Artigo Acesso aberto Revisado por pares

Multidimensional Proteomics Reveals a Role of UHRF2 in the Regulation of Epithelial-Mesenchymal Transition (EMT)

2016; Elsevier BV; Volume: 15; Issue: 7 Linguagem: Inglês

10.1074/mcp.m115.057448

ISSN

1535-9484

Autores

Mi Lai, Lizhu Liang, Jiwei Chen, Naiqi Qiu, Sai Ge, Shuhui Ji, Tieliu Shi, Bei Zhen, Mingwei Liu, Chen Ding, Yì Wáng, Jun Qin,

Tópico(s)

RNA modifications and cancer

Resumo

UHRF1 is best known for its positive role in the maintenance of DNMT1-mediated DNA methylation and is implicated in a variety of tumor processes. In this paper, we provided evidence to demonstrate a role of UHRF2 in cell motility and invasion through the regulation of the epithelial-mesenchymal transition (EMT) process by acting as a transcriptional co-regulator of the EMT-transcription factors (TFs). We ectopically expressed UHRF2 in gastric cancer cell lines and performed multidimensional proteomics analyses. Proteome profiling analysis suggested a role of UHRF2 in repression of cell-cell adhesion; analysis of proteome-wide TF DNA binding activities revealed the up-regulation of many EMT-TFs in UHRF2-overexpressing cells. These data suggest that UHRF2 is a regulator of cell motility and the EMT program. Indeed, cell invasion experiments demonstrated that silencing of UHRF2 in aggressive cells impaired their abilities of migration and invasion in vitro. Further ChIP-seq identified UHRF2 genomic binding motifs that coincide with several TF binding motifs including EMT-TFs, and the binding of UHRF2 to CDH1 promoter was validated by ChIP-qPCR. Moreover, the interactome analysis with IP-MS uncovered the interaction of UHRF2 with TFs including TCF7L2 and several protein complexes that regulate chromatin remodeling and histone modifications, suggesting that UHRF2 is a transcription co-regulator for TFs such as TCF7L2 to regulate the EMT process. Taken together, our study identified a role of UHRF2 in EMT and tumor metastasis and demonstrated an effective approach to obtain clues of UHRF2 function without prior knowledge through combining evidence from multidimensional proteomics analyses. UHRF1 is best known for its positive role in the maintenance of DNMT1-mediated DNA methylation and is implicated in a variety of tumor processes. In this paper, we provided evidence to demonstrate a role of UHRF2 in cell motility and invasion through the regulation of the epithelial-mesenchymal transition (EMT) process by acting as a transcriptional co-regulator of the EMT-transcription factors (TFs). We ectopically expressed UHRF2 in gastric cancer cell lines and performed multidimensional proteomics analyses. Proteome profiling analysis suggested a role of UHRF2 in repression of cell-cell adhesion; analysis of proteome-wide TF DNA binding activities revealed the up-regulation of many EMT-TFs in UHRF2-overexpressing cells. These data suggest that UHRF2 is a regulator of cell motility and the EMT program. Indeed, cell invasion experiments demonstrated that silencing of UHRF2 in aggressive cells impaired their abilities of migration and invasion in vitro. Further ChIP-seq identified UHRF2 genomic binding motifs that coincide with several TF binding motifs including EMT-TFs, and the binding of UHRF2 to CDH1 promoter was validated by ChIP-qPCR. Moreover, the interactome analysis with IP-MS uncovered the interaction of UHRF2 with TFs including TCF7L2 and several protein complexes that regulate chromatin remodeling and histone modifications, suggesting that UHRF2 is a transcription co-regulator for TFs such as TCF7L2 to regulate the EMT process. Taken together, our study identified a role of UHRF2 in EMT and tumor metastasis and demonstrated an effective approach to obtain clues of UHRF2 function without prior knowledge through combining evidence from multidimensional proteomics analyses. UHRF (ubiquitin-like, containing PHD and RING finger domains, inverted CCAAT Box-Binding Protein of 90 kDa) family contains four members characterized with multiple domains in structure (1Bronner C. Achour M. Arima Y. Chataigneau T. Saya H. Schini-Kerth V.B. The UHRF family: oncogenes that are drugable targets for cancer therapy in the near future?.Pharmacol. Therapeutics. 