, Chalon P, Lelias JM, Dumont X, et al. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90(4):809–19.PubMedCrossRef
57.
go back to reference Ikawa S, Obinata M, Ikawa Y. Human p53-p51 (p53-related) fusion protein: a potent BAX transactivator. Jpn J Cancer Res. 1999;90(6):596–9.PubMedCrossRef Ikawa S, Obinata M, Ikawa Y. Human p53-p51 (p53-related) fusion protein: a potent BAX transactivator. Jpn J Cancer Res. 1999;90(6):596–9.PubMedCrossRef
58.
go back to reference Basu S, Murphy ME. Genetic modifiers of the p53 pathway. Cold Spring Harb Perspect Med. 2016;6(4):a026302.PubMedCrossRef Basu S, Murphy ME. Genetic modifiers of the p53 pathway. Cold Spring Harb Perspect Med. 2016;6(4):a026302.PubMedCrossRef
59.
go back to reference Haupt S, Raghu D, Haupt Y. Mutant p53 drives cancer by subverting multiple tumor suppression pathways. Front Oncol. 2016;6:12.PubMedPubMedCentral Haupt S, Raghu D, Haupt Y. Mutant p53 drives cancer by subverting multiple tumor suppression pathways. Front Oncol. 2016;6:12.PubMedPubMedCentral
60.
go back to reference Murray-Zmijewski F, Lane DP, Bourdon JC. p53/p63/p73 isoforms: an orchestra of isoforms to harmonise cell differentiation and response to stress. Cell Death Differ. 2006;13(6):962–72.PubMedCrossRef Murray-Zmijewski F, Lane DP, Bourdon JC. p53/p63/p73 isoforms: an orchestra of isoforms to harmonise cell differentiation and response to stress. Cell Death Differ. 2006;13(6):962–72.PubMedCrossRef
62.
go back to reference Arima Y, Hirota T, Bronner C, Mousli M, Fujiwara T, Niwa S, Ishikawa H, Saya H. Down-regulation of nuclear protein ICBP90 by p53/p21Cip1/WAF1-dependent DNA-damage checkpoint signals contributes to cell cycle arrest at G1/S transition. Genes Cells. 2004;9(2):131–42.PubMedCrossRef Arima Y, Hirota T, Bronner C, Mousli M, Fujiwara T, Niwa S, Ishikawa H, Saya H. Down-regulation of nuclear protein ICBP90 by p53/p21Cip1/WAF1-dependent DNA-damage checkpoint signals contributes to cell cycle arrest at G1/S transition. Genes Cells. 2004;9(2):131–42.PubMedCrossRef
63.
go back to reference Alhosin M, Abusnina A, Achour M, Sharif T, Muller C, Peluso J, Chataigneau T, Lugnier C, Schini-Kerth VB, Bronner C, et al. Induction of apoptosis by thymoquinone in lymphoblastic leukemia Jurkat cells is mediated by a p73-dependent pathway which targets the epigenetic integrator UHRF1. Biochem Pharmacol. 2010;79(9):1251–60.PubMedCrossRef Alhosin M, Abusnina A, Achour M, Sharif T, Muller C, Peluso J, Chataigneau T, Lugnier C, Schini-Kerth VB, Bronner C, et al. Induction of apoptosis by thymoquinone in lymphoblastic leukemia Jurkat cells is mediated by a p73-dependent pathway which targets the epigenetic integrator UHRF1. Biochem Pharmacol. 2010;79(9):1251–60.PubMedCrossRef
64.
go back to reference Sharif T, Auger C, Alhosin M, Ebel C, Achour M, Etienne-Selloum N, Fuhrmann G, Bronner C, Schini-Kerth VB. Red wine polyphenols cause growth inhibition and apoptosis in acute lymphoblastic leukaemia cells by inducing a redox-sensitive up-regulation of p73 and down-regulation of UHRF1. Eur J Cancer (Oxford, England : 1990). 2010;46(5):983–94.CrossRef Sharif T, Auger C, Alhosin M, Ebel C, Achour M, Etienne-Selloum N, Fuhrmann G, Bronner C, Schini-Kerth VB. Red wine polyphenols cause growth inhibition and apoptosis in acute lymphoblastic leukaemia cells by inducing a redox-sensitive up-regulation of p73 and down-regulation of UHRF1. Eur J Cancer (Oxford, England : 1990). 2010;46(5):983–94.CrossRef
66.
