BIOMARKERS FOR BREAST CANCER

Abstract
The present invention uses 2-dimensional differential gel electrophoresisgel (2D-DIGE) and mass spetrum techniques to identify breast cancer biomarkers in transformed breast cells. In summary, the present invention identifies numerous putative breast cancer markers from various stages of breast cancer. The results of the invention aids in developing proteins identified as useful diagnostic and therapeutic candidates on breast cancer research.
Description
FIELD OF THE INVENTION

The present invention relates to biomarkers for breast cancer. More specifically, the invention relates to biomarkers that can be used in diagnosis, determination of disease severity, and monitoring of therapeutic response of patients with breast cancer. The method is based on the use of 2-dimensional differential gel electrophoresisgel (2D-DIGE) to quantitatively identify biomarkers in breast cancer.


BACKGROUND OF THE INVENTION

Breast cancer is one of the leading causes of death among women around the world. The 5-year survival rate for breast cancer is close to 97% when tumors are confined to breast tissue, but decrease dramatically to 23% when tumors have metastasized to other organs at the time of diagnosis. Presymtomatic screening to detect early-stage breast cancer while it is still resectable with potential for cure can greatly reduce breast cancer-related mortality. Unfortunately, only 63% (1992-1999, US) of the breast cancers are localized at the time of diagnosis (Jemal, A. et al. (2004) CA Cancer J. Clin. 54:8-29). Small lesions are frequently missed and may not be visible, even by mammography, particularly in young women and women with dense breast tissue. Molecular markers that can potentially identify these small lesions that are invisible to imaging techniques will provide a real opportunity to treat a neoplasm before it invades tissue.


Previous inventions indicate that the transformation and metastasis of normal breast cells are correlated to altered expression in both transcription and translation levels (Kulasingam, V. & Diamandis, E. P. Mol. Cell. Proteomics 2007, 6, 1997). To better understand the molecular mechanisms associated with tumorigenesis and metastasis, it is necessary to identify the gene expression signatures and protein expression markers among normal breast cells, noninvasive breast cancer cells, and invasive breast cancer cells. At the transcription level, microarray strategies have been used to classify breast tumors as highly invasive and noninvasive cancer. At the translation level, proteomic strategies have been used to discern cancer markers from noninvasive and invasive breast cells. Nagaraja et al. compared the proteomic profiling of cell lines corresponding to normal breast cells, non-invasive breast cancer cells, and invasive breast cancer cells using 2-DE (Nagaraja, G. M.; Othman, M.; Fox, B. P.; Alsaber, R.; Pellegrino, C. M.; Zeng, Y.; Khanna, R.; Tamburini, P.; Swaroop, A.; Kandpal, R. P. Oncogene 2006, 25, 2328). Although they identified 26 spots as potential cancer markers, no statistical analysis was included in their study. Pucci-Minafra et al. compared a ductal infiltrating carcinoma-derived cell line with a non-tumoral mammary epithelial cell line using 2-DE, silver staining, immunodetection, and N-terminal sequencing and identified 58 differentially expressed proteins (Pucci-Minafra, I.; Fontana, S.; Cancemi, P.; Alaimo, G; Minafra, S. Ann. N.Y. Acad. Sci. 2002, 963, 122). In contrast to these cell line based experiments, Pawlik analyzed differentially expressed proteins among nipple aspirate fluid samples from tumor-bearing and disease-free breasts (Pawlik, T. M.; Hawke, D. H.; Liu, Y.; Krishnamurthy, S.; Fritsche, H.; Hunt, K. K.; Kuerer, H. M. BMC. Cancer 2006, 6, 68). Although these identified proteins are primarily abundant proteins, few of them have been validated as biomarkers.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Optimization of starvation time for secretomic analysis. MCF-10A, MCF-7 and MDA-MB-231 on cell culture dishes were used to check starvation induced cell autolysis by detecting the release of cytoplasmic proteins, LDH and β-tubulin in serum-free media. The serum-free media were harvested and concentrated 1000-fold at indicative starvation periods prior to performing immunoblotting analysis.