2007; 115: 419-434Crossref PubMed Scopus (138) Google Scholar). The founding member of this family, UHRF1 is the best characterized. UHRF1 has been reported to play critical roles in various processes, including the maintenance of DNMT1-mediated DNA methylation. UHRF1 binds to hemi-methylated DNA through its SRA domain and therefore plays an important role in targeting DNMT1 to sites of replication (2Sharif J. Muto M. Takebayashi S. Suetake I. Iwamatsu A. Endo T.A. Shinga J. Mizutani-Koseki Y. Toyoda T. Okamura K. Tajima S. Mitsuya K. Okano M. Koseki H. The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA.Nature. 2007; 450: 908-912Crossref PubMed Scopus (924) Google Scholar, 3Bostick M. Kim J.K. Estève P.-O. Clark A. Pradhan S. Jacobsen S.E. UHRF1 plays a role in maintaining DNA methylation in mammalian cells.Science. 2007; 317: 1760-1764Crossref PubMed Scopus (982) Google Scholar, 4Hashimoto H. Horton J.R. Zhang X. Bostick M. Jacobsen S.E. Cheng X. The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix.Nature. 2008; 455: 826-829Crossref PubMed Scopus (332) Google Scholar, 5Avvakumov G.V. Walker J.R. Xue S. Li Y. Duan S. Bronner C. Arrowsmith C.H. Dhe-Paganon S. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1.Nature. 2008; 455: 822-825Crossref PubMed Scopus (365) Google Scholar, 6Arita 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: 818-821Crossref PubMed Scopus (373) Google Scholar, 7Qian C. Li S. Jakoncic J. Zeng L. Walsh M.J. Zhou M.M. Structure and hemimethylated CpG binding of the SRA domain from human UHRF1.J. Biol. Chem. 2008; 283: 34490-34494Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar, 8Rottach A. Frauer C. Pichler G. Bonapace I.M. Spada F. Leonhardt H. The multi-domain protein Np95 connects DNA methylation and histone modification.Nucleic Acids Res. 2010; 38: 1796-1804Crossref PubMed Scopus (127) Google Scholar). In addition, UHRF1 recognizes dimethylated/trimethylated lysine 9 of histone H3 (H3K9me2/3) via its PHD and TTD domains (8Rottach A. Frauer C. Pichler G. Bonapace I.M. Spada F. Leonhardt H. The multi-domain protein Np95 connects DNA methylation and histone modification.Nucleic Acids Res. 2010; 38: 1796-1804Crossref PubMed Scopus (127) Google Scholar, 9Citterio E. Papait R. Nicassio F. Vecchi M. Gomiero P. Mantovani R. Di Fiore P.P. Bonapace I.M. Np95 is a histone-binding protein endowed with ubiquitin ligase activity.Mol. Cell. Biol. 2004; 24: 2526-2535Crossref PubMed Scopus (157) Google Scholar, 10Karagianni P. Amazit L. Qin J. Wong J. ICBP90, a novel methyl K9 H3 binding protein linking protein ubiquitination with heterochromatin formation.Mol. Cell. Biol. 2008; 28: 705-717Crossref PubMed Scopus (191) Google Scholar, 11Nady N. Lemak A. Walker J.R. Avvakumov G.V. Kareta M.S. Achour M. Xue S. Duan S. Allali-Hassani A. Zuo X. Wang Y.X. Bronner C. Chedin F. Arrowsmith C.H. Dhe-Paganon S. Recognition of multivalent histone states associated with heterochromatin by UHRF1 protein.J. Biol. Chem. 2011; 286: 24300-24311Abstract Full Text Full Text PDF PubMed Scopus (154) Google Scholar, 12Arita K. Isogai S. Oda T. Unoki M. Sugita K. Sekiyama N. Kuwata K. Hamamoto R. Tochio H. Sato M. Recognition of modification status on