Identifying the role of Wilms tumor 1 associated protein in cancer prediction using integrative genomic analyses

  • Authors:
    • Li‑Sheng Wu
    • Jia‑Yi Qian
    • Minghai Wang
    • Haiwei Yang
  • View Affiliations

  • Published online on: July 18, 2016     https://doi.org/10.3892/mmr.2016.5528
  • Pages: 2823-2831
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The Wilms tumor suppressor, WT1 was first identified due to its essential role in the normal development of the human genitourinary system. Wilms tumor 1 associated protein (WTAP) was subsequently revealed to interact with WT1 using yeast two-hybrid screening. The present study identified 44 complete WTAP genes in the genomes of vertebrates, including fish, amphibians, birds and mammals. The vertebrate WTAP proteins clustered into the primate, rodent and teleost lineages using phylogenetic tree analysis. From 1,347 available SNPs in the human WTAP gene, 19 were identified to cause missense mutations. WTAP was expressed in bladder, blood, brain, breast, colorectal, esophagus, eye, head and neck, lung, ovarian, prostate, skin and soft tissue cancers. A total of 17 out of 328 microarrays demonstrated an association between WTAP gene expression and cancer prognosis. However, the association between WTAP gene expression and prognosis varied in distinct types of cancer, and even in identical types of cancer from separate microarray databases. By searching the Catalogue of Somatic Mutations in Cancer database, 65 somatic mutations were identified in the human WTAP gene from the cancer tissue samples. These results suggest that the function of WTAP in tumor formation may be multidimensional. Furthermore, signal transducer and activator of transcription 1, forkhead box protein O1, interferon regulatory factor 1, glucocorticoid receptor and peroxisome proliferator-activated receptor γ transcription factor binding sites were identified in the upstream (promoter) region of the human WTAP gene, suggesting that these transcription factors may be involved in WTAP functions in tumor formation.

Introduction

The Wilms tumor suppressor gene WT1 was first identified due to its essential role in the normal development of the human genitourinary system (1). WT1 functions as a transcription factor regulating target gene expression (1). In addition, WT1 was revealed to regulate the expression of genes involved in the Wnt signaling pathway via a genome-wide screening analysis (2).

Wilms tumor 1 associated protein (WTAP) was demonstrated to interact with WT1 using a yeast two-hybrid screening (3). WTAP and WT1 were observed to localize to the nucleoplasm and nuclear speckles, where they were partially co-localized with splicing factors (3). WTAP is the mammalian homolog of the Drosophila gene female-lethal(2) D [fl(2)D], and fl(2)D is involved in activating female-specific patterns of alternative splicing of sex-lethal and transformer pre-mRNA (4,5). Previous studies demonstrated that WTAP-null and heterozygous mice succumbed between embryonic day 6.5 and 10.5, and exhibited marked defects in cell proliferation, which resulted in defects in endoderm and mesoderm formation (6,7). Furthermore, it has been demonstrated in mice that WTAP is required for G2/M cell cycle transition by stabilizing the cyclin A2 mRNA and, thus, is vital in early development (6). In addition, WTAP is involved in the function of human spliceosomes (8). Recently, it was demonstrated that WTAP and virilizer are subunits of the N6-methyladenosine methylation complex, which regulates mRNA stability (9,10).

A limited number of studies have been performed on the role of WTAP in tumor genesis. WTAP is overexpressed in cholangiocarcinoma and WTAP expression was observed to correlate with metastasis (11). In addition, WTAP was demonstrated to be overexpressed in glioblastoma, and regulated glioblastoma cell migration and invasion (12). Bansal et al (13) identified WTAP as an oncogenic protein in acute myeloid leukemia. Carbonic anhydrase 4, a novel tumor suppressor in colorectal cancer, inhibited the Wnt signaling pathway by targeting the WTAP-WT1-transducin β-like 1 axis (14). The aim of the present study was to investigate the role of WTAP in tumor formation by identifying novel WTAP genes in vertebrate genomes. The expression of these genes in healthy and tumor tissue samples was determined, and functionally relevant single nucleotide polymorphisms (SNPs) and somatic mutations in WTAP were identified. Conserved transcription factor binding sites within the promoter region of the human WTAP gene were identified. Furthermore, meta-analysis of the prognostic value of WTAP gene expression in various cancers was performed.

Materials and methods

Identification of novel WTAP genes in vertebrate genomes and transcription factor-binding sites

The DNA and amino acid sequences of novel vertebrate WTAPs were obtained by searching Ensembl genome databases (ensembl.org/index.html) using orthologous and paralogous associations. The prospective WTAP sequences were confirmed using the Basic Local Alignment Search Tool (BLAST; http://blast.ncbi.nlm.nih.gov/Blast.cgi) (1518). Conserved transcription factor-binding sites in the promoter regions of the human WTAP gene were identified from the SABiosciences' proprietary database, DECODE (Qiagen, Inc., Valencia, CA, USA), which combines text mining with the University of California, Santa Cruz genome browser data (1921).