go back to reference Liu X, Chen X, Yu X, Tao Y, Bode AM, Dong Z, Cao Y. Regulation of microRNAs by epigenetics and their interplay involved in cancer. J Exp Clin Cancer Res. 2013;32:96.PubMedPubMedCentralCrossRef Liu X, Chen X, Yu X, Tao Y, Bode AM, Dong Z, Cao Y. Regulation of microRNAs by epigenetics and their interplay involved in cancer. J Exp Clin Cancer Res. 2013;32:96.PubMedPubMedCentralCrossRef
69.
go back to reference Rokavec M, Li H, Jiang L, Hermeking H. The p53/microRNA connection in gastrointestinal cancer. Clin Exp Gastroenterol. 2014;7:395–413.PubMedPubMedCentral Rokavec M, Li H, Jiang L, Hermeking H. The p53/microRNA connection in gastrointestinal cancer. Clin Exp Gastroenterol. 2014;7:395–413.PubMedPubMedCentral
70.
go back to reference Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6(4):259–69.PubMedCrossRef Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6(4):259–69.PubMedCrossRef
71.
go back to reference Osada H, Takahashi T. let-7 and miR-17-92: small-sized major players in lung cancer development. Cancer Sci. 2011;102(1):9–17.PubMedCrossRef Osada H, Takahashi T. let-7 and miR-17-92: small-sized major players in lung cancer development. Cancer Sci. 2011;102(1):9–17.PubMedCrossRef
72.
go back to reference Sun C, Liu Z, Li S, Yang C, Xue R, Xi Y, Wang L, Wang S, He Q, Huang J, et al. Down-regulation of c-Met and Bcl2 by microRNA-206, activates apoptosis, and inhibits tumor cell proliferation, migration and colony formation. Oncotarget. 2015;6(28):25533–74.PubMedPubMedCentralCrossRef Sun C, Liu Z, Li S, Yang C, Xue R, Xi Y, Wang L, Wang S, He Q, Huang J, et al. Down-regulation of c-Met and Bcl2 by microRNA-206, activates apoptosis, and inhibits tumor cell proliferation, migration and colony formation. Oncotarget. 2015;6(28):25533–74.PubMedPubMedCentralCrossRef
73.
go back to reference Tang S, Pei R, Li K, Ma J, Zhang P, Lu Y, Liu X, Du X, Chen D, Sha K, et al. DNA methylation-mediated epigenetic silencing of miR-720 contributes to leukemogenesis in acute myeloid leukemia. Zhonghua Xue Ye Xue Za Zhi. 2014;35(11):1009–12.PubMed Tang S, Pei R, Li K, Ma J, Zhang P, Lu Y, Liu X, Du X, Chen D, Sha K, et al. DNA methylation-mediated epigenetic silencing of miR-720 contributes to leukemogenesis in acute myeloid leukemia. Zhonghua Xue Ye Xue Za Zhi. 2014;35(11):1009–12.PubMed
74.
go back to reference Zhou L, Zhao X, Han Y, Lu Y, Shang Y, Liu C, Li T, Jin Z, Fan D, Wu K. Regulation of UHRF1 by miR-146a/b modulates gastric cancer invasion and metastasis. FASEB J. 2013;27(12):4929–39.PubMedCrossRef Zhou L, Zhao X, Han Y, Lu Y, Shang Y, Liu C, Li T, Jin Z, Fan D, Wu K. Regulation of UHRF1 by miR-146a/b modulates gastric cancer invasion and metastasis. FASEB J. 2013;27(12):4929–39.PubMedCrossRef
75.