a histone H3 tail by linked histone reader modules of the epigenetic regulator UHRF1.Proc. Natl. Acad. Sci. U. S. A. 2012; 109: 12950-12955Crossref PubMed Scopus (140) Google Scholar, 13Rothbart S.B. Krajewski K. Nady N. Tempel W. Xue S. Badeaux A.I. Barsyte-Lovejoy D. Martinez J.Y. Bedford M.T. Fuchs S.M. Arrowsmith C.H. Strahl B.D. Association of UHRF1 with methylated H3K9 directs the maintenance of DNA methylation.Nat. Structural Mol. Biol. 2012; 19: 1155-1160Crossref PubMed Scopus (264) Google Scholar, 14Xie S. Jakoncic J. Qian C. UHRF1 double tudor domain and the adjacent PHD finger act together to recognize K9me3-containing histone H3 tail.J. Mol. Biol. 2012; 415: 318-328Crossref PubMed Scopus (71) Google Scholar, 15Cheng J. Yang Y. Fang J. Xiao J. Zhu T. Chen F. Wang P. Li Z. Yang H. Xu Y. Structural insight into coordinated recognition of trimethylated histone H3 lysine 9 (H3K9me3) by the plant homeodomain (PHD) and tandem tudor domain (TTD) of UHRF1 (ubiquitin-like, containing PHD and RING finger domains, 1) protein.J. Biol. Chem. 2013; 288: 1329-1339Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar), and exerts ubiquitin E3 ligase activity on DNMT1 and histone substrates via its RING domain (9Citterio E. Papait R. Nicassio F. Vecchi M. Gomiero P. Mantovani R. Di Fiore P.P. Bonapace I.M. Np95 is a histone-binding protein endowed with ubiquitin ligase activity.Mol. Cell. Biol. 2004; 24: 2526-2535Crossref PubMed Scopus (157) Google Scholar, 10Karagianni P. Amazit L. Qin J. Wong J. ICBP90, a novel methyl K9 H3 binding protein linking protein ubiquitination with heterochromatin formation.Mol. Cell. Biol. 2008; 28: 705-717Crossref PubMed Scopus (191) Google Scholar, 16Qin W. Leonhardt H. Spada F. Usp7 and Uhrf1 control ubiquitination and stability of the maintenance DNA methyltransferase Dnmt1.J. Cell. Biochem. 2011; 112: 439-444Crossref PubMed Scopus (114) Google Scholar, 17Du Z. Song J. Wang Y. Zhao Y. Guda K. Yang S. Kao H.-Y. Xu Y. Willis J. Markowitz S.D. DNMT1 stability is regulated by proteins coordinating deubiquitination and acetylation-driven ubiquitination.Sci. Signaling. 2010; 3: ra80Crossref PubMed Scopus (238) Google Scholar). Importantly, several studies have demonstrated the oncogenic role of UHRF1 in tumors by promoting proliferation and metastasis of cancer cells (18Mudbhary R. Hoshida Y. Chernyavskaya Y. Jacob V. Villanueva A. Fiel M.I. Chen X. Kojima K. Thung S. Bronson R.T. Lachenmayer A. Revill K. Alsinet C. Sachidanandam R. Desai A. SenBanerjee S. Ukomadu C. Llovet J.M. Sadler K.C. UHRF1 overexpression drives DNA hypomethylation and hepatocellular carcinoma.Cancer Cell. 2014; 25: 196-209Abstract Full Text Full Text PDF PubMed Scopus (219) Google Scholar, 19Zhou 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: 4929-4939Crossref PubMed Scopus (86) Google Scholar, 20Guan D. Factor D. Liu Y. Wang Z. Kao H.Y. The epigenetic regulator UHRF1 promotes ubiquitination-mediated degradation of the tumor-suppressor protein promyelocytic leukemia protein.Oncogene. 2013; 32: 3819-3828Crossref PubMed Scopus (52) Google Scholar, 21Bronner 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: 1643-1649Crossref PubMed Scopus (77) Google Scholar, 22Li X.L. Xu J.H. Nie J.H. Fan S.J. Exogenous expression of UHRF1 promotes proliferation and metastasis of breast cancer cells.Oncol. Rep. 2012; 28: 375-383PubMed Google Scholar, 23Babbio F. Pistore C. Curti L. Castiglioni I. Kunderfranco P. Brino L. Oudet P. Seiler R. Thalman G.N. Roggero E. Sarti M. Pinton S. Mello-Grand M. Chiorino G. Catapano C.V. Carbone G.M. Bonapace I.M. The SRA protein UHRF1 promotes epigenetic crosstalks and is involved in prostate cancer progression.Oncogene. 