Proteomic identification of potential cancer markers in human urine using subtractive analysis

  • Authors:
    • Holger Husi
    • Richard J.E. Skipworth
    • Andrew Cronshaw
    • Kenneth C.H. Fearon
    • James A. Ross
  • View Affiliations

  • Published online on: March 7, 2016     https://doi.org/10.3892/ijo.2016.3424
  • Pages: 1921-1932
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Urine is an ideal medium in which to focus diagnostic cancer research due to the non-invasive nature and ease of sampling. Many large-scale proteomic studies have shown that urine is unexpectedly complex. We hypothesised that novel diagnostic cancer biomarkers could be discovered using a comparative proteomic analysis of pre-existing data. We assembled a database of 100 published datasets of 5,620 urinary proteins, as well as 46 datasets of 8,620 non-redundant proteins derived from kidney and blood proteome analyses. The data were then used to either subtract or compare molecules from a novel urinary proteome profiling dataset that we generated. We identified 1,161 unique proteins in samples from either cancer-bearing or healthy subjects. Subtractive analysis yielded a subset of 44 proteins that were found uniquely in urine from cancer patients, 30 of which were linked previously to cancer. In conclusion, this approach is useful in discovering novel biomarkers in tissues where unrelated profiling data is available. Only a limited disease-specific novel dataset is required to define new targets or substantiate previous findings. We have shared this discovery platform in the form of our Large Scale Screening Resource database, accessible through the Proteomic Analysis DataBase portal (www.PADB.org).

Introduction

Screening of human tissues for cancer biomarkers is an important task in cancer diagnosis and treatment, which is hindered by the complexity of the sample systems studied. A less complex system such as urine is a preferred medium to screen for protein or peptide biomarkers due to the non-invasive sampling of patients, ease of sampling and the unrestricted quantities obtainable. Urine is relatively stable in terms of protein/peptide composition and fragmentation compared with other bodily fluids such as serum, where proteolytic degradation by endogenous proteases has been shown to occur during or after sample collection (1).

Several investigations have been published describing the urinary peptidome and proteome (as well as biomarker discoveries for several diseases) using methodologies ranging from traditional 2D gel electrophoresis alone (2), or coupled with mass spectrometry (2-DE-MS) (3), immunohistochemistry (4), liquid chromatography mass spectrometry (LC-MS) (5), and surface enhanced laser desorption ionisation-time of flight mass spectrometry (SELDI-TOF-MS) (69).

The proteomic screening of urine for potential cancer markers has shown several proteins to be differentially present in ovarian cancer (10). Bladder cancer biomarkers constitute a different non-overlapping set of molecules (1113), as do potential biomarkers for upper gastrointestinal cancers (9). An improvement in the reliability of diagnostic tests is to employ more than one biomarker synchronously (9,14). For example, one previous study employed an antibody-based array of 810 different antibodies to define peptide patterns in urine associated with cancer (15). A different approach was used successfully in recent years, combining urinary mass spectroscopy with protein/peptide pattern analysis to identify kidney disease (16).

There is a clear need to collect and cross-correlate the wealth of data published in the scientific literature. Currently, there are a number of urinary databases available. The majority consist of lists of identified proteins derived from tryptic digests analysed by liquid chromatography tandem mass spectrometry (LC-MS/MS), such as the Max-Planck Unified Proteome Database (MAPU) (17) and Sys-BodyFluid (18). More recently, a urinary database combining chromatographic reverse-phase retention times and m/z values has been established (19).

However, there is no database available which integrates all of the data. In order to fill this gap, we have assembled datasets from 100 urinary proteomic studies in our novel proteomic database termed the Large Scale Screening Resource (LSSR). LSSR is accessible and downloadable through the Proteomic Analysis DataBase (PADB) portal at www.PADB.org.

In this study, we explore the possibility of discovering novel cancer-associated molecular markers in human urine by subtractive analysis using a novel dataset of the human cancer urinary proteome [derived from patients with upper gastrointestinal (GI) cancer] and comparing it to non-cancer urinary datasets.

Materials and methods

Materials

Tris/Tricine peptide gels, gel-running buffers, CM and IMAC resins, and chromatography buffers were from Bio-Rad (Hemel Hempstead, UK). All other chemicals were obtained from Sigma-Aldrich (Gillingham, UK).

Sample collection

Urine samples were obtained from upper GI cancer patients (n=41) and non-cancer controls (n=21) as described previously (9). Summary participant demographics are shown in Table I. Participant age ranged between 21 and 84 (control group), and 43 and 82 (cancer group). Random morning urine samples were collected over a time period of 2 years. Cancer urine samples were collected prior to surgery if the patient was being considered for resection. All procedures were approved by the local research ethics committee, and written informed consent was obtained. The study conformed to the standards set by the Declaration of Helsinki. All urine samples were kept at −40°C for short-term or −80°C for long-term storage.

Table I

Demographics of the study cohort.

Table I

Demographics of the study cohort.

Cancer (n=41)Control (n=21)Entire cohort (n=62)
Age (years)64 (9.5)62.1 (23.5)63.4 (15.6)
Male (M:F)26:1517:0443:19
Primary tumor origin
Pancreas15N/A
Oesophagus9
OGJ7
Stomach5
Duodenum1
Unknown4
Histology
Adenocarcinoma34N/A
Squamous carcinoma3
Unknown4

[i] Urine specimens were analysed from cancer patients (n=41) and healthy controls (n=21). Data are presented as means with standard deviations in brackets. OGJ, oesophago-gastric junction.

Chromatographic enrichment of urine proteins and peptides, and sample preparation

Aliquots of 0.5 ml from individual cancer or control urine samples was added to either 30 μl CM10 (n=33 cancer urines, n=8 control urines) or 30 μl IMAC30 (Cu2+-chelated) (n=21 cancer urines, n=19 control urines) spin column resin (Bio-Rad) and 0.75 ml binding buffer (either 0.1 M NaH3C2O2 pH 4.0 for CM resin, or 0.1 M NaHPO4 pH 7.0 including 0.5 M NaCl for IMAC30 resin) and incubated for 1 h at room temperature under constant agitation. Sample and resin combinations were chosen based on independent analyses using peak stratification by SELDI mass spectrometry (9). Unbound material was removed and the resin washed four times with 0.3 ml binding buffer. Bound material was separated by electrophoresis on a 16.5% Tris-Tricine gel (Bio-Rad), and gel bands in the region of 2–10 kDa were excised after Coomassie staining (BioSafe Coomassie; Bio-Rad). The molecular mass range of 2–10 kDa was selected since many urinary proteins are derived from proteolytic processing and urinary shedding as described (20). Additionally, we previously observed potential urinary cancer markers in this mass range (9).

LC-MS/MS mass spectrometry

Proteins and peptides from gel bands were digested in situ with trypsin. The resulting peptides were eluted with acetonitrile (ACN), and analysed by LC-MS/ MS (21). The LC-MS system consisted of an Agilent 1200 Series HPLC (Agilent Technologies, Yarnton, UK) with a Kasil sealed fused silica pre-column (Next Advance, New York, NY, USA) packed to a length of ~3 cm with Pursuit C18, 5 μm particle size (Varian, Crawley, UK) and PicoTip Emitter analytical column PF 360-75-15-N-5 (New Objective, Woburn, MA, USA) packed to a length of ~20 cm with Pursuit C18, 5 μm particle size (Varian). The column was equilibrated with solvent A (0.1% formic acid in 2.5% acetonitrile) and eluted with a linear gradient from 0 to 10% over 6 to 8 min; from 8 to 60% over 8 to 35 min; from 60 to 100% over 35 to 40 min; solvent B (0.1% formic acid, 0.025% TFA in 90% acetonitrile) over 45 min at a flow rate of 5 μl/min. The LTQ mass spectrometer (Thermo Scientific, Epsom, UK) was fitted with a NanoLC ESI source. Data-dependent acquisition was controlled by XCalibur software. Fragmentation spectra were then processed by XCalibur and BioWorks software (Thermo Fisher Scientific, Loughborough, UK) and submitted to the Mascot search engine (Matrix Science, London, UK) using UniProt/SwissProt (release May 2011, Homo sapiens, 18055 sequences) as the reference database. Mascot search parameters were: enzyme specificity trypsin, maximum missed cleavage 1, fixed modifications cysteine carbamidomethylation, variable modification methionine oxidation, precursor mass tolerance +/−3 kDa, fragment ion mass tolerance +/−0.4 kDa. Only Mascot hits with a false discovery rate (FDR) ≤0.05 were taken into consideration.

Meta-analysis and subtractive data analysis

Proteins with at least two peptide matches were analysed further by comparing molecules that were only observed in urine samples from cancer patients with a database consisting of proteins found by other studies in urine, blood and kidney. This database was assembled from 136 publications, listing 146 tissue-specific datasets. The blood datasets covered plasma, serum and erythrocytes; the kidney studies were derived from analyses of cortex, medulla, epithelium, glomerulus, inner medullary collecting duct, mesangium, parenchyma, peroxisomal membrane, peroxisome, basolateral membrane vesicles, brush border membrane vesicles, urothelial mucosa and whole kidney; and urine datasets described either the whole or exosomal proteomes. All entries were then matched to the UniProt database, followed by clustering to individual (unique) entries by annotating splice and variant entries to common parent molecules and ultimately assigning each unique cluster an in-house specific accession number. Additionally, all proteins mapping to immunoglobulins were clustered into one generic cluster, as well as all proteins belonging to the Major Histocompatibility Complex (MHC). Merging and subtraction analysis was done using software written in-house. We also manually added our own functional classification tags to each molecular cluster, based on known properties of each molecule, giving an abridged view of proteome compositions.

Results

Urine samples were extracted from 21 healthy non-cancer controls and 41 patients with upper GI cancer (n=41) (Table I). Of the 41 cancer patients, staging investigations demonstrated that at least 29 (70.7%) had nodal or metastatic disease. We analysed all 62 urine samples by LC-MS/MS in the region of 2–10 kDa by chromatographic enrichment using either CM10, IMAC30, or both resin types individually, resulting in a total of 81 chromatographic enrichments, followed by gel analysis, tryptic digestion and mass spectrometry. All molecular weight regions cut from gels were identical in at least three samples from each cohort group, thus also allowing comparison of identified molecules on a gel-region by gel-region basis After data extraction by Mascot searching (resulting in 35,801 peptides covering 7,639 proteins) and applying discovery criteria of a FDR ≤0.05 and a minimal Mascot score of 13, the resulting 81 datasets were further analysed by merging all protein lists. This yielded 1,228 unique non-redundant entries (data not shown). Additionally, all molecules relating to either immunoglobulins or MHC were also merged into two individual clusters since members of these two families are well known to show a great degree of hypervariability, and therefore they may skew any analysis towards single entries from those classes, since they are not expected to show any duplications across the datasets analysed in this study. The final list consisted of 1,161 molecular clusters. Furthermore, we re-classified all molecules in the datasets available by manually annotating every protein with a single molecular property or functionality tag as listed in the legend of Fig. 1. The properties or functionalities were assigned based on known properties of each individual protein, either from original publications or derived from database annotations, such as enzyme nomenclatures, sequence homologies and domain analysis. The compositional analysis of the merged datasets of blood, urine and kidney proteomes, as well as our urinary dataset is shown in Fig. 2. It was clear that all merged datasets consist of ~25% enzymes, 10% cell-shape molecules, 10% transcriptional or translational elements and 10% transport molecules. However, our novel dataset appeared to contain more cell-shape and transcriptional/translational proteins and less transport molecules, which may reflect an association with disease, rather than a general breakdown of cellular components.

The 1,161 molecules were then split into groups depending on whether they were observed in cancer urine samples, or urine from healthy individuals (Fig. 3A). The 745 proteins only found in cancer urine samples were then tagged and the entire dataset compared to data of 31,743 unmerged entries derived from 146 tissue-specific datasets from 137 publications (data not shown). This external data consisted of 9,707 merged entries, covering proteomic studies from urine, kidney and blood (Table II). A comparative analysis of our dataset with the three largest urinary proteome profiling datasets showed a 46% overlap of our data with the dataset from Kentsis et al (22), a 41% overlap with the study by Adachi et al (23), and a 21% with the urinary exosome dataset from Gonzales et al (24) (Fig. 3B). A global comparison between proteomes from urine, kidney and blood (Fig. 3C) demonstrated a slightly larger overlap of the urinary proteome with the kidney proteome than the blood proteome.

Table II

Number of entries listed in the LSSR database for analysed samples derived from blood, urine and kidney.

Table II

Number of entries listed in the LSSR database for analysed samples derived from blood, urine and kidney.

No. of entries prior to mergeMerged entriesNo. of studies
Urine13,6355,868101
Blood4,4333,66012
Kidney13,6754,96434

[i] The number of entries by tissue type is given either as numbers derived directly from the studies analysed, or after merging all datasets based on unique identifiers assigned by our database.

We then performed subtractive analysis on our urinary proteome data by eliminating any potential cancer candidate molecule if it was found in any of the urinary datasets unrelated to cancer. This reduced dataset of 268 proteins (data not shown) was further condensed by removing any entries which did not have a spectral count of at least two, resulting in 44 proteins, of which 24 were found uniquely in our study (in comparison to all other datasets), and 20 which were also found in the other tissues (Table III). All 44 of these proteins were then analysed by searching the Online Mendelian Inheritance in Man (OMIM) database for publications where these molecules were reported to be directly associated with human disease or cancer. Fourteen proteins were annotated in OMIM to be causative for a disease, and 30 were known to be involved in cancer.

Table III

List of potential cancer candidate markers from human urine.

Table III

List of potential cancer candidate markers from human urine.

Peptide countSpectral countGeneProteinOMIM diseasePADB classificationTissueMolecular functionCancer typePubMed (cancer association)
Only detected in cancer patient urine, high confidence dataset
1011POTEKPPutative β-actin-like protein 3CS: Cell shapeUrineActin filament de-/re-polymerizationHepatocellular carcinoma16824795
1943DCP1AmRNA-decapping enzyme 1AENZ: enzyme, enzymatic propertiesUrineTranscriptional co-activatorGastric cancer23932921
933NAV1Neuron navigator 1DEV: developmentUrineneuronal migration
803ZFYVE20Rabenosyn-5TP: transport, storage, endocytosis, exocytosis, vesiclesUrineendosomal transport
773PLA1APhospholipase A1 member AENZ: enzyme, enzymatic propertiesUrineLipid metabolismProstate cancer22904677
53GLB1L β-galactosidase-1-like proteinENZ: enzyme, enzymatic propertiesUrineGlycosyl hydrolase, carbohydrate metabolism
322COX4I2Cytochrome c oxidase subunit 4 isoform 2, mitochondrialExocrine pancreatic insufficiency, dyserythropoietic anemia, calvarial hyperostosisTP: transport, storage, endocytosis, exocytosis, vesiclesUrineMitochondrial electron transportGeneral (Warburg effect)22320183
202SOS2Son of sevenless homolog 2MOD: modulator, regulatorUrineGuanine-nucleotide releasing factor
202GALNT6Polypeptide N-acetylgalactosa minyltransferase 6ENZ: enzyme, enzymatic propertiesUrinePost-translational protein O-linked glycosylationBreast cancer20215525
172CCDC88CProtein DapleAutosomal recessive nonsyndromic hydrocephalus HYC1SIG: signalingUrineNegative regulator of canonical Wnt signaling pathwayBreast cancer23593120
152TTI1TEL2-interacting protein 1 homologMOD: modulator, regulatorUrineRegulator of DNA damage responseMultiple myeloma23263282
132RPGRIP1X-linked retinitis-pigmentosa GTPase regulator interacting protein 1Leber congenital amaurosis 6CS: Cell shapeUrineSensory transduction
132GBP4Guanylate-binding protein 4DEV: developmentUrineGTP hydrolysis
112MTTPMicrosomal triglyceride transfer protein large subunitAβ lipoproteinemiaTP: transport, storage, endocytosis, exocytosis, vesiclesUrineLipid transport, plasma lipoprotein secretionSmall intestinal cancer12630961
112ERBB2Receptor tyrosine-protein kinase erbB-2Glioma susceptibility 1; ovarian cancer; lung cancer; gastric cancerENZ: enzyme, enzymatic propertiesUrineProtein tyrosine kinase involved in transcriptional regulationMultiple22014070
92PLEKHG2Pleckstrin homology domain-containing family G member 2MOD: modulator, regulatorUrineGuanine-nucleotide releasing factorPancreatic cancer24041470
72POLA2DNA polymerase α subunit BTF: transcription and translationUrineDNA replication and cell proliferationMelanoma24987109
62GPSM2G-protein-signaling modulator 2Deafness, autosomal recessive 82CC: cell cycle (turnover, mitosis, meiosis)UrineG-protein coupled receptor signaling pathway, spindle pole orientationBreast cancer20589935
62GDNFGlial cell line-derived neurotrophic factorCentral hypoventilation syndrome; Hirschsprung disease, susceptibility to, 3SIG: signalingUrineneurotrophic factorPancreatic cancer20960036
52DDHD2Phospholipase DDHD2ENZ: enzyme, enzymatic propertiesUrineLipid degradation and metabolismBreast cancer20940404
42TEAD2Transcriptional enhancer factor TEF-4TF: transcription and translationUrineTranscription regulationProstate cancer19478945
32SARM1Sterile α and TIR motif-containing protein 1MOD: modulator, regulatorUrineRegulator of Toll-like receptor signaling pathwayColorectal cancer20426761
32DOK7Downstream of tyrosine kinase 7Myasthenia, limb-girdleSIG: signalingUrineNeuromuscular synaptogenesisBreast cancer23054610
22ZDHHC6Probable palmitoyltransferase ZDHHC6ENZ: enzyme, enzymatic propertiesUrineProtein palmitoylation
Detected in urine from cancer patients and other tissues, high confidence dataset
95HIST3H3Histone H3.1tTF: transcription and translationKidney, urineTranscription regulation, DNA repair, DNA replication
254HIST1H1EHistone H1.4TF: transcription and translationKidney, urineRegulator of gene transcriptionEndometrial cancer cells23682076
34BOLA2BolA-like protein 2UK: unknownKidney, urineRedox controlLiver cancer22653869
1163NAV2Neuron navigator 2ENZ: enzyme, enzymatic propertiesBlood, urineNeuronal developmentColorectal carcinoma22810696
253MLL3Histone-lysine N-methyltransferase MLL3ENZ: enzyme, enzymatic propertiesBlood, urineTranscriptional coactivationColorectal cancer21853109
392FMR1Fragile X mental retardation 1 proteinFragile x tremor/ataxia syndrome; fragile x mental retardation syndrome; premature ovarian failure 1TF: transcription and translationKidney, urineTranslation repressorHepatocellular carcinoma17786358
372TJP1Tight junction protein ZO-1CS: cell shapeKidney, urineTight junction assembly, cell migrationNon-small cell lung cancer24294375
352PCDH17 Protocadherin-17CS: cell shapeBlood, urineCalcium-dependent cell-adhesion proteinLaryngeal squamous cell carcinoma21213369
302NSUN5Putative methyltransferase NSUN5Williams-Beuren syndromeENZ: enzyme, enzymatic propertiesBlood, urineMethyl-transferase, embryonic development
132ST14Suppressor of tumorigenicity 14 proteinIchthyosis with hypotrichosis, autosomal recessiveENZ: enzyme, enzymatic propertiesKidney, urineDegradation of extracellular matrixBreast cancer20716618
132SONBax antagonist selected in saccharomyces 1TF: transcription and translationKidney, urineSplicing cofactor
92CPB1Carboxypeptidase BENZ: enzyme, enzymatic propertiesBlood, urineCarboxypeptidase, protein degradationPancreatic cancer1688389
62HBG2Hemoglobin subunit γ-2Cyanosis transient neonatalTP: transport, storage, endocytosis, exocytosis, vesiclesBlood, kidney, urineOxygen transport
62AKAP2A-kinase anchor protein 2SCA: scaffolder, docking, adaptorKidney, urineProtein kinase A-anchoring proteinOvarian cancer19123201
52DYNLL2Dynein light chain 2, cytoplasmicTP: transport, storage, endocytosis, exocytosis, vesiclesBlood, kidney, urineMicrotubule-based transport
52NCOR1Nuclear receptor corepressor 1TF: transcription and translationBlood, urineTranscriptional repressorProstate cancer20466759
52MKXHomeobox protein MohawkTF: transcription and translationKidney, urineMorphogenetic regulator of cell adhesion
42ACTN2α-actinin-2Cardiomyopathy, dilated, 1aaCS: cell shapeBlood, kidney, urine Actin-anchoringMetastatic pancreatic endocrine neoplasm15448002
42CORO1ACoronin-1AImmunodeficiency 8CS: cell shapeBlood, kidney, urineCrucial component of cytoskeletal modulationBreast cancer21489049
22CELF5CUGBP Elav-like family member 5TF: transcription and translationKidney, urineRegulation of pre-mRNA alternative splicing

[i] Molecules found uniquely in our urinary dataset (with peptide and spectral counts of at least two) but not in non-cancer urine samples are listed by gene and protein names, their individual peptide and spectral counts, and whether they are known to be associated with human disease based on the OMIM database. The tissue type in which the molecule was found, based on meta-analysis of external datasets, a classification-tag, and the molecular function are included. Additionally, a PubMed identification number is listed if the protein has been described to be directly associated with cancer, including the cancer type. The dataset is divided based on whether the proteins were only found in our analysis, or whether they were also detected in other proteomic non-urinary screens.

Discussion

Proteomic large-scale analysis of tissues to define a cancer state can be time- and resource-consuming, especially in light of an unknown end-point. Therefore, it could be helpful to compare a novel dataset with known data in order to establish whether potential disease markers are observable, and thereby analyse a simplified dataset for the disease in question. This approach does not address the issue of quantitative comparisons, but it is rather a qualitative approach. However, the resulting list of potential candidate molecules will have a specificity of 100%. Here, we test this hypothesis by applying a subtractive analysis method in conjunction with large-scale meta-analysis of urinary datasets to screen for potential novel cancer markers observable in human urine.

An initial comparison of functional profiles of urine, blood and kidney proteomes showed no major discernible difference between those datasets. This finding, in itself is not surprising, since it is expected that these systems should reflect an overall similar composition through a combination of immediate environment and source. Blood, containing a substantial amount of cells, is also expected to show a reasonably uniform functional composition profile compared with other tissues e.g. kidney. Our novel urinary dataset, having an expected bias towards an aberrant functional profile due to overexpressed molecules associated with disease, contains more molecules involved in cellular contacts, morphology and cytoskeletal aspects, as well as transcriptional/translational components, which may be directly linked to abnormal and uncontrolled cellular growth.

Comparison of our dataset with known non-cancer urinary proteomes yielded a set of only 44 molecules specific for our cancer data, of which 68% are already known to be involved in cancer. The functional profile of those 44 proteins in comparison to the merged urinary proteome profile showed mainly an enrichment of developmental proteins (5%), signaling molecules (7%) and, most strikingly, transcriptional/ translational proteins (20%). The known cancer-associated molecules described have been suggested to be involved in hepatocellular carcinoma [κ actin (POTEKP) (25); BolA-like protein 2 (BOLA2) (26); fragile X mental retardation 1 protein (FMR1) (27)]; mammary carcinogenesis [polypeptide N-acetylgalactosaminyltrans ferase 6 (GALNT6) (28); protein Daple (CCDC88C) (29); G-protein-signaling modulator 2 (GPSM2) (30); phospho-lipase DDHD2 (DDHD2) (31); downstream of tyrosine kinase 7 (DOK7) (32); suppressor of tumorigenicity 14 protein (ST14) (33); coronin-1A (CORO1A) (34)], lung cancer [tight junction protein ZO-1 (TJP1) (35)], prostate cancer [phos-pholipase A1 member A (PLA1A) (36); transcriptional enhancer factor TEF-4 (TEAD2) (37); nuclear receptor corepressor 1 (NCOR1) (38)], ovarian cancer [A-kinase anchor protein 2 (AKAP2) (39)], colorectal cancer [sterile α and TIR motif-containing protein 1 (SARM1) (40); neuron navigator 2 (NAV2) (41); histone-lysine N-methyltransferase MLL3 (MLL3) (42)], pancreatic cancer [pleckstrin homology domain-containing family G member 2 (PLEKHG2) (43); glial cell line-derived neurotrophic factor (GDNF) (44); carboxypeptidase B (CPB1) (45); α-actinin-2 (ACTN2) (46)], gastric cancer [mRNA-decapping enzyme 1A (DCP1A) (47), a co-activator in TGF-β signaling (48)], melanoma [DNA polymerase α subunit B (POLA2) (49)], multiple myeloma [TEL2-interacting protein 1 homolog (TTI1) (50)], endometrial cancer cells [Histone H1.4 (HIST1H1E (51)], laryngeal squamous cell carcinoma [protocadherin-17 (PCDH17) (52)], and adenocarcinoma [microsomal triglyceride transfer protein large subunit (MTTP) (53)]. Additionally, the latter protein was also described to be a pivotal element in the cancer-associated muscle-wasting disease cachexia (54). Some of these proteins may be differentially regulated across a range of different cancer types and may therefore represent key cancer markers. For example, receptor tyrosine-protein kinase erbB-2 (ERBB2) has been described to be a marker for various cancer types, such as gastroesophageal (55), breast (56), lung (57), gallbladder (58) and pancreatic cancer (59), as well as uterine serous adenocarcinoma (60), and others. Another known protein to be involved in cancer progression is the mitochondrial cytochrome c oxidase subunit 4 isoform 2 (COX4I2), which is part of the Warburg effect, where cancer cells show higher propensity to produce lactate independent of oxygen presence or absence (61).

Of the proteins not previously described in association with cancer, transcription factor Bax antagonists selected in Saccharomyces 1 (SON), homeobox protein Mohawk (MKX) and CUGBP Elav-like family member 5 (CELF5) may represent other potential lead candidates in cancer stratification. Other important markers may include developmental molecules, such as guanylate-binding protein 4 GBP4, which is a negative regulator of virus-triggered cellular responses (62) and is involved in GTP hydrolysis, or neuron navigator NAV1, which has been reported to be a neuronal guidance molecule (63). However, its role in cancer or outside the neuronal environment remains to be elucidated.

In conclusion, we have demonstrated that a subtractive analysis of proteomic datasets can yield a number of potential diagnostic cancer targets in human urine. Further specific screening of urine, based on our findings, using, for example, an antibody-based approach, will establish whether our potential markers are associated with a general cancer status, or if they are specific for a defined cancer type such as pancreatic or esophageal cancer. Additionally, since the data in our database can easily be expanded to contain further datasets, there are other, as yet undefined diseases, which can be addressed by establishing and comparing a relatively small disease-specific dataset. This approach also has the advantage of rapid turnover and increased cost-effectiveness relating to large-scale analyses of tissue and cell proteomes for the discovery of novel molecular markers. In this regard, we are encouraging researchers to submit their published datasets to be incorporated in the LSSR database. All data will be freely available through the PADB portal at www.PADB.org.

Acknowledgements

We thank C.A. Greig, N.A. Stephens and H. Wackerhage for patient recruitment. Funding of this study was provided by the University of Edinburgh.

References

1 

Good DM, Thongboonkerd V, Novak J, Bascands JL, Schanstra JP, Coon JJ, Dominiczak A and Mischak H: Body fluid proteomics for biomarker discovery: Lessons from the past hold the key to success in the future. J Proteome Res. 6:4549–4555. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Marshall T and Williams K: Two-dimensional electrophoresis of human urinary proteins following concentration by dye precipitation. Electrophoresis. 17:1265–1272. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Pieper R, Gatlin CL, McGrath AM, Makusky AJ, Mondal M, Seonarain M, Field E, Schatz CR, Estock MA, Ahmed N, et al: Characterization of the human urinary proteome: A method for high-resolution display of urinary proteins on two-dimensional electrophoresis gels with a yield of nearly 1400 distinct protein spots. Proteomics. 4:1159–1174. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Büeler MR, Wiederkehr F and Vonderschmitt DJ: Electrophoretic, chromatographic and immunological studies of human urinary proteins. Electrophoresis. 16:124–134. 1995. View Article : Google Scholar : PubMed/NCBI