go back to reference Bandres E, Agirre X, Bitarte N, Ramirez N, Zarate R, Roman-Gomez J, Prosper F, Garcia-Foncillas J. Epigenetic regulation of microRNA expression in colorectal cancer. Int J Cancer. 2009;125(11):2737–43.PubMedCrossRef Bandres E, Agirre X, Bitarte N, Ramirez N, Zarate R, Roman-Gomez J, Prosper F, Garcia-Foncillas J. Epigenetic regulation of microRNA expression in colorectal cancer. Int J Cancer. 2009;125(11):2737–43.PubMedCrossRef
76.
go back to reference Cekaite L, Rantala JK, Bruun J, Guriby M, Agesen TH, Danielsen SA, Lind GE, Nesbakken A, Kallioniemi O, Lothe RA, et al. MiR-9, −31, and −182 deregulation promote proliferation and tumor cell survival in colon cancer. Neoplasia. 2012;14(9):868–79.PubMedPubMedCentralCrossRef Cekaite L, Rantala JK, Bruun J, Guriby M, Agesen TH, Danielsen SA, Lind GE, Nesbakken A, Kallioniemi O, Lothe RA, et al. MiR-9, −31, and −182 deregulation promote proliferation and tumor cell survival in colon cancer. Neoplasia. 2012;14(9):868–79.PubMedPubMedCentralCrossRef
77.
go back to reference Zhu M, Xu Y, Ge M, Gui Z, Yan F. Regulation of UHRF1 by microRNA-9 modulates colorectal cancer cell proliferation and apoptosis. Cancer Sci. 2015;106(7):833–9.PubMedPubMedCentralCrossRef Zhu M, Xu Y, Ge M, Gui Z, Yan F. Regulation of UHRF1 by microRNA-9 modulates colorectal cancer cell proliferation and apoptosis. Cancer Sci. 2015;106(7):833–9.PubMedPubMedCentralCrossRef
78.
go back to reference Yu T, Li J, Yan M, Liu L, Lin H, Zhao F, Sun L, Zhang Y, Cui Y, Zhang F, et al. MicroRNA-193a-3p and -5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene. 2015;34(4):413–23.PubMedCrossRef Yu T, Li J, Yan M, Liu L, Lin H, Zhao F, Sun L, Zhang Y, Cui Y, Zhang F, et al. MicroRNA-193a-3p and -5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene. 2015;34(4):413–23.PubMedCrossRef
79.
go back to reference Deng W, Yan M, Yu T, Ge H, Lin H, Li J, Liu Y, Geng Q, Zhu M, Liu L, et al. Quantitative proteomic analysis of the metastasis-inhibitory mechanism of miR-193a-3p in non-small cell lung cancer. Cell Physiol Biochem. 2015;35(5):1677–88.PubMedCrossRef Deng W, Yan M, Yu T, Ge H, Lin H, Li J, Liu Y, Geng Q, Zhu M, Liu L, et al. Quantitative proteomic analysis of the metastasis-inhibitory mechanism of miR-193a-3p in non-small cell lung cancer. Cell Physiol Biochem. 2015;35(5):1677–88.PubMedCrossRef
80.
go back to reference Matsushita R, Yoshino H, Enokida H, Goto Y, Miyamoto K, Yonemori M, Inoguchi S, Nakagawa M, Seki N. Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p): Inhibition of bladder cancer cell aggressiveness. Oncotarget. 2016;7(19):28460-87. doi: 10.18632/oncotarget.8668. Matsushita R, Yoshino H, Enokida H, Goto Y, Miyamoto K, Yonemori M, Inoguchi S, Nakagawa M, Seki N. Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p): Inhibition of bladder cancer cell aggressiveness. Oncotarget. 2016;7(19):28460-87. doi: 10.​18632/​oncotarget.​8668.