Comparative proteomic analyses of WTAP proteins

The amino acid sequences of identified vertebrate WTAPs were aligned using ClustalW (ebi.ac.uk/Tools/msa/). A maximum likelihood (ML) tree of vertebrate WTAPs was constructed using Molecular Evolutionary Genetics Analysis version 5.05 (megasoftware.net/) with the optimal model (Kimura 2-parameter model). The relative support of internal nodes was determined by bootstrap analyses with 1,000 replications for ML reconstructions (22). The program, codeml within the Phylogenetic Analysis by ML version 4.7 software package (abacus.gene.ucl.ac.uk/software/paml.html) was used to investigate whether WTAP proteins were positively selected (23).

Identification of functionally relevant SNPs in the human WTAP gene and somatic mutations in human cancer

The functionally relevant SNPs of the human WTAP gene were extracted from the Ensembl genome databases and the Short Genetic Variations database (ncbi.nlm.nih.gov/snp), as previously described (1521). The SNPs causing missense mutations were then identified. The somatic mutations of the human WTAP gene in cancer tissues were extracted from the Catalogue of Somatic Mutations in Cancer (COSMIC) database (cancer.sanger.ac.uk/cosmic), which mines somatic mutations in complete cancer genomes (24).

In silico expression analyses of the human WTAP gene

Expression profiles of the human WTAP gene in normal tissues were obtained from the GeneAnnot (genecards.weizmann.ac.il/geneannot/index.shtml) (25) and ArrayExpress (ebi.ac.uk/arrayexpress/) databases (26).

Meta-analysis of the prognostic value of human WTAP gene expression in cancer tissues

The expression of the WTAP gene and the biological association between gene expression and prognosis were determined by inputting human WTAP gene (NP_001257460) into the PrognoScan database (prognoscan.org/) (27).

Results

Comparative proteomics of WTAP proteins identified in vertebrate genomes

WTAP DNA and protein sequences were collected from the Ensembl genome database and confirmed by BLASTing. Completed WTAP genes were identified in the following genomes: Human, chimpanzee, gibbon, macaque, gorilla, orangutan, olive baboon, vervet monkey, marmoset, tarsier, bush baby, armadillo, sloth, squirrel, elephant, guinea pig, mouse, rat, pika, horse, microbat, ferret, dolphin, dog, pig, sheep, cow, alpaca, chicken, duck, turkey, flycatcher, zebra finch, Chinese softshell turtle, anole lizard, spotted gar, Amazon molly, platyfish, stickleback, tilapia, medaka, cave fish and zebrafish. In the armadillo genome, two WTAP genes were identified. The maximum likelihood method was used to construct the phylogenetic tree of vertebrate WTAPs (Fig. 1). The vertebrate WTAP genes clustered into the primate, rodent and teleost lineages. Furthermore, site-specific analysis for positive selection with six models of codon substitution, M0 (one-ratio), M1a (nearly neutral), M2a (positive selection), M3 (discrete), M7 (β), and M8 (β and ω) were performed in vertebrate, mammalian, bird, reptile and teleost lineages. No sites were identified under positive selection with any models in the various WTAP groups. Therefore, it was concluded that WTAP proteins were under purifying selection (data not shown).

Expression profile of the human WTAP gene

Investigation of available microarray data revealed that the human WTAP gene was predominantly expressed in the following tissues: Bone marrow, whole blood, lymph node, brain, cerebellum, retina, spinal cord, heart, smooth muscle, skeletal muscle, small intestine, colon, adipocyte, kidney, liver, lung, pancreas, thyroid, salivary gland, adrenal gland, skin, ovary, uterus, placenta, prostate and testis. In addition, the human WTAP gene was expressed in the following types of cancer: Bladder, blood, brain, breast, colorectal, esophagus, eye, head and neck, lung, ovarian, prostate, skin and soft tissue.

Comparative genomics on the human WTAP gene

Signal transducer and activator of transcription 1 (STAT1), forkhead box protein O1 (FOXO1), interferon regulatory factor 1 (IRF1), glucocorticoid receptor and peroxisome proliferator-activated receptor γ (PPARγ) transcription factor binding sites were identified in the upstream (promoter) region of the WTAP gene.

Functionally relevant SNP identification in the human WTAP gene

A total of 1,347 available SNPs were identified in the human WTAP gene. Among these, 19 SNPs were functionally relevant, causing missense mutations (Table I).

Table I

Functionally relevant SNP identification in the human WTAP gene.

Table I

Functionally relevant SNP identification in the human WTAP gene.

SNPChr 6 position sequenceSequenceTypeAmino acid
rs1543500159,755,455(+) GGCAG(G/T)GAAAAMissenseS (Ser) ⇒ R (Arg)
rs112093927159,753,489(+) GAAGT(A/G)TCGAAMissenseC (Cys) ⇒ Y (Tyr)
rs140439442159,755,519(+) AGAAA(A/G)CAGTGMissenseA (Ala) ⇒ T (Thr)
rs146208471159,755,499(+) CAGTC(A/G)TGACCMissenseH (His) ⇒ R (Arg)
rs144625269159,755,303(+) ACAGG(A/G)AGGGCMissenseE (Glu) ⇒ K (Lys)
rs148080007159,739,015(+) CTTCA(A/G)AGTTAMissenseK (Lys) ⇒ R (Arg)
rs149103382159,755,447(+) CTCCC(A/G)CGGGCMissenseT (Thr) ⇒ A (Ala)
rs150215853159,743,764(+) AGCAA(C/G)CAAGGMissenseT (Thr) ⇒ S (Ser)
rs187127278159,755,555(+) GTTCC(C/T)GCCACMissenseR (Arg) ⇒ C (Cys)
rs373161821159,755,405(+) GTTAC(A/G)TAAATMissenseV (Val) ⇒ I (Ile)
rs375163417159,755,312(+) GCAAC(A/G)CAACCMissenseT (Thr) ⇒ A (Ala)
rs375840138159,755,421(+) CAGTG(C/T)GGGGTMissenseA (Ala) ⇒ V (Val)
rs528166112159,755,353(+) GGTAA(A/T)AAGTCMissenseN (Asn) ⇒ K (Lys)
rs532857576159,748,230(+) CGAGC(A/G)TTGCCMissenseV (Val) ⇒ I (Ile)
rs543667028159,755,190(+) AGCTT(C/T)TGAACMissenseS (Ser) ⇒ F (Phe)
rs546830649159,755,349(+) GAATG(A/G)TAATAMissenseG (Gly) ⇒ D (Asp)
rs558627827159,742,104(+) ATGAA(A/G)CATATMissenseA (Ala) ⇒ T (Thr)
rs560214098159,748,326(+) GCATC(A/G)TCTGCMissenseQ (Gln) ⇒ K (Lys)
rs563264867159,755,400(+) TAGTG(A/G)TTACGMissenseG (Gly) ⇒ D (Asp)

[i] SNP, single nucleotide polymorphism; WTAP, Wilms tumor 1 associated protein; Chr, chromosome.

Identification of somatic mutations of the WTAP gene in human cancer

By searching the COSMIC database, 65 somatic mutations of the human WTAP gene were identified in various types of cancer tissue (Table II).

Table II

Somatic mutations of WTAP in cancer tissues.

Table II

Somatic mutations of WTAP in cancer tissues.

Position (AA)Mutation (CDS)Mutation (amino acid)Mutation ID (COSM)CountMutation type
12c.34C>Tp.R12aCOSM2073783 Substitution-nonsense
30c.90G>Ap.W30aCOSM2231361 Substitution-nonsense
32c.96A>Tp.Q32HCOSM36224551 Substitution-missense
33c.98A>Gp.Y33CCOSM49303741 Substitution-missense
35c.104C>Tp.A35VCOSM44450391 Substitution-missense
44c.132C>Ap.Y44aCOSM4508221 Substitution-nonsense
51c.152A>Cp.D51ACOSM4508231 Substitution-missense
54c.161G>Ap.G54DCOSM38601031 Substitution-missense
66c.198G>Ap.Q66QCOSM10755341Substitution-coding silent
71c.211C>Tp.R71CCOSM38601041 Substitution-missense
79c.235C>Tp.R79aCOSM14419241 Substitution-nonsense
79c.236G>Ap.R79QCOSM4508241 Substitution-missense
86c.256G>Ap.E86KCOSM7413381 Substitution-missense
91c.271A>Gp.T91ACOSM14962561 Substitution-missense
95c.283C>Tp.Q95aCOSM16211612 Substitution-nonsense
103c.307C>Tp.P103SCOSM10755351 Substitution-missense
104c.312C>Tp.S104SCOSM14419251Substitution-coding silent
108c.324G>Cp.L108LCOSM13117781Substitution-coding silent
116c.347C>Tp.A116VCOSM13117791 Substitution-missense
117c.350T>Ap.I117NCOSM5952101 Substitution-missense
119c.355T>Gp.L119VCOSM10755361 Substitution-missense
135c.405G>Tp.L135LCOSM4508251Substitution-coding silent
141c.421G>Ap.E141KCOSM48067621 Substitution-missense
147c.440T>Cp.F147SCOSM10755371 Substitution-missense
148c.443C>Tp.T148MCOSM12328731 Substitution-missense
150c.448G>Ap.D150NCOSM30232841 Substitution-missense
150c.448G>Cp.D150HCOSM48283261 Substitution-missense
154c.461G>Ap.G154ECOSM3419872 Substitution-missense
156c.466A>Gp.K156ECOSM50035721 Substitution-missense
156c.467A>Gp.K156RCOSM4209941 Substitution-missense
157c.470T>Cp.L157SCOSM15454491 Substitution-missense
159c.476C>Ap.A159ECOSM2483611 Substitution-missense
159c.476C>Tp.A159VCOSM30232871 Substitution-missense
162c.485G>Ap.R162QCOSM14419261 Substitution-missense
162c.485G>Cp.R162PCOSM14874251 Substitution-missense
163c.489G>Ap.M163ICOSM4836321 Substitution-missense
163 c.489_494delGCTTAT p.M163_L164delMLCOSM2422711Deletion-in frame
164c.491T>Cp.L164PCOSM13117801 Substitution-missense
209c.627C>Ap.I209ICOSM33663701Substitution-coding silent
211c.632T>Gp.L211RCOSM48803071 Substitution-missense
212c.635A>Cp.D212ACOSM5952091 Substitution-missense
226c.676C>Tp.Q226aCOSM47414941 Substitution-nonsense
231c.693G>Ap.E231ECOSM2073801Substitution-coding silent
248c.744C>Tp.A248ACOSM12705421Substitution-coding silent
250c.749G>Cp.S250TCOSM3953211 Substitution-missense
255c.763A>Gp.T255ACOSM46455581 Substitution-missense
255c.765A>Tp.T255TCOSM7413371Substitution-coding silent
271c.811A>Tp.S271CCOSM30232942 Substitution-missense
305c.913T>Cp.S305PCOSM36224561 Substitution-missense
308c.922G>Tp.G308WCOSM14419271 Substitution-missense
309c.927T>Gp.N309KCOSM41603421 Substitution-missense
314c.941C>Gp.S314CCOSM16451281 Substitution-missense
334c.1001C>Tp.A334VCOSM39282042 Substitution-missense
335c.1003G>Tp.G335WCOSM5952081 Substitution-missense
342 c.1020_1021delCTp.P342fsa4COSM37324021 Deletion-frameshift
343c.1029G>Ap.T343TCOSM47414951Substitution-coding silent
353c.1058C>Gp.S353aCOSM15454481 Substitution-nonsense
367c.1099G>Tp.A367SCOSM10755381 Substitution-missense
368c.1104G>Cp.V368VCOSM48328021Substitution-coding silent
374c.1120C>Tp.R374aCOSM47414961 Substitution-nonsense
374c.1121G>Ap.R374QCOSM10755391 Substitution-missense
377c.1130G>Cp.G377ACOSM49806941 Substitution-missense
384c.1150G>Ap.G384SCOSM46379941 Substitution-missense
391c.1173A>Gp.V391VCOSM30233071Substitution-coding silent
393c.1178G>Tp.G393VCOSM5952071 Substitution-missense

{ label (or @symbol) needed for fn[@id='tfn2-mmr-14-03-2823'] } Mutation (amino acid) refers to the change in amino acid sequence resulting from the mutation.

a Stop codon. Count refers to the mutation number identified in the database. WTAP, wilms tumor 1 associated protein; AA, amino acid; CDS, coding DNA sequence; COSM, catalogue of somatic mutations.

Meta-analysis of the prognostic value of human WTAP gene expression in cancer tissues

A total of 17 out of 328 microarrays identified an association between WTAP gene expressions and cancer prognosis (bladder cancers, 1/7; blood cancers, 0/37; brain cancers, 1/23; breast cancers, 5/110; colorectal cancers, 3/48; esophagus cancers, 0/1; eye cancers, 2/5; head and neck cancers, 0/6; lung cancers, 4/56; ovarian cancers, 0/25; prostate cancers, 0/1; skin cancers, 0/6; and soft tissue cancers, 1/3), P<0.05 (Table III) (2838). In bladder, brain, eye and soft tissue cancers, reduced expression of the WTAP gene was associated with poor survival. However, an increased expression of the WTAP gene was associated with poor survival in lung cancer. Of the six breast cancer microarrays, reduced expression of the WTAP gene was associated with poor survival in two cases from the same database (GSE2990) and in the database GSE1456-GPL96, while increased expression of the WTAP gene was associated with poor survival in the GSE1456-GPL96 and GSE1456-GPL97 databases. Of the colorectal cancer microarrays, reduced expression of the WTAP gene was associated with poor survival in two cases (GSE17537 and GSE17538), while increased expression of the WTAP gene was associated with poor survival in the GSE14333 database.

Table III

Associations between microarray WTAP expression and cancer prognosis.

Table III

Associations between microarray WTAP expression and cancer prognosis.

Author, yearDatabaseCancer typeSubtypePatient numberEndpointCutpointP-valuePrognosisRef.
Kim, 2010GSE5287BladderTransitional cell carcinoma165Disease specific survival0.110.025526128
Freije, 2004GSE4412-GPL97BrainGlioma74Overall survival0.140.000331129
Pawitan, 2005GSE1456-GPL96Breast159Overall survival0.430.003810130
Pawitan, 2005GSE1456-GPL96Breast159Relapse free survival0.780.023849230
Pawitan, 2005GSE1456-GPL97Breast159Relapse free survival0.860.047352230
Sotiriou, 2006GSE2990Breast54Distant metastasis free survival0.150.039634131
Sotiriou, 2006GSE2990Breast62Relapse free survival0.130.038288131
Jorissen, 2009GSE14333Colorectal226Disease free survival0.850.011858232
Smith, 2010GSE17537Colorectal49Disease specific survival0.240.010751133
Smith, 2010GSE17538Colorectal55Overall survival0.250.010567133
Laurent, 2011GSE22138EyeUveal melanoma63Distant metastasis free survival0.490.049578134
Laurent, 2011GSE22138EyeUveal melanoma63Distant metastasis free survival0.210.007475134
Tomida, 2009GSE13213LungAdenocarcinoma117Overall survival0.890.000567235
Tomida, 2009GSE13213LungAdenocarcinoma117Overall survival0.900.012852235
Okayama, 2012GSE31210LungAdenocarcinoma204Overall survival0.880.043703236
Bild, 2006GSE3141LungNSCLC111Overall survival0.890.002284237
Gobble, 2011GSE30929Soft tissueLiposarcoma140Distant recurrence free survival0.210.008282138

[i] The PrognoScan database was used to determine associations between expression of the WTAP gene and prognosis. P<0.05 was considered to indicate a statistically significant difference. WTAP, Wilms tumor 1 associated protein; NSCLC, non-small cell lung cancer.

Discussion

WT1 was first identified due to its essential role in the normal development of the human genitourinary system (1) and WTAP was identified as a protein that interacted with WT1 (3). A total of 44 complete WTAP genes were identified in the human, chimpanzee, gibbon, macaque, gorilla, orangutan, olive baboon, vervet monkey, marmoset, tarsier, bush baby, armadillo, sloth, squirrel, elephant, guinea pig, mouse, rat, pika, horse, microbat, ferret, dolphin, dog, pig, sheep, cow, alpaca, chicken, duck, turkey, flycatcher, zebra finch, Chinese softshell turtle, anole lizard, spotted gar, Amazon molly, platyfish, stickleback, tilapia, medaka, cave fish and zebrafish genomes. It was observed that WTAP genes were widely expressed in vertebrates, existing in fish, amphibians, birds and mammals. The phylogenetic tree revealed that the vertebrate WTAP proteins were clustered into the primate, rodent and teleost lineages. All vertebrate WTAPs are conserved according to the analysis of alignment and phylogenetic tree construction. Furthermore, the vertebrate WTAPs were under purifying selection. These results suggest that WTAP performs an essential physiological role in all vertebrates.

WTAP was predominantly expressed in bone marrow, whole blood, lymph node, brain, cerebellum, retina, spinal cord, heart, smooth muscle, skeletal muscle, small intestine, colon, adipocyte, kidney, liver, lung, pancreas, thyroid, salivary gland, adrenal gland, skin, ovary, uterus, placenta, prostate and testis. The expression pattern of WTAP appeared to be ubiquitous, which is indicative of a housekeeping role. By contrast, WT1 is expressed at low levels in only the spleen, heart, gonad and kidney (3). A total of 19 SNPs that cause missense mutations were identified in the human WTAP gene; however, it remains unclear whether these SNPs affect the physiological or pathological functions of WTAP.

WTAP and WT1 partially co-localize with splicing factors, and are distributed together in the nucleoplasm and in nuclear speckles (3). It has been demonstrated in numerous tumors that WT1 is a tumor suppressor, exerting effects including inhibiting cell proliferation and enhancing apoptosis (3942). However, WTAP is an oncogene, which is overexpressed in cholangiocarcinoma (11), glioblastoma (12) and acute myeloid leukemia (13). In the present study, it was demonstrated that WTAP was expressed in bladder, blood, brain, breast, colorectal, esophagus, eye, head and neck, lung, ovarian, prostate, skin and soft tissue cancers. Of a total of 328 microarrays, 17 revealed an association between microarray WTAP expression and cancer prognosis (bladder cancers, 1; brain cancers, 1; breast cancers, 6; colorectal cancers, 3; eye cancers, 2; lung cancers, 4; and soft tissue cancers, 1). The majority of microarrays did not reveal an association between microarray WTAP expression and cancer prognosis. This may be due to a lack of WTAP expression information in the database. Notably, WTAP was not involved in all tumor types. In addition, it is notable that the association between WTAP expression and prognosis varied between the different cancer types, and even in identical cancers from separate databases. This suggests that the function of WTAP in these tumors may not be solely as an oncogene, but may be multidimensional (1113). The differing WTAP expression in various tumors may be due to the distinct oncogenes or tumor suppressors stabilized by WTAP in particular tumors (9,10).