2012; 31: 4878-4887Crossref PubMed Scopus (99) Google Scholar, 24Unoki M. Daigo Y. Koinuma J. Tsuchiya E. Hamamoto R. Nakamura Y. UHRF1 is a novel diagnostic marker of lung cancer.Br. J. Cancer. 2010; 103: 217-222Crossref PubMed Scopus (97) Google Scholar, 25Unoki M. Kelly J. Neal D. Ponder B. Nakamura Y. Hamamoto R. UHRF1 is a novel molecular marker for diagnosis and the prognosis of bladder cancer.Br. J. Cancer. 2009; 101: 98-105Crossref PubMed Scopus (94) Google Scholar, 26Unoki 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: 1279-1288Crossref PubMed Scopus (83) Google Scholar). UHRF2 has similar sequence and domain architectures as UHRF1. This similarity suggests a potential functional conservation between these two proteins. Like UHRF1, UHRF2 also recognizes hemimethylated DNA substrates and H3K9me2/3, and interacts with DNMTs and histone methyltransferase G9a in vitro (27Mori T. Ikeda D.D. Yamaguchi Y. Unoki M. Project N. NIRF/UHRF2 occupies a central position in the cell cycle network and allows coupling with the epigenetic landscape.FEBS Lett. 2012; 586: 1570-1583Crossref PubMed Scopus (34) Google Scholar). However, there are substantial differences between UHRF1 and UHRF2. UHRF2 cannot rescue the DNA methylation defect in Uhrf1−/− ES cells because of its inability to recruit DNMT1 to replication foci during S phase (28Zhang J. Gao Q. Li P. Liu X. Jia Y. Wu W. Li J. Dong S. Koseki H. Wong J. S phase-dependent interaction with DNMT1 dictates the role of UHRF1 but not UHRF2 in DNA methylation maintenance.Cell Res. 2011; 21: 1723-1739Crossref PubMed Scopus (91) Google Scholar). In addition, UHRF2 was reported as a specific binder of 5hmC with its SRA domain, whereas UHRF1-SRA does not have this binding preference (29Spruijt C.G. Gnerlich F. Smits A.H. Pfaffeneder T. Jansen P.W. Bauer C. Munzel M. Wagner M. Muller M. Khan F. Eberl H.C. Mensinga A. Brinkman A.B. Lephikov K. Muller U. Walter J. Boelens R. van Ingen H. Leonhardt H. Carell T. Vermeulen M. Dynamic readers for 5-(hydroxy)methylcytosine and its oxidized derivatives.Cell. 2013; 152: 1146-1159Abstract Full Text Full Text PDF PubMed Scopus (760) Google Scholar, 30Zhou T. Xiong J. Wang M. Yang N. Wong J. Zhu B. Xu R.M. Structural basis for hydroxymethylcytosine recognition by the SRA domain of UHRF2.Mol. Cell. 2014; 54: 879-886Abstract Full Text Full Text PDF PubMed Scopus (77) Google Scholar). Unlike UHRF1, which is often found in ESCs, UHRF2 is more commonly expressed in differentiated cells. A function of UHRF2 in regulation of cell cycle was also speculated. UHRF2 was found to interact with cyclins, CDKs, p53, pRB, PCNA, and was able to induce G1 arrest by ubiquitinating cyclins D1 and E1 (31Li Y. Mori T. Hata H. Homma Y. Kochi H. NIRF induces G1 arrest and associates with Cdk2.Biochem. Biophys. Res. Commun. 2004; 319: 464-468Crossref PubMed Scopus (35) Google Scholar, 32Mori T. Ikeda D.D. Fukushima T. Takenoshita S. Kochi H. NIRF constitutes a nodal point in the cell cycle network and is a candidate tumor suppressor.Cell Cycle. 2011; 10: 3284-3299Crossref PubMed Scopus (34) Google Scholar). Other substrates of UHRF2 E3 Ub ligase include PCNP, nuclear aggregates containing polyglutamine repeats, hepatitis B virus core protein and zinc finger protein 131 (ZNF131) (33Iwata A. Nagashima Y. Matsumoto L. Suzuki T. Yamanaka T. Date H. Deoka K. Nukina N. Tsuji S. Intranuclear degradation of polyglutamine aggregates by the ubiquitin-proteasome system.J. Biol. Chem. 2009; 284: 9796-9803Abstract Full Text Full Text PDF PubMed Scopus (76) Google Scholar, 34Mori T. Li Y. Hata H. Kochi H. NIRF is a ubiquitin ligase that is capable of ubiquitinating PCNP, a PEST-containing nuclear protein.FEBS Lett. 2004; 557: 209-214Crossref