81.
go back to reference Goto Y, Kurozumi A, Nohata N, Kojima S, Matsushita R, Yoshino H, Yamazaki K, Ishida Y, Ichikawa T, Naya Y, et al. The microRNA signature of patients with sunitinib failure: regulation of UHRF1 pathways by microRNA-101 in renal cell carcinoma. Oncotarget. 2016. doi: 10.18632/oncotarget.10887. [Epub ahead of print]. Goto Y, Kurozumi A, Nohata N, Kojima S, Matsushita R, Yoshino H, Yamazaki K, Ishida Y, Ichikawa T, Naya Y, et al. The microRNA signature of patients with sunitinib failure: regulation of UHRF1 pathways by microRNA-101 in renal cell carcinoma. Oncotarget. 2016. doi: 10.​18632/​oncotarget.​10887. [Epub ahead of print].
82.
go back to reference Shi H, Zhou S, Liu J, Zhu J, Xue J, Gu L, Chen Y. miR-34a inhibits the in vitro cell proliferation and migration in human esophageal cancer. Pathol Res Pract. 2016;212(5):444–9.PubMedCrossRef Shi H, Zhou S, Liu J, Zhu J, Xue J, Gu L, Chen Y. miR-34a inhibits the in vitro cell proliferation and migration in human esophageal cancer. Pathol Res Pract. 2016;212(5):444–9.PubMedCrossRef
83.
go back to reference Manikandan M, Deva Magendhra Rao AK, Arunkumar G, Rajkumar KS, Rajaraman R, Munirajan AK. Down regulation of miR-34a and miR-143 may indirectly inhibit p53 in oral squamous cell carcinoma: a pilot study. Asian Pac J Cancer Prev. 2015;16(17):7619–25.PubMedCrossRef Manikandan M, Deva Magendhra Rao AK, Arunkumar G, Rajkumar KS, Rajaraman R, Munirajan AK. Down regulation of miR-34a and miR-143 may indirectly inhibit p53 in oral squamous cell carcinoma: a pilot study. Asian Pac J Cancer Prev. 2015;16(17):7619–25.PubMedCrossRef
84.
go back to reference Bhattacharya S, Ahir M, Patra P, Mukherjee S, Ghosh S, Mazumdar M, Chattopadhyay S, Das T, Chattopadhyay D, Adhikary A. PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a. Biomaterials. 2015;51:91–107.PubMedCrossRef Bhattacharya S, Ahir M, Patra P, Mukherjee S, Ghosh S, Mazumdar M, Chattopadhyay S, Das T, Chattopadhyay D, Adhikary A. PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a. Biomaterials. 2015;51:91–107.PubMedCrossRef
85.
go back to reference Abusnina A, Alhosin M, Keravis T, Muller CD, Fuhrmann G, Bronner C, Lugnier C. Down-regulation of cyclic nucleotide phosphodiesterase PDE1A is the key event of p73 and UHRF1 deregulation in thymoquinone-induced acute lymphoblastic leukemia cell apoptosis. Cell Signal. 2011;23(1):152–60.PubMedCrossRef Abusnina A, Alhosin M, Keravis T, Muller CD, Fuhrmann G, Bronner C, Lugnier C. Down-regulation of cyclic nucleotide phosphodiesterase PDE1A is the key event of p73 and UHRF1 deregulation in thymoquinone-induced acute lymphoblastic leukemia cell apoptosis. Cell Signal. 2011;23(1):152–60.PubMedCrossRef
86.
go back to reference Oldenborg PA. Role of CD47 in erythroid cells and in autoimmunity. Leuk Lymphoma. 2004;45(7):1319–27.PubMedCrossRef Oldenborg PA. Role of CD47 in erythroid cells and in autoimmunity. Leuk Lymphoma. 2004;45(7):1319–27.PubMedCrossRef
87.
go back to reference Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11(3):130–5.PubMedCrossRef Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11(3):130–5.PubMedCrossRef
88.
go back to reference Barclay AN, Van den Berg TK. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: structure, function, and therapeutic target. Annu Rev Immunol. 2014;32:25–50.PubMedCrossRef Barclay AN, Van den Berg TK. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: structure, function, and therapeutic target. Annu Rev Immunol. 2014;32:25–50.PubMedCrossRef
90.
go back to reference Cheng QS, Wang XB. CD47 and leukemia stem cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2010;18(4):1088–91.PubMed Cheng QS, Wang XB. CD47 and leukemia stem cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2010;18(4):1088–91.PubMed
91.
go back to reference Zhang L, Huang H. Targeting the cancer biomarker CD47: a review on the diverse mechanisms of the CD47 pathway in cancer treatment. Anticancer Agents Med Chem. 2016;16(6):658–67.PubMedCrossRef Zhang L, Huang H. Targeting the cancer biomarker CD47: a review on the diverse mechanisms of the CD47 pathway in cancer treatment. Anticancer Agents Med Chem. 2016;16(6):658–67.PubMedCrossRef
92.
go back to reference Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, Finetti P, Van Egmond M, Matozaki T, Kraal G, et al. CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A. 2011;108(45):18342–7.PubMedPubMedCentralCrossRef Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, Finetti P, Van Egmond M, Matozaki T, Kraal G, et al. CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A. 2011;108(45):18342–7.PubMedPubMedCentralCrossRef
93.
go back to reference Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109(17):6662–7.PubMedPubMedCentralCrossRef Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109(17):6662–7.PubMedPubMedCentralCrossRef
94.
go back to reference Edris B, Weiskopf K, Volkmer AK, Volkmer JP, Willingham SB, Contreras-Trujillo H, Liu J, Majeti R, West RB, Fletcher JA, et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc Natl Acad Sci U S A. 2012;109(17):6656–61.PubMedPubMedCentralCrossRef Edris B, Weiskopf K, Volkmer AK, Volkmer JP, Willingham SB, Contreras-Trujillo H, Liu J, Majeti R, West RB, Fletcher JA, et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc Natl Acad Sci U S A. 2012;109(17):6656–61.PubMedPubMedCentralCrossRef
95.
go back to reference Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs Jr KD, van Rooijen N, Weissman IL. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138(2):286–99.PubMedPubMedCentralCrossRef Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs Jr KD, van Rooijen N, Weissman IL. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138(2):286–99.PubMedPubMedCentralCrossRef
96.
go back to reference Kim D, Wang J, Willingham SB, Martin R, Wernig G, Weissman IL. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia. 2012;26(12):2538–45.PubMedCrossRef Kim D, Wang J, Willingham SB, Martin R, Wernig G, Weissman IL. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia. 2012;26(12):2538–45.PubMedCrossRef
97.
go back to reference Wang Y, Xu Z, Guo S, Zhang L, Sharma A, Robertson GP, Huang L. Intravenous delivery of siRNA targeting CD47 effectively inhibits melanoma tumor growth and lung metastasis. Mol Ther. 2013;21(10):1919–29.PubMedPubMedCentralCrossRef Wang Y, Xu Z, Guo S, Zhang L, Sharma A, Robertson GP, Huang L. Intravenous delivery of siRNA targeting CD47 effectively inhibits melanoma tumor growth and lung metastasis. Mol Ther. 2013;21(10):1919–29.PubMedPubMedCentralCrossRef
98.
go back to reference Broom OJ, Zhang Y, Oldenborg PA, Massoumi R, Sjolander A. CD47 regulates collagen I-induced cyclooxygenase-2 expression and intestinal epithelial cell migration. PLoS One. 2009;4(7):e6371.PubMedPubMedCentralCrossRef Broom OJ, Zhang Y, Oldenborg PA, Massoumi R, Sjolander A. CD47 regulates collagen I-induced cyclooxygenase-2 expression and intestinal epithelial cell migration. PLoS One. 2009;4(7):e6371.PubMedPubMedCentralCrossRef
99.
go back to reference Nogueira L, Ruiz-Ontanon P, Vazquez-Barquero A, Moris F, Fernandez-Luna JL. The NFkappaB pathway: a therapeutic target in glioblastoma. Oncotarget. 2011;2(8):646–53.PubMedPubMedCentralCrossRef Nogueira L, Ruiz-Ontanon P, Vazquez-Barquero A, Moris F, Fernandez-Luna JL. The NFkappaB pathway: a therapeutic target in glioblastoma. Oncotarget. 2011;2(8):646–53.PubMedPubMedCentralCrossRef
100.