FIG. 2. Secretomic comparisons across MCF-10A, MCF-7 and MDA-MB-231 cells using 2D-DIGE. Protein sample (50 μg each) enriched from serum-free media was labeled with Cy-dyes and separated using 24 cm, pH 3-10 non-linear IPG strips. 2D-DIGE images of MCF-10A, MCF-7, and MDA-MB-231 at appropriate excitation and emission wavelengths were pseudo-colored and overlaid with ImageQuant Tool (GE Healthcare).



FIG. 3. Percentage of secreted proteins identified from serum-free media by 2D-DIGE/MALDI-TOF MS for MCF-10A, MCF-7 and MDA-MB-231 cells according to their sub-cellular locations (A) and biological functions (B).



FIG. 4. Proteomic comparisons among MCF-10A, MCF-7 and MDA-MB-231 cells using 2D-DIGE. Protein samples (150 μg each) purified from total cell lysates were labeled with Cy-dyes and separated using 24 cm, pH 3-10 non-linear IPG strips. 2D-DIGE images of MCF-10A, MCF-7 and MDA-MB-231 at appropriate excitation and emission wavelengths were pseudo-colored and overlaid with ImageQuant Tool (GE Healthcare).



FIG. 5. Percentage of total cellular proteins identified by 2D-DIGE/MALDI-TOF MS for MCF-10A, MCF-7 and MDA-MB-231 cells according to their sub-cellular locations (A) and biological functions (B).



FIG. 6. Representative immunoblotting and immunofluorescent analyses for selected differentially expressed proteins identified by proteomic analysis in MCF-10A, MCF-7 and MDA-MB-231 cells. The levels of identified proteins in serum-free media, (A) Cyclophilin A, (B) 14-3-3 delta and (C) Peroxiredoxin 2 and total cellular proteins, (D) Profilin, (E) Cathepsin D, (F) Annexin 2 and (G) Protein disulfide isomerase A1 in MDA-MB-231 and MCF-7 versus MCF-10A confirmed by immunoblot (left top panels), densitometry results with normalized values using nonspecific bands (NS) of secreted proteins and β-tubulin as loading controls (left bottom panels), protein expression map (right top panels) and three-dimensional spot image (right bottom panels). (H) MCF-10A, MCF-7 and MDA-MB-231 cells were fixed and incubated with anti-HDAC antibody and stained with a Texas Red-conjugated secondary antibody (Red). Nuclei were stained with DAPI (Blue). Each set of three fields was taken using the same exposure, and images are representative of three different fields. Scale bar=20 μm.



FIG. 7. Immunoblotting and immunofluorescence analyses of the expression and protein localization changes of newly identified putative breast cancer markers across MCF-10A, MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 cells. (A) The profile of the secreted proteome changes across MCF-10A, MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 cells. The serum-free media from the cell lines was concentrated and 10 μg of the total protein was resolved using SDS-PAGE and immunoblotted for MPP2 and Bestrophin 3. NS represents a nonspecific band used to show equal loading of secreted proteins. (B) 5×104 MCF-10A, MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 cells were seeded on cover slips before fixation and staining for Parvabumin, BANF1, PdLIM1 and IFIT3. Each set of three fields was taken using the same exposure, and images are representative of three different fields. Scale bar=20 μm.



FIG. 8. Expression profiles for proteins potentially contributing to (A) cell cycle (B) redox regulation (C) carbohydrate metabolism (D) calcium regulation (E) vascular transport (F) protease inhibition in comparing MCF-7 and MDA-MB-231 with MCF-10A. White bars represent fold change in protein expression in MDA-MB-231 versus MCF-10A. Black bars represent fold change in protein expression in MCF-7 versus MCF-10A. The vertical axis indicates the identified proteins; the horizontal axis indicates the fold change in protein expression.