PubMed Scopus (51) Google Scholar, 35Qian G. Jin F. Chang L. Yang Y. Peng H. Duan C. NIRF, a novel ubiquitin ligase, interacts with hepatitis B virus core protein and promotes its degradation.Biotechnol. Lett. 2012; 34: 29-36Crossref PubMed Scopus (24) Google Scholar, 36Oh Y. Chung K.C. UHRF2, a ubiquitin E3 ligase, acts as a small ubiquitin-like modifier E3 ligase for zinc finger protein 131. J.Biol. Chem. 2013; 288: 9102-9111Abstract Full Text Full Text PDF Scopus (20) Google Scholar). Like UHRF1, UHRF2 was also implicated in tumors; but reports about the role of UHRF2 in tumors are contradictory and uncertain. Some studies demonstrated that UHRF2 behaves like a tumor suppressor to inhibit the inappropriate cell cycle progression (31Li Y. Mori T. Hata H. Homma Y. Kochi H. NIRF induces G1 arrest and associates with Cdk2.Biochem. Biophys. Res. Commun. 2004; 319: 464-468Crossref PubMed Scopus (35) Google Scholar, 32Mori T. Ikeda D.D. Fukushima T. Takenoshita S. Kochi H. NIRF constitutes a nodal point in the cell cycle network and is a candidate tumor suppressor.Cell Cycle. 2011; 10: 3284-3299Crossref PubMed Scopus (34) Google Scholar), whereas other studies suggested potential oncogenic characteristics of UHRF2 with up-regulated expression in cancers (37Wu J. Liu S. Liu G. Dombkowski A. Abrams J. Martin-Trevino R. Wicha M.S. Ethier S.P. Yang Z.Q. Identification and functional analysis of 9p24 amplified genes in human breast cancer.Oncogene. 2012; 31: 333-341Crossref PubMed Scopus (64) Google Scholar, 38Lu S. Yan D. Wu Z. Jiang T. Chen J. Yuan L. Lin J. Peng Z. Tang H. Ubiquitin-like with PHD and ring finger domains 2 is a predictor of survival and a potential therapeutic target in colon cancer.Oncol. Reports. 2014; 31: 1802-1810Crossref PubMed Scopus (18) Google Scholar, 39Wang F. Zhang P. Ma Y. Yang J. Moyer M.P. Shi C. Peng J. Qin H. NIRF is frequently upregulated in colorectal cancer and its oncogenicity can be suppressed by let-7a microRNA.Cancer Lett. 2012; 314: 223-231Crossref PubMed Scopus (47) Google Scholar, 40Wu T.-F. Zhang W. Su Z.-P. Chen S.-S. Chen G.-L. Wei Y.-X. Sun T. Xie X.-S. Li B. Zhou Y.-X. Du Z.-W. UHRF2 mRNA Expression is Low in Malignant Glioma but Silencing Inhibits the Growth of U251 Glioma Cells in vitro.Asian Pacific J. Cancer Prevention. 2012; 13: 5137-5142Crossref PubMed Scopus (11) Google Scholar). Metastasis is an important characteristics of cancer and responsible for more than 90% of cancer associated mortality. Metastasis of cancer cells is a complex process that is partly regulated by activation of epithelial to mesenchymal transition (EMT) 1The abbreviations used are:EMTEpithelial to mesenchymal transitionWCEwhole cell extractsNEnucleus extractsTFREtranscription factor response elementssRPmanual RPLC-MS/MSLiquid chromatography - tandem mass spectrometryFDRfalse discovery rateiBAQintensity based absolute quantificationFOTfraction of totalLFQlabel-free quantificationTFstranscription factorsCoRscoregulatorsGOgene ontologygRNAguide RNAq-PCRquantitative real time PCRChIPchromatin immunoprecipitation. to acquire the ability to invade and metastasize (41Spano D. Heck C. De Antonellis P. Christofori G. Zollo M. Molecular networks that regulate cancer metastasis.Semin. Cancer Biol. 2012; 22: 234-249Crossref PubMed Scopus (275) Google Scholar). During EMT, epithelial cells lose cell-cell contacts and cell polarity, and acquire mesenchymal-like characteristics with increased ability of migration and invasion. EMT is orchestrated by transcription factor cascades that regulate the expression of proteins involved in cell-cell contacts, cell polarity, cytoskeleton structure and extracellular matrix degradation. For instance, EMT-TFs repress one