go back to reference Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-kappaB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother. 2014;14(11):1293–306.PubMedPubMedCentralCrossRef Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-kappaB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother. 2014;14(11):1293–306.PubMedPubMedCentralCrossRef
101.
go back to reference Sick E, Boukhari A, Deramaudt T, Ronde P, Bucher B, Andre P, Gies JP, Takeda K. Activation of CD47 receptors causes proliferation of human astrocytoma but not normal astrocytes via an Akt-dependent pathway. Glia. 2011;59(2):308–19.PubMedCrossRef Sick E, Boukhari A, Deramaudt T, Ronde P, Bucher B, Andre P, Gies JP, Takeda K. Activation of CD47 receptors causes proliferation of human astrocytoma but not normal astrocytes via an Akt-dependent pathway. Glia. 2011;59(2):308–19.PubMedCrossRef
102.
go back to reference Sinha R, Yen PM. Cellular action of thyroid hormone. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, et al., editors. Endotext. South Dartmouth MA: MDText.com, Inc; 2000. Sinha R, Yen PM. Cellular action of thyroid hormone. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, et al., editors. Endotext. South Dartmouth MA: MDText.com, Inc; 2000.
103.
go back to reference Kim WG, Cheng SY. Thyroid hormone receptors and cancer. Biochim Biophys Acta. 2013;1830(7):3928–36.PubMedCrossRef Kim WG, Cheng SY. Thyroid hormone receptors and cancer. Biochim Biophys Acta. 2013;1830(7):3928–36.PubMedCrossRef
104.
go back to reference Wu SM, Cheng WL, Liao CJ, Chi HC, Lin YH, Tseng YH, Tsai CY, Chen CY, Lin SL, Chen WJ, et al. Negative modulation of the epigenetic regulator, UHRF1, by thyroid hormone receptors suppresses liver cancer cell growth. Int J Cancer. 2015;137(1):37–49.PubMedCrossRef Wu SM, Cheng WL, Liao CJ, Chi HC, Lin YH, Tseng YH, Tsai CY, Chen CY, Lin SL, Chen WJ, et al. Negative modulation of the epigenetic regulator, UHRF1, by thyroid hormone receptors suppresses liver cancer cell growth. Int J Cancer. 2015;137(1):37–49.PubMedCrossRef
105.
go back to reference Kim MY, Park SJ, Shim JW, Yang K, Kang HS, Heo K. Naphthazarin enhances ionizing radiation-induced cell cycle arrest and apoptosis in human breast cancer cells. Int J Oncol. 2015;46(4):1659–66.PubMed Kim MY, Park SJ, Shim JW, Yang K, Kang HS, Heo K. Naphthazarin enhances ionizing radiation-induced cell cycle arrest and apoptosis in human breast cancer cells. Int J Oncol. 2015;46(4):1659–66.PubMed
106.
go back to reference Jang SY, Hong D, Jeong SY, Kim JH. Shikonin causes apoptosis by up-regulating p73 and down-regulating ICBP90 in human cancer cells. Biochem Biophys Res Commun. 2015;465(1):71–6.PubMedCrossRef Jang SY, Hong D, Jeong SY, Kim JH. Shikonin causes apoptosis by up-regulating p73 and down-regulating ICBP90 in human cancer cells. Biochem Biophys Res Commun. 2015;465(1):71–6.PubMedCrossRef
107.
go back to reference Abusnina A, Keravis T, Yougbare I, Bronner C, Lugnier C. Anti-proliferative effect of curcumin on melanoma cells is mediated by PDE1A inhibition that regulates the epigenetic integrator UHRF1. Mol Nutr Food Res. 2011;55(11):1677–89.PubMedCrossRef Abusnina A, Keravis T, Yougbare I, Bronner C, Lugnier C. Anti-proliferative effect of curcumin on melanoma cells is mediated by PDE1A inhibition that regulates the epigenetic integrator UHRF1. Mol Nutr Food Res. 2011;55(11):1677–89.PubMedCrossRef
108.