5 

Spahr CS, Davis MT, McGinley MD, Robinson JH, Bures EJ, Beierle J, Mort J, Courchesne PL, Chen K, Wahl RC, et al: Towards defining the urinary proteome using liquid chromatography-tandem mass spectrometry. I Profiling an unfractionated tryptic digest. Proteomics. 1:93–107. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Cadieux PA, Beiko DT, Watterson JD, Burton JP, Howard JC, Knudsen BE, Gan BS, McCormick JK, Chambers AF, Denstedt JD, et al: Surface-enhanced laser desorption/ionization-time of flight-mass spectrometry (SELDI-TOF-MS): A new proteomic urinary test for patients with urolithiasis. J Clin Lab Anal. 18:170–175. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Roelofsen H, Alvarez-Llamas G, Schepers M, Landman K and Vonk RJ: Proteomics profiling of urine with surface enhanced laser desorption/ionization time of flight mass spectrometry. Proteome Sci. 5:22007. View Article : Google Scholar : PubMed/NCBI

8 

Vanhoutte KJ, Laarakkers C, Marchiori E, Pickkers P, Wetzels JF, Willems JL, van den Heuvel LP, Russel FG and Masereeuw R: Biomarker discovery with SELDI-TOF MS in human urine associated with early renal injury: Evaluation with computational analytical tools. Nephrol Dial Transplant. 22:2932–2943. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Husi H, Stephens N, Cronshaw A, MacDonald A, Gallagher I, Greig C, Fearon KC and Ross JA: Proteomic analysis of urinary upper gastrointestinal cancer markers. Proteomics Clin Appl. 5:289–299. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Petri AL, Simonsen AH, Yip TT, Hogdall E, Fung ET, Lundvall L and Hogdall C: Three new potential ovarian cancer biomarkers detected in human urine with equalizer bead technology. Acta Obstet Gynecol Scand. 88:18–26. 2009. View Article : Google Scholar

11 

Tsui KH, Tang P, Lin CY, Chang PL, Chang CH and Yung BY: Bikunin loss in urine as useful marker for bladder carcinoma. J Urol. 183:339–344. 2010. View Article : Google Scholar

12 

Chen YT, Chen CL, Chen HW, Chung T, Wu CC, Chen CD, Hsu CW, Chen MC, Tsui KH, Chang PL, et al: Discovery of novel bladder cancer biomarkers by comparative urine proteomics using iTRAQ technology. J Proteome Res. 9:5803–5815. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Tan LB, Chen KT, Yuan YC, Liao PC and Guo HR: Identification of urine PLK2 as a marker of bladder tumors by proteomic analysis. World J Urol. 28:117–122. 2010. View Article : Google Scholar

14 

Xue A, Scarlett CJ, Chung L, Butturini G, Scarpa A, Gandy R, Wilson SR, Baxter RC and Smith RC: Discovery of serum biomarkers for pancreatic adenocarcinoma using proteomic analysis. Br J Cancer. 103:391–400. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Schröder C, Jacob A, Tonack S, Radon TP, Sill M, Zucknick M, Rüffer S, Costello E, Neoptolemos JP, Crnogorac-Jurcevic T, et al: Dual-color proteomic profiling of complex samples with a microarray of 810 cancer-related antibodies. Mol Cell Proteomics. 9:1271–1280. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Good DM, Zürbig P, Argilés A, Bauer HW, Behrens G, Coon JJ, Dakna M, Decramer S, Delles C, Dominiczak AF, et al: Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics. 9:2424–2437. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Zhang Y, Zhang Y, Adachi J, Olsen JV, Shi R, de Souza G, Pasini E, Foster LJ, Macek B, Zougman A, et al: MAPU: Max-Planck Unified database of organellar, cellular, tissue and body fluid proteomes. Nucleic Acids Res. 35:D771–D779. 2007. View Article : Google Scholar :

18 

Li SJ, Peng M, Li H, Liu BS, Wang C, Wu JR, Li YX and Zeng R: Sys-BodyFluid: A systematical database for human body fluid proteome research. Nucleic Acids Res. 37:D907–D912. 2009. View Article : Google Scholar :

19 

Agron IA, Avtonomov DM, Kononikhin AS, Popov IA, Moshkovskii SA and Nikolaev EN: Accurate mass tag retention time database for urine proteome analysis by chromatography-mass spectrometry. Biochemistry (Mosc). 75:636–641. 2010. View Article : Google Scholar

20 

Carson JM, Okamura K, Wakashin H, McFann K, Dobrinskikh E, Kopp JB and Blaine J: Podocytes degrade endocytosed albumin primarily in lysosomes. PLoS One. 9:e997712014. View Article : Google Scholar : PubMed/NCBI

21 

Collins MO, Yu L and Choudhary JS: Analysis protein complexes by 1D-SDS-PAGE and tandem mass spectrometry. Protocol Exchange. 2008. View Article : Google Scholar

22 

Kentsis A, Monigatti F, Dorff K, Campagne F, Bachur R and Steen H: Urine proteomics for profiling of human disease using high accuracy mass spectrometry. Proteomics Clin Appl. 3:1052–1061. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Adachi J, Kumar C, Zhang Y, Olsen JV and Mann M: The human urinary proteome contains more than 1500 proteins, including a large proportion of membrane proteins. Genome Biol. 7:R802006. View Article : Google Scholar : PubMed/NCBI

24 

Gonzales PA, Pisitkun T, Hoffert JD, Tchapyjnikov D, Star RA, Kleta R, Wang NS and Knepper MA: Large-scale proteomics and phosphoproteomics of urinary exosomes. J Am Soc Nephrol. 20:363–379. 2009. View Article : Google Scholar :

25 

Chang KW, Yang PY, Lai HY, Yeh TS, Chen TC and Yeh CT: Identification of a novel actin isoform in hepatocellular carcinoma. Hepatol Res. 36:33–39. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Hunecke D, Spanel R, Länger F, Nam SW and Borlak J: MYC-regulated genes involved in liver cell dysplasia identified in a transgenic model of liver cancer. J Pathol. 228:520–533. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Liu Y, Zhu X, Zhu J, Liao S, Tang Q, Liu K, Guan X, Zhang J and Feng Z: Identification of differential expression of genes in hepatocellular carcinoma by suppression subtractive hybridization combined cDNA microarray. Oncol Rep. 18:943–951. 2007.PubMed/NCBI

28 

Park JH, Nishidate T, Kijima K, Ohashi T, Takegawa K, Fujikane T, Hirata K, Nakamura Y and Katagiri T: Critical roles of mucin 1 glycosylation by transactivated polypeptide N-acetylgalactosaminyltransferase 6 in mammary carcinogenesis. Cancer Res. 70:2759–2769. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Long J, Zhang B, Signorello LB, Cai Q, Deming-Halverson S, Shrubsole MJ, Sanderson M, Dennis J, Michailidou K, Easton DF, et al: Evaluating genome-wide association study-identified breast cancer risk variants in African-American women. PLoS One. 8:e583502013. View Article : Google Scholar : PubMed/NCBI

30 

Fukukawa C, Ueda K, Nishidate T, Katagiri T and Nakamura Y: Critical roles of LGN/GPSM2 phosphorylation by PBK/TOPK in cell division of breast cancer cells. Genes Chromosomes Cancer. 49:861–872. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Yang ZQ, Liu G, Bollig-Fischer A, Giroux CN and Ethier SP: Transforming properties of 8p11–12 amplified genes in human breast cancer. Cancer Res. 70:8487–8497. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Heyn H, Carmona FJ, Gomez A, Ferreira HJ, Bell JT, Sayols S, Ward K, Stefansson OA, Moran S, Sandoval J, et al: DNA methylation profiling in breast cancer discordant identical twins identifies DOK7 as novel epigenetic biomarker. Carcinogenesis. 34:102–108. 2013. View Article : Google Scholar :

33 

Kauppinen JM, Kosma VM, Soini Y, Sironen R, Nissinen M, Nykopp TK, Kärjä V, Eskelinen M, Kataja V and Mannermaa A: ST14 gene variant and decreased matriptase protein expression predict poor breast cancer survival. Cancer Epidemiol Biomarkers Prev. 19:2133–2142. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Hattori N, Okochi-Takada E, Kikuyama M, Wakabayashi M, Yamashita S and Ushijima T: Methylation silencing of angio-poietin-like 4 in rat and human mammary carcinomas. Cancer Sci. 102:1337–1343. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Ni S, Xu L, Huang J, Feng J, Zhu H, Wang G and Wang X: Increased ZO-1 expression predicts valuable prognosis in non-small cell lung cancer. Int J Clin Exp Pathol. 6:2887–2895. 2013.PubMed/NCBI

36 

Paulo P, Ribeiro FR, Santos J, Mesquita D, Almeida M, Barros-Silva JD, Itkonen H, Henrique R, Jerónimo C, Sveen A, et al: Molecular subtyping of primary prostate cancer reveals specific and shared target genes of different ETS rearrangements. Neoplasia. 14:600–611. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Blum R, Gupta R, Burger PE, Ontiveros CS, Salm SN, Xiong X, Kamb A, Wesche H, Marshall L, Cutler G, et al: Molecular signatures of prostate stem cells reveal novel signaling pathways and provide insights into prostate cancer. PLoS One. 4:e57222009. View Article : Google Scholar : PubMed/NCBI

38 

Battaglia S, Maguire O, Thorne JL, Hornung LB, Doig CL, Liu S, Sucheston LE, Bianchi A, Khanim FL, Gommersall LM, et al: Elevated NCOR1 disrupts PPARalpha/gamma signaling in prostate cancer and forms a targetable epigenetic lesion. Carcinogenesis. 31:1650–1660. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Quinn MC, Filali-Mouhim A, Provencher DM, Mes-Masson AM and Tonin PN: Reprogramming of the transcriptome in a novel chromosome 3 transfer tumor suppressor ovarian cancer cell line model affected molecular networks that are characteristic of ovarian cancer. Mol Carcinog. 48:648–661. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Quyun C, Ye Z, Lin SC and Lin B: Recent patents and advances in genomic biomarker discovery for colorectal cancers. Recent Pat DNA Gene Seq. 4:86–93. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Watanabe Y, Castoro RJ, Kim HS, North B, Oikawa R, Hiraishi T, Ahmed SS, Chung W, Cho MY, Toyota M, et al: Frequent alteration of MLL3 frameshift mutations in microsatellite deficient colorectal cancer. PLoS One. 6:e233202011. View Article : Google Scholar : PubMed/NCBI

43 

Shain AH, Salari K, Giacomini CP and Pollack JR: Integrative genomic and functional profiling of the pancreatic cancer genome. BMC Genomics. 14:6242013. View Article : Google Scholar : PubMed/NCBI

44 

Liu H, Ma Q and Li J: High glucose promotes cell proliferation and enhances GDNF and RET expression in pancreatic cancer cells. Mol Cell Biochem. 347:95–101. 2011. View Article : Google Scholar

45 

Fernstad R, Pousette A, Carlström K and Sköldefors H: A novel assay for pancreatic cellular damage: IV. Serum concentrations of pancreas-specific protein (PASP) in acute pancreatitis and other abdominal diseases. Pancreas. 5:42–49. 1990. View Article : Google Scholar : PubMed/NCBI

46 

Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ and Maitra A: Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 10:6152–6158. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Iio A, Takagi T, Miki K, Naoe T, Nakayama A and Akao Y: DDX6 post-transcriptionally down-regulates miR-143/145 expression through host gene NCR143/145 in cancer cells. Biochim Biophys Acta. 1829:1102–1110. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Bai RY, Koester C, Ouyang T, Hahn SA, Hammerschmidt M, Peschel C and Duyster J: SMIF, a Smad4-interacting protein that functions as a co-activator in TGFbeta signaling. Nat Cell Biol. 4:181–190. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Lu YC, Yao X, Crystal JS, Li YF, El-Gamil M, Gross C, Davis L, Dudley ME, Yang JC, Samuels Y, et al: Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin Cancer Res. 20:3401–3410. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Fernández-Sáiz V, Targosz BS, Lemeer S, Eichner R, Langer C, Bullinger L, Reiter C, Slotta-Huspenina J, Schroeder S, Knorn AM, et al: SCFFbxo9 and CK2 direct the cellular response to growth factor withdrawal via Tel2/Tti1 degradation and promote survival in multiple myeloma. Nat Cell Biol. 15:72–81. 2013. View Article : Google Scholar

51 

Lee LR, Teng PN, Nguyen H, Hood BL, Kavandi L, Wang G, Turbov JM, Thaete LG, Hamilton CA, Maxwell GL, et al: Progesterone enhances calcitriol antitumor activity by upregulating vitamin D receptor expression and promoting apoptosis in endometrial cancer cells. Cancer Prev Res (Phila). 6:731–743. 2013. View Article : Google Scholar

52 

Giefing M, Zemke N, Brauze D, Kostrzewska-Poczekaj M, Luczak M, Szaumkessel M, Pelinska K, Kiwerska K, Tönnies H, Grenman R, et al: High resolution ArrayCGH and expression profiling identifies PTPRD and PCDH17/PCH68 as tumor suppressor gene candidates in laryngeal squamous cell carcinoma. Genes Chromosomes Cancer. 50:154–166. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Al-Shali K, Wang J, Rosen F and Hegele RA: Ileal adenocarcinoma in a mild phenotype of abetalipoproteinemia. Clin Genet. 63:135–138. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Silvério R, Laviano A, Rossi Fanelli F and Seelaender M: L-Carnitine induces recovery of liver lipid metabolism in cancer cachexia. Amino Acids. 42:1783–1792. 2012. View Article : Google Scholar

55 

Hjortland GO, Meza-Zepeda LA, Beiske K, Ree AH, Tveito S, Hoifodt H, Bohler PJ, Hole KH, Myklebost O, Fodstad O, et al: Genome wide single cell analysis of chemotherapy resistant metastatic cells in a case of gastroesophageal adenocarcinoma. BMC Cancer. 11:4552011. View Article : Google Scholar : PubMed/NCBI

56 

Adachi R, Horiuchi S, Sakurazawa Y, Hasegawa T, Sato K and Sakamaki T: ErbB2 down-regulates microRNA-205 in breast cancer. Biochem Biophys Res Commun. 411:804–808. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Janku F, Garrido-Laguna I, Petruzelka LB, Stewart DJ and Kurzrock R: Novel therapeutic targets in non-small cell lung cancer. J Thorac Oncol. 6:1601–1612. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Goldin RD and Roa JC: Gallbladder cancer: A morphological and molecular update. Histopathology. 55:218–229. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Komoto M, Nakata B, Amano R, Yamada N, Yashiro M, Ohira M, Wakasa K and Hirakawa K: HER2 overexpression correlates with survival after curative resection of pancreatic cancer. Cancer Sci. 100:1243–1247. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Elsahwi KS and Santin AD: erbB2 overexpression in uterine serous cancer: A molecular target for trastuzumab therapy. Obstet Gynecol Int. 2011:1282952011. View Article : Google Scholar : PubMed/NCBI

61 

Mazzio EA, Boukli N, Rivera N and Soliman KF: Pericellular pH homeostasis is a primary function of the Warburg effect: Inversion of metabolic systems to control lactate steady state in tumor cells. Cancer Sci. 103:422–432. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Hu Y, Wang J, Yang B, Zheng N, Qin M, Ji Y, Lin G, Tian L, Wu X, Wu L, et al: Guanylate binding protein 4 negatively regulates virus-induced type I IFN and antiviral response by targeting IFN regulatory factor 7. J Immunol. 187:6456–6462. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Maes T, Barceló A and Buesa C: Neuron navigator: A human gene family with homology to unc-53, a cell guidance gene from Caenorhabditis elegans. Genomics. 80:21–30. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 48 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Husi H, Skipworth RJ, Cronshaw A, Fearon KC and Ross JA: Proteomic identification of potential cancer markers in human urine using subtractive analysis. Int J Oncol 48: 1921-1932, 2016
APA
Husi, H., Skipworth, R.J., Cronshaw, A., Fearon, K.C., & Ross, J.A. (2016). Proteomic identification of potential cancer markers in human urine using subtractive analysis. International Journal of Oncology, 48, 1921-1932. https://doi.org/10.3892/ijo.2016.3424
MLA
Husi, H., Skipworth, R. J., Cronshaw, A., Fearon, K. C., Ross, J. A."Proteomic identification of potential cancer markers in human urine using subtractive analysis". International Journal of Oncology 48.5 (2016): 1921-1932.
Chicago
Husi, H., Skipworth, R. J., Cronshaw, A., Fearon, K. C., Ross, J. A."Proteomic identification of potential cancer markers in human urine using subtractive analysis". International Journal of Oncology 48, no. 5 (2016): 1921-1932. https://doi.org/10.3892/ijo.2016.3424