of the key epithelial genes E-cadherin through binding the promoter region of CDH1 directly or indirectly. The reported key EMT-TFs include SNAIL1/2, TWIST1/2, ZEB1/2, TCF3 and FOXC2 (42Lamouille S. Xu J. Derynck R. Molecular mechanisms of epithelial-mesenchymal transition.Nature reviews. Mol. Cell Biol. 2014; 15: 178-196Crossref PubMed Scopus (5230) Google Scholar, 43Puisieux A. Brabletz T. Caramel J. Oncogenic roles of EMT-inducing transcription factors.Nat. Cell Biol. 2014; 16: 488-494Crossref PubMed Scopus (707) Google Scholar, 44De Craene B. Berx G. Regulatory networks defining EMT during cancer initiation and progression.Nat. Rev. Cancer. 2013; 13: 97-110Crossref PubMed Scopus (1899) Google Scholar, 45Tam W.L. Weinberg R.A. The epigenetics of epithelial-mesenchymal plasticity in cancer.Nat. Med. 2013; 19: 1438-1449Crossref PubMed Scopus (883) Google Scholar). Because of the limited number of studies on the involvement of UHRF2 in tumorigenesis, the precise biological functions of UHRF2 in cancer and whether it also functions like UHRF1 remain to be investigated. Epithelial to mesenchymal transition whole cell extracts nucleus extracts transcription factor response elements manual RP Liquid chromatography - tandem mass spectrometry false discovery rate intensity based absolute quantification fraction of total label-free quantification transcription factors coregulators gene ontology guide RNA quantitative real time PCR chromatin immunoprecipitation. MS-based proteomics is a powerful approach for large scale protein analysis in biological research (46Aebersold R. Mann M. Mass spectrometry-based proteomics.Nature. 2003; 422: 198-207Crossref PubMed Scopus (5585) Google Scholar, 47Cravatt B.F. Simon G.M. Yates Iii, J.R. The biological impact of mass-spectrometry-based proteomics.Nature. 2007; 450: 991-1000Crossref PubMed Scopus (571) Google Scholar). Our lab has developed a fast, label-free quantification workflow (Fast-quan) for protein identification, in which 7,000 proteins can be identified and quantified with 12 h of MS running time (48Ding C. Jiang J. Wei J. Liu W. Zhang W. Liu M. Fu T. Lu T. Song L. Ying W. Chang C. Zhang Y. Ma J. Wei L. Malovannaya A. Jia L. Zhen B. Wang Y. He F. Qian X. Qin J. A fast workflow for identification and quantification of proteomes.Mol. Cell. Proteomics. 2013; 12: 2370-2380Abstract Full Text Full Text PDF PubMed Scopus (89) Google Scholar). This has enabled analysis of multiple samples. We also developed a concatenated tandem array of transcription factor response elements (catTFRE) pull-down assay that allows for enrichment and identification of endogenous transcription factors (TFs) (49Ding C. Chan D.W. Liu W. Liu M. Li D. Song L. Li C. Jin J. Malovannaya A. Jung S.Y. Zhen B. Wang Y. Qin J. Proteome-wide profiling of activated transcription factors with a concatenated tandem array of transcription factor response elements.Proc. Natl. Acad. Sci. U.S.A. 2013; 110: 6771-6776Crossref PubMed Scopus (81) Google Scholar). The combination of measuring changes in DNA binding activity of TFs and proteome-wide profiling of protein abundance allows us to correlate TF activity with target genes in response to exogenous stimulation. Thus, the proteome-wide identification of activated TFs when cells are perturbed can provide important biological clues about the mechanisms and signal transduction pathways. Current UHRF researches focused on how UHRF proteins impact genome DNA methylation. This direction is important in studying cancer initiation when changes in UHRF proteins can reprogram the epigenome. It is entirely not clear whether and what roles UHRF2 may play when cells become cancerous. We thus ectopically expressed UHRF2 in gastric cancer cell lines and performed multidimensional proteomics analyses to obtain clues for UHRF2 functions in a consistent manner. The MS profiling revealed down-regulation of a number of epithelial markers including CDH1, JUP, TJP1, DSG2, INADL, CXADR, SPINT1, and TJP2. The catTFRE-MS analysis also demonstrated up-regulation of multiple key transcription factors involved in EMT, including TWIST2, FOXC2, and TCF family of transcription factors. Furthermore, we demonstrated that silencing UHRF2 in gastric cancer cells could inhibit the ability of cell migration and invasion in vitro. Together, these results suggested that UHRF2 play a role in tumor metastasis. SGC7901, MKN74, N87, and MKN45 human gastric cancer cell lines were cultured in DMEM supplemented with 10% FBS (fetal bovine serum) (Gibco, Carlsbad, CA), 1% Penicillin-Streptomycin (Sigma-Aldrich, St. Louis, MO) and incubated at 37 °C and 5% CO2. Antibody specific to UHRF2 was generated against a recombinant GST-tagged UHRF2 (N-terminal 351aa) fragment. Specific antibody was purified from serum of immunized rabbit with His-tagged-UHRF2 protein. The specific recognition of endogenous UHRF2 by the antibody was confirmed by IP-MS. The human UHRF2 coding sequence was amplified by PCR from cDNA libraries. The amplified fragment was cloned into the pENTR-vector. The lentiviral vector containing UHRF2 cDNA was constructed by recombination of pHAGE-EF-ZsG-DEST with pENTR. The sequence was verified by DNA sequencing. Lentivirus supernatants were collected 48 h after transfecting pHAGE-EF-UHRF2-ZsG-DEST with the packaging vectors pMD2.G and psPAX2 into 293T cells using lipofectamine 2000 reagent (Invitrogen). The lentivirus packaged with the empty pHAGE-EF-ZsG-DEST vector was used as control. The concentrated and purified lentivirus from supernatants were used to infect cells with 8 μg/ml Polybrene to generated control or UHRF2-expressing stable cells of SGC7901, MKN74, N87, and MKN45. The expression of UHRF2 in different cell lines was analyzed by Western blot. CatTFRE was done as previously described (49Ding C. Chan D.W. Liu W. Liu M. Li D. Song L. Li C. Jin J. Malovannaya A. Jung S.Y. Zhen B. Wang Y. Qin J. Proteome-wide profiling of activated transcription factors with a concatenated tandem array of transcription factor response elements.Proc. Natl. Acad. Sci. U.S.A. 2013; 110: 6771-6776Crossref PubMed Scopus (81) Google Scholar). Briefly, the catTFRE DNA was prepared by PCR amplification with biotinylated primers. Nuclear extracts (NEs) were prepared with NE-PER nuclear extraction reagents (Thermo Fisher Scientific, Boston, MA). Two milligram of control or OE NEs was used to incubate with pre-immobilized biotinylated catTFRE on Dynabeads (Dynabeads M-280 Streptavidin, Invitrogen, Carlsbad, CA). The mixture was supplemented with EDTA to a final concentration of 1 mm and adjusted with NaCl to 200–250 mm total salt concentration and incubated for 2h at 4 °C. The supernatant was discarded and Dynabeads were washed with NETN [100 mm NaCl, 20 mm Tris-Cl, 0.5 mm EDTA, and 0.5% (v/v) Nonidet P-40] twice followed by PBS twice. Beads were added to 20 μl 2× loading buffer and incubated in 95 °C for 5 min. The supernatant was put on SDS-PAGE for separation. SDS-PAGE gels were stained with Coomassie Blue R-250 to visualize the protein bands and each lane was cut into 12 gel slices. In-gel trypsin digestion was performed. After incubation with rotation for overnight at 37 °C, the digested products were extracted with acetonitrile (ACN) and dried in vacuum. Whole cellular lysates were extracted using 8 m urea. MKN74 control and UHRF2-OE cells (MKN74 is a gastric cancer cell line with low invasiveness, which expresses low level of