go back to reference Chen H, Zhang T, Sheng Y, Zhang C, Peng Y, Wang X, Zhang C. Methylation profiling of multiple tumor suppressor genes in hepatocellular carcinoma and the epigenetic mechanism of 3OST2 regulation. J Cancer. 2015;6(8):740–9.PubMedPubMedCentralCrossRef Chen H, Zhang T, Sheng Y, Zhang C, Peng Y, Wang X, Zhang C. Methylation profiling of multiple tumor suppressor genes in hepatocellular carcinoma and the epigenetic mechanism of 3OST2 regulation. J Cancer. 2015;6(8):740–9.PubMedPubMedCentralCrossRef
109.
go back to reference Abu-Alainin W, Gana T, Liloglou T, Olayanju A, Barrera LN, Ferguson R, Campbell F, Andrews T, Goldring C, Kitteringham N, et al. UHRF1 regulation of the Keap1-Nrf2 pathway in pancreatic cancer contributes to oncogenesis. J Pathol. 2016;238(3):423–33.PubMedCrossRef Abu-Alainin W, Gana T, Liloglou T, Olayanju A, Barrera LN, Ferguson R, Campbell F, Andrews T, Goldring C, Kitteringham N, et al. UHRF1 regulation of the Keap1-Nrf2 pathway in pancreatic cancer contributes to oncogenesis. J Pathol. 2016;238(3):423–33.PubMedCrossRef
110.
go back to reference Parashar G, Capalash N. Promoter methylation-independent reactivation of PAX1 by curcumin and resveratrol is mediated by UHRF1. Clin Exp Med. 2016;16(3):471–8.PubMedCrossRef Parashar G, Capalash N. Promoter methylation-independent reactivation of PAX1 by curcumin and resveratrol is mediated by UHRF1. Clin Exp Med. 2016;16(3):471–8.PubMedCrossRef
111.
go back to reference Yu C, Xing F, Tang Z, Bronner C, Lu X, Di J, Zeng S, Liu J. Anisomycin suppresses Jurkat T cell growth by the cell cycle-regulating proteins. Pharmacol Rep. 2013;65(2):435–44.PubMedCrossRef Yu C, Xing F, Tang Z, Bronner C, Lu X, Di J, Zeng S, Liu J. Anisomycin suppresses Jurkat T cell growth by the cell cycle-regulating proteins. Pharmacol Rep. 2013;65(2):435–44.PubMedCrossRef
112.
go back to reference Krifa M, Leloup L, Ghedira K, Mousli M, Chekir-Ghedira L. Luteolin induces apoptosis in BE colorectal cancer cells by downregulating calpain, UHRF1, and DNMT1 expressions. Nutr Cancer. 2014;66(7):1220–7.PubMedCrossRef Krifa M, Leloup L, Ghedira K, Mousli M, Chekir-Ghedira L. Luteolin induces apoptosis in BE colorectal cancer cells by downregulating calpain, UHRF1, and DNMT1 expressions. Nutr Cancer. 2014;66(7):1220–7.PubMedCrossRef
113.
go back to reference Ding G, Chen P, Zhang H, Huang X, Zang Y, Li J, Li J, Wong J. Regulation of ubiquitin-like with plant homeodomain and RING finger domain 1 (UHRF1) protein stability by heat shock protein 90 chaperone machinery. J Biol Chem. 2016;291(38):20125–35.PubMedCrossRef Ding G, Chen P, Zhang H, Huang X, Zang Y, Li J, Li J, Wong J. Regulation of ubiquitin-like with plant homeodomain and RING finger domain 1 (UHRF1) protein stability by heat shock protein 90 chaperone machinery. J Biol Chem. 2016;291(38):20125–35.PubMedCrossRef
Metadata
Title
Signalling pathways in UHRF1-dependent regulation of tumor suppressor genes in cancer
Authors
Mahmoud Alhosin
Ziad Omran
Mazin A. Zamzami
Abdulrahman L. Al-Malki
Hani Choudhry
Marc Mousli
Christian Bronner
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2016
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-016-0453-5

Other articles of this Issue 1/2016

Journal of Experimental & Clinical Cancer Research 1/2016 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