Furthermore, 65 somatic mutations of WTAP were identified in cancer tissues. The effects of these mutations on tumor formation remain to be elucidated and require future investigation. The results of the present study suggest that WTAP has a comprehensive and complex role in tumor formation. STAT1, FOXO1, IRF1, glucocorticoid receptor and PPARγ transcription factor binding sites were identified in the upstream (promoter) region of the WTAP gene. STAT1 is a cytoplasmic protein, which functions as a signal messenger and transcription factor in cellular responses to cytokines and growth factors (43). It exhibits anti-tumor functions via control of the immune system and promotion of tumor immune surveillance (4446). FOXO1 is an important transcriptional regulator of cell proliferation and is considered to be essential for tumor growth and progression (47). Deregulation of FOXO1 promotes cell proliferation and tumorigenesis, and has thus become a primary target of tumorigenesis prevention (48,49). IRF1 is involved in the regulation of interferon α and β transcription, and it has been demonstrated that IRF1 gene deletion or rearrangement correlates with the development of human cancers (50,51). The glucocorticoid receptor is a member of the nuclear receptor family, which acts as a ligand-dependent transcription factor to regulate gene expression. In addition, the estrogen and androgen receptors are members of the nuclear receptor family. In breast cancer, the estrogen receptor drives cell growth, proliferation and metastasis, and the androgen receptor has a similar role in prostate cancer (52,53). These tumor-associated transcriptional factors may affect the expression of WTAP and contribute to tumor formation (1214).

In conclusion, 44 complete WTAP genes were identified in vertebrate genomes. The vertebrate WTAP proteins clustered into the primate, rodent and teleost lineages. The association between WTAP gene expression and prognosis varied in distinct cancers, and even in identical cancers from separate microarray databases. Furthermore, a total of 65 somatic mutations were identified in the human WTAP gene from cancer tissue samples. The results of the present study suggest that the function of WTAP in tumor formation may be multidimensional.

Acknowledgments

The present study was supported by Anhui Science and Technology Research Projects (grant no. 130zc04065), the National Natural Science Foundation of China (grant nos. 81372828 and 810001329), and the Priority Academic Program Development of Jiangsu Higher Education Institutions.

References

1 

Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, Housman D and Jaenisch R: WT-1 is required for early kidney development. Cell. 74:679–691. 1993. View Article : Google Scholar : PubMed/NCBI

2 

Kim MK, McGarry TJ, O Broin P, Flatow JM, Golden AA and Licht JD: An integrated genome screen identifies the Wnt signaling pathway as a major target of WT1. Proc Natl Acad Sci USA. 106:11154–11159. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Little NA, Hastie ND and Davies RC: Identification of WTAP, a novel Wilms' tumour 1-associating protein. Hum Mol Genet. 9:2231–2239. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Granadino B, Penalva LO and Sánchez L: The gene fl(2)d is needed for the sex-specific splicing of transformer pre-mRNA but not for double-sex pre-mRNA in Drosophila melanogaster. Mol Gen Genet. 253:26–31. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Penalva LO, Ruiz MF, Ortega A, Granadino B, Vicente L, Segarra C, Valcárcel J and Sánchez L: The drosophila fl(2) d gene, required for female-specific splicing of Sxl and tra pre-mRNAs, encodes a novel nuclear protein with a HQ-rich domain. Genetics. 155:129–139. 2000.PubMed/NCBI

6 

Horiuchi K, Umetani M, Minami T, Okayama H, Takada S, Yamamoto M, Aburatani H, Reid PC, Housman DE, Hamakubo T and Kodama T: Wilms' tumor 1-associating protein regulates G2/M transition through stabilization of cyclin A2 mRNA. Proc Natl Acad Sci USA. 103:17278–17283. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Naruse C, Fukusumi Y, Kakiuchi D and Asano M: A novel gene trapping for identifying genes expressed under the control of specific transcription factors. Biochem Biophys Res Commun. 361:109–115. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Horiuchi K, Kawamura T, Iwanari H, Ohashi R, Naito M, Kodama T and Hamakubo T: Identification of Wilms' tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J Biol Chem. 288:33292–33302. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Schwartz S, Mumbach MR, Jovanovic M, Wang T, Maciag K, Bushkin GG, Mertins P, Ter-Ovanesyan D, Habib N, Cacchiarelli D, et al: Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5′ sites. Cell Rep. 8:284–296. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al: Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24:177–189. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Jo HJ, Shim HE, Han ME, Kim HJ, Kim KS, Baek S, Choi KU, Hur GY and Oh SO: WTAP regulates migration and invasion of cholangiocarcinoma cells. J Gastroenterol. 48:1271–1282. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Jin DI, Lee SW, Han ME, Kim HJ, Seo SA, Hur GY, Jung S, Kim BS and Oh SO: Expression and roles of Wilms' tumor 1-associating protein in glioblastoma. Cancer Sci. 103:2102–2109. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Bansal H, Yihua Q, Iyer SP, Ganapathy S, Proia DA, Penalva LO, Uren PJ, Suresh U, Carew JS, Karnad AB, et al: WTAP is a novel oncogenic protein in acute myeloid leukemia. Leukemia. 28:1171–1174. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Zhang J, Tsoi H, Li X, Wang H, Gao J, Wang K, Go MY, Ng SC, Chan FK, Sung JJ and Yu J: Carbonic anhydrase IV inhibits colon cancer development by inhibiting the Wnt signalling pathway through targeting the WTAP-WT1-TBL1 axis. Gut. pii: gutjnl-2014-308614. 2015.Epub ahead of print.

15 

Ding Z, Yang HW, Xia TS, Wang B and Ding Q: Integrative genomic analyses of the RNA-binding protein, RNPC1, and its potential role in cancer prediction. Int J Mol Med. 36:473–484. 2015.PubMed/NCBI

16 

Wang B, Xu W, Tan M, Xiao Y, Yang H and Xia TS: Integrative genomic analyses of a novel cytokine, interleukin-34 and its potential role in cancer prediction. Int J Mol Med. 35:92–102. 2015.

17 

Yang L, Luo Y and Wei J: Integrative genomic analyses on Ikaros and its expression related to solid cancer prognosis. Oncol Rep. 24:571–577. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Yang L, Luo Y, Wei J and He S: Integrative genomic analyses on IL28RA, the common receptor of interferon-lambda1, -lambda2 and -lambda3. Int J Mol Med. 25:807–812. 2010.PubMed/NCBI

19 

Yang L, Wei J and He S: Integrative genomic analyses on interferon-lambdas and their roles in cancer prediction. Int J Mol Med. 25:299–304. 2010.PubMed/NCBI

20 

Wang M, Wei X, Shi L, Chen B, Zhao G and Yang H: Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction. Int J Mol Med. 33:1019–1026. 2014.PubMed/NCBI

21 

Wang B, Chen K, Xu W, Chen D, Tang W and Xia TS: Integrative genomic analyses of secreted protein acidic and rich in cysteine and its role in cancer prediction. Mol Med Rep. 10:1461–1468. 2014.PubMed/NCBI

22 

Kumar S, Nei M, Dudley J and Tamura K: MEGA: A biologist-centric software for evolutionary analysis of DNA and protein sequences. Brief Bioinform. 9:299–306. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Yang Z: PAML: A program package for phylogenetic analysis by maximum likelihood. Comput Appl Biosci. 13:555–556. 1997.PubMed/NCBI

24 

Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al: COSMIC: Mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res. 39(Database issue): D945–D950. 2011. View Article : Google Scholar :

25 

Chalifa-Caspi V, Yanai I, Ophir R, Rosen N, Shmoish M, Benjamin-Rodrig H, Shklar M, Stein TI, Shmueli O, Safran M and Lancet D: GeneAnnot: Comprehensive two-way linking between oligonucleotide array probesets and GeneCards genes. Bioinformatics. 20:1457–1458. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Parkinson H, Sarkans U, Shojatalab M, Abeygunawardena N, Contrino S, Coulson R, Farne A, Lara GG, Holloway E, Kapushesky M, et al: ArrayExpress-a public repository for microarray gene expression data at the EBI. Nucleic Acids Res. 33(Database issue): D553–D555. 2005. View Article : Google Scholar

27 

Mizuno H, Kitada K, Nakai K and Sarai A: PrognoScan: A new database for meta-analysis of the prognostic value of genes. BMC Med Genomics. 2:182009. View Article : Google Scholar : PubMed/NCBI

28 

Kim WJ, Kim EJ, Kim SK, Kim YJ, Ha YS, Jeong P, Kim MJ, Yun SJ, Lee KM, Moon SK, et al: Predictive value of progression-related gene classifier in primary non-muscle invasive bladder cancer. Mol Cancer. 9:32010. View Article : Google Scholar : PubMed/NCBI

29 

Freije WA, Castro-Vargas FE, Fang Z, Horvath S, Cloughesy T, Liau LM, Mischel PS and Nelson SF: Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 64:6503–6510. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Pawitan Y, Bjöhle J, Amler L, Borg AL, Egyhazi S, Hall P, Han X, Holmberg L, Huang F, Klaar S, et al: Gene expression profiling spares early breast cancer patients from adjuvant therapy: Derived and validated in two population-based cohorts. Breast Cancer Res. 7:R953–R964. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, et al: Gene expression profiling in breast cancer: Understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 98:262–272. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Jorissen RN, Gibbs P, Christie M, Prakash S, Lipton L, Desai J, Kerr D, Aaltonen LA, Arango D, Kruhøffer M, et al: Metastasis-associated gene expression changes: Predict poor outcomes in patients with dukes stage B and C colorectal cancer. Clin Cancer Res. 15:7642–7651. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Smith JJ, Deane NG, Wu F, Merchant NB, Zhang B, Jiang A, Lu P, Johnson JC, Schmidt C, Bailey CE, et al: Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer. Gastroenterology. 138:958–968. 2010. View Article : Google Scholar

34 

Laurent C, Valet F, Planque N, Silveri L, Maacha S, Anezo O, Hupe P, Plancher C, Reyes C, Albaud B, et al: High PTP4A3 phosphatase expression correlates with metastatic risk in uveal melanoma patients. Cancer Res. 71:666–674. 2011. View Article : Google Scholar

35 

Tomida S, Takeuchi T, Shimada Y, Arima C, Matsuo K, Mitsudomi T, Yatabe Y and Takahashi T: Relapse-related molecular signature in lung adenocarcinomas identifies patients with dismal prognosis. J Clin Oncol. 27:2793–2799. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar

37 

Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, Joshi MB, Harpole D, Lancaster JM, Berchuck A, et al: Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 439:353–357. 2006. View Article : Google Scholar

38 

Gobble RM, Qin LX, Brill ER, Angeles CV, Ugras S, O'Connor RB, Moraco NH, Decarolis PL, Antonescu C and Singer S: Expression profiling of liposarcoma yields a multigene predictor of patient outcome and identifies genes that contribute to liposarcomagenesis. Cancer Res. 71:2697–2705. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Li Y, Wang J, Li X, Jia Y, Huai L, He K, Yu P, Wang M, Xing H, Rao Q, et al: Role of the Wilms' tumor 1 gene in the aberrant biological behavior of leukemic cells and the related mechanisms. Oncol Rep. 32:2680–2686. 2014.PubMed/NCBI

40 

Lee SY, Choe YJ, Park JY, Lee SS, Kim YH, Shin SJ, Chung YJ and Kim HS: Wilms' tumor gene 1 enhances nutlin-3-induced apoptosis. Oncol Rep. 31:131–136. 2014.

41 

Liu Y and Liu S: Berberine inhibits Wilms' tumor cell progression through upregulation of Wilms' tumor gene on the X chromosome. Mol Med Rep. 8:1537–1541. 2013.PubMed/NCBI

42 

Li M, Cai MY, Lu JB, Hou JH, Wu QL and Luo RZ: Clinicopathological investigation of four cases of desmoplastic small round cell tumor. Oncol Lett. 4:423–428. 2012.

43 

Ding Y, Yang M, She S, Min H, Xv X, Ran X, Wu Y, Wang W, Wang L, Yi L, et al: iTRAQ-based quantitative proteomic analysis of cervical cancer. Int J Oncol. 46:1748–1758. 2015.PubMed/NCBI

44 

Kaewpiboon C, Srisuttee R, Malilas W, Moon J, Oh S, Jeong HG, Johnston RN, Assavalapsakul W and Chung YH: Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells. Mol Med Rep. 11:2315–2321. 2015.

45 

Chen S, Li C, Wu B, Zhang C, Liu C, Lin X, Wu X, Sun L, Liu C, Chen B, et al: Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors. Int J Oncol. 45:1133–1142. 2014.PubMed/NCBI

46 

Li W, Wei Q and Liang J: Phosphorylated signal transducer and activator of transcription 1 is a potential predictor of interferon response in patients with advanced renal cell carcinoma. Mol Med Rep. 9:1929–1934. 2014.PubMed/NCBI

47 

Zhao Z, Li C, Xi H, Gao Y and Xu D: Curcumin induces apoptosis in pancreatic cancer cells through the induction of forkhead box O1 and inhibition of the PI3K/Akt pathway. Mol Med Rep. 12:5415–5422. 2015.PubMed/NCBI

48 

Gao F and Wang W: MicroRNA-96 promotes the proliferation of colorectal cancer cells and targets tumor protein p53 inducible nuclear protein 1, forkhead box protein O1 (FOXO1) and FOXO3a. Mol Med Rep. 11:1200–1266. 2015.

49 

Zhu H: Targeting forkhead box transcription factors FOXM1 and FOXO in leukemia (Review). Oncol Rep. 32:1327–1334. 2014.PubMed/NCBI

50 

Xu Y, Wang W, Gou A, Li H, Tian Y, Yao M and Yang R: Effects of suppressor of cytokine signaling 1 silencing on human melanoma cell proliferation and interferon-γ sensitivity. Mol Med Rep. 11:583–588. 2015.

51 

Chen FF, Jiang G, Xu K and Zheng JN: Function and mechanism by which interferonregulatory factor-1 inhibits oncogenesis. Oncol Lett. 5:417–423. 2013.PubMed/NCBI

52 

Pufall MA: Glucocorticoids and Cancer. Adv Exp Med Biol. 872:315–333. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Volden PA and Conzen SD: The influence of glucocorticoid signaling on tumor progression. Brain Behav Immun. 30(Suppl): S26–S31. 2013. View Article : Google Scholar

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu LS, Qian JY, Wang M and Yang H: Identifying the role of Wilms tumor 1 associated protein in cancer prediction using integrative genomic analyses. Mol Med Rep 14: 2823-2831, 2016
APA
Wu, L., Qian, J., Wang, M., & Yang, H. (2016). Identifying the role of Wilms tumor 1 associated protein in cancer prediction using integrative genomic analyses. Molecular Medicine Reports, 14, 2823-2831. https://doi.org/10.3892/mmr.2016.5528
MLA
Wu, L., Qian, J., Wang, M., Yang, H."Identifying the role of Wilms tumor 1 associated protein in cancer prediction using integrative genomic analyses". Molecular Medicine Reports 14.3 (2016): 2823-2831.
Chicago
Wu, L., Qian, J., Wang, M., Yang, H."Identifying the role of Wilms tumor 1 associated protein in cancer prediction using integrative genomic analyses". Molecular Medicine Reports 14, no. 3 (2016): 2823-2831. https://doi.org/10.3892/mmr.2016.5528