UHRF2 and high level of CDH1 on its surface) were lysed with 8 m urea containing protease inhibitor PMSF for 30min at 4 °C. The lysate was centrifuged at 24,000 × g and the supernatant was collected as whole cell extracts (WCE). Protein concentration was determined by BCA assay. Twenty micrograms of control and OE proteins were digested with trypsin. Tryptic peptides were separated on a C18 column with acetonitrile of different percentage as 6%, 9%, 12%, 15%, 18%, 21%, 25%, 30%, and 35%. Nine separations were combined to six fractions and dried in vacuum. Peptides were stored at −80 °C until re-dissolved for MS analysis. NEs were prepared with NE-PER Kit (Thermo) from control and UHRF2-OE cells of N87, MKN45, SGC7901, and MKN74. Equal amounts (1 mg∼10 mg) of NEs from control and OE cells were incubated with 5 μg UHRF2 antibody. The incubation solution was adjusted with NaCl to 200 mm total salt concentration and incubated at 4 °C for overnight. After the addition of 30 μl protein A/G-Sepharose for each IP reaction and incubation for another 2h, the immunoprecipitate was washed with NETN twice followed by PBS buffer twice. The Sepharose beads were re-suspended in 20 μl 2× loading buffer and incubated at 95 °C for 5 min. The supernatant was resolved by 10% SDS-PAGE and in-gel trypsin digestion was performed after the gel was sliced. Dried peptide samples were re-dissolved in solvent A (0.1% formic acid in water). Liquid chromatography - tandem mass spectrometry (LC-MS/MS) analysis was performed with Q-Exactive Plus or FUSION mass spectrometer (Thermo) equipped with an online Easy-nLC 1000 nano-HPLC system (Thermo). The injected peptides were separated on a reversed phase nano-HPLC C18 column (Pre-column: 5 μm, 300 Å, 2 cm × 100 μm ID; analytical column: 3 μm, 120 Å, 15 cm × 75 μm ID) at a flow rate of 350 nl/min with a 75-min gradient of 3 to 30% solvent B (0.1% formic acid in acetonitrile). For the detection with FUSION mass spectrometry, a precursor scan was measured in the Orbitrap by scanning from m/z 300–1400 with a resolution of 120,000. Ions selected under top-speed mode were isolated in Quadrupole and fragmented by higher energy collision-induced dissociation (HCD) with normalized collision energy of 35%, then measured in the linear ion trap. Typical mass spectrometric conditions were: AGC targets were 5e5 ions for Orbitrap scans and 5e3 for MS/MS scans; dynamic exclusion was employed for 18 s. For the Q-Exactive Plus, the instrument was operated in the data-dependent acquisition mode with a resolution of 70,000 at full scan mode and 17,500 at MS/MS mode. The full scan was processed in the Orbitrap from m/z 300–1400, the top 20 most intense ions in each scan were automatically selected for HCD fragmentation with normalized collision energy of 27% and measured in Orbitrap. Typical mass spectrometric conditions were: AGC targets were 3e6 ions for full scans and 5e4 for MS/MS scans; dynamic exclusion was employed for 18 s. The acquired MS/MS spectra were searched by Mascot 2.3 (Matrix Science Inc, MA) implemented on Proteome Discoverer 1.4 (Thermo) against the human National Center for Biotechnology Information (NCBI) RefSeq protein databases (updated on April 7, 2013, 32,015 protein entries). The parameter settings were: the mass tolerances were 20 ppm for precursor and 50mmu for product ions from Q-Exactive Plus and 20 ppm for precursor and 0.5 Da for product ions from FUSION respectively; two missed cleavages were allowed; the fixed modification was set as carbamidomethyl (C), dynamic modifications were protein acetyl (protein N-term), oxidation(M) for profiling data; dynamic modification for catTFRE were phosphor (Y), phosphor (ST), deStreak (

Referência(s)