SUMMARY OF THE INVENTION

The present invention provides a breast cancer biomarker library, comprising bestrophin-3, carbonic anhydrase 2, dynein heavy chain 6, ecto-ADP-ribosyltransferase 4, GRAM domain-containing protein 2, interferon-induced protein with tetratricopeptide repeat 3, phosphoglycerate mutase 1, proteasome subunit alpha type-1, proteasome subunit alpha type-3, rab GTPase-binding effector protein 2, Ras-related protein Rab-2B, selenium-binding protein 1, transmembrane protein C14orf180, vascular protein sorting-associated protein 54, achaete-scute homologue 4, aconitate hydratase, aminopeptidase B, annexin A3, barrier-to-autointegration factor, bifunctional purine biosynthesis, calumenin, carbonic anhydrase 2, coiled-coil domain-containing protein, erlin-2, F-actin-capping protein subunit beta, flavin reductase, fructose-1,6-biphosphatase 1, fructose-biphosphate ldolase A, heat shock protein 75 kDa, heterogeneous nuclear ribonucleoproteins A2/B1, leukotriene A-4 hydrolase, microfibrillar-associated protein 3 like, microtubule-associated protein RP, nuclear distribution protein nudE homologue 1, parvalbumin alpha, PDZ and LIM domain protein 1, peptidylprolyl isomerase domain and WD repeat-containing protein 1, phosphoserine aminotransferase, plastin-3, programmed cell death 6-interacting protein, proteasome activator complex subunit 1, proteasome activator complex subunit 2, protein canopy homologue 2, protein CASC2, protein disulfide-isomerase A6, protein SHQ1, Rab GDP dissociation inhibitor beta, reticulocalbin-2, Rho GTPase-activating protein 25, Rho GTPase-activating protein 5, ribonuclease inhibitor, selenium-binding protein 1, septin-11, septin-8, serine/threonine-protein kinase Nek7, serine/threonine-protein kinase PCTAIRE-1, small ubiqutin-related modifier 3, stress-induced phosphoprotein 1, thioredoxin domain-containing protein 5, ubiquitin-conjugating enzyme E2, UPF0492 protein C20orf94, voltage-dependent anion-selective channel protein and zinc finger protein 433.


The present invention also provides a method of predicting an increased likelihood of developing breast cancer progression of a subject comprising:

    • (a) detecting the expression of at least one biomarker in a sample from a subject with bioassays, wherein the biomarker is selected from the biomarker library aformentioned; and
    • (b) comparing the pattern of biomarker expression in the previous step to a reference biomarker expression pattern from normal tissue, wherein at least one fold of increasing or decreasing biomarker expression relative to the reference indicating an increased likelihood of breast cancer development.


DETAILED DESCRIPTION OF THE INVENTION

The present invention provides a biomarker library for breast cancer, and a method of predicting an increased likelihood of developing breast cancer progression using the same.


Biomarker Library

Breast cancer is the leading cause of cancer-induced mortality in women. Early detection of breast cancer greatly improves its survival rates. The identification of cellular targets that play a role in highly invasive breast cancer may also contribute to a better understanding of the biological mechanisms inherent in the aggressive progression of cancer and may also be used in the development of new diagnostic or therapeutic strategies for breast cancer.


Accordingly, the aim of the present invention is to discover biomarkers with the greatest potential to facilitate early detection of breast cancer and monitor the progress of breast tumorigenesis. Numerous proteins, including bestrophin-3 and parvalbumin, are highly expressed in both low-invasive and aggressive breast cancer cells and are verified as breast cancer markers in this invention. Importantly, several of these identified proteins, including bestrophin-3, GRAMD2, and nuclear distribution protein nudE homolog 1, have not been reported in previous breast cancer studies, thus implying these proteins are valuable breast cancer markers.


Biomarkers of the present invention can be used in diagnosis, including determination of disease severity and monitoring of therapeutic response of patients with breast cancer. The biomarker library is listed as follows: bestrophin-3, carbonic anhydrase 2, dynein heavy chain 6, ecto-ADP-ribosyltransferase 4, GRAM domain-containing protein 2, interferon-induced protein with tetratricopeptide repeat 3, phosphoglycerate mutase 1, proteasome subunit alpha type-1, proteasome subunit alpha type-3, rab GTPase-binding effector protein 2, Ras-related protein Rab-2B, selenium-binding protein 1, transmembrane protein C14orf180, vascular protein sorting-associated protein 54, achaete-scute homologue 4, aconitate hydratase, aminopeptidase B, annexin A3, barrier-to-autointegration factor, bifunctional purine biosynthesis, calumenin, carbonic anhydrase 2, coiled-coil domain-containing protein, erlin-2, F-actin-capping protein subunit beta, flavin reductase, fructose-1,6-biphosphatase 1, fructose-biphosphate ldolase A, heat shock protein 75 kDa, heterogeneous nuclear ribonucleoproteins A2/B1, leukotriene A-4 hydrolase, microfibrillar-associated protein 3 like, microtubule-associated protein RP, nuclear distribution protein nudE homologue 1, parvalbumin alpha, PDZ and LIM domain protein 1, peptidylprolyl isomerase domain and WD repeat-containing protein 1, phosphoserine aminotransferase, plastin-3, programmed cell death 6-interacting protein, proteasome activator complex subunit 1, proteasome activator complex subunit 2, protein canopy homologue 2, protein CASC2, protein disulfide-isomerase A6, protein SHQ1, Rab GDP dissociation inhibitor beta, reticulocalbin-2, Rho GTPase-activating protein 25, Rho GTPase-activating protein 5, ribonuclease inhibitor, selenium-binding protein 1, septin-11, septin-8, serine/threonine-protein kinase Nek7, serine/threonine-protein kinase PCTAIRE-1, small ubiqutin-related modifier 3, stress-induced phosphoprotein 1, thioredoxin domain-containing protein 5, ubiquitin-conjugating enzyme E2, UPF0492 protein C20orf94, voltage-dependent anion-selective channel protein and zinc finger protein 433.


Predicting Method

When using biomarkers in diagnosis, the present invention provides a direct way of predicting an increased likelihood of developing breast cancer progression of a subject comprising following steps:

    • (a) detecting the expression of at least one biomarker in a sample from a subject with bioassays, wherein the biomarker is selected from the biomarker library aforementioned; and
    • (b) comparing the pattern of biomarker expression in the previous step to a reference biomarker expression pattern from normal tissue, wherein at least one fold of increasing or decreasing biomarker expression relative to the reference indicating an increased likelihood of breast cancer development.


The expression pattern indicates the quantity of biomarker expression. In a better embodiment, the increasing or decreasing of biomarker expression is 1.5 fold relative to the biomarker expression of normal tissue.


The method further comprises applying software for protein expression comparison between normal tissue and tumor tissue.


The breast cancer progression includes the presence or absence of breast tumor, the stage of breast cancer and the effectiveness of breast cancer treatment.


The stage of breast cancer includes invasive and non-invasive tumor pregression. Invasive tumor is another synonym of cancer, and the name refers to invasion of surrounding tissues. Non-invasive tumor is a neoplasm which is not invasive but has the potential to progress to cancer (become invasive) if left untreated.


A neoplasm is the abnormal proliferation of cells. The growth of this clone of cells exceeds, and is uncoordinated with, that of the normal tissues around it. The growth persists in the same excessive manner even after cessation of the stimuli. It usually causes a lump or tumor.


The subject mentioned herein is human or mammal, and the sample is selected from blood, serum, plasma, ductal lavage fluid and nipple aspiration fluid.


The bioassay used herein comprises immunoassay, electrophoresis and mass spectrum.


The immunoassay is measured using an immunoblotting, especially an antibody-based assay. The antibody-based assay used herein is for biomarker detection, comprsing at least one vessel for antibody and biomarker interaction and a detectable label to attached with antibody. Useful detectable labels include but are not limited to radioactive labels such as 32P, 3H, and 14C; fluorescent dyes such as fluorescein isothiocyanate (FITC), rhodamine, lanthanide phosphors, Texas red, and ALEXA Fluor Dyes™ (Molecular Probes), CY™ dyes (Amersham), Spectrum Dyes (Abbott Labs); electron-dense reagents such as gold; enzymes such as horseradish peroxidase, beta-galactosidase, luciferase, and alkaline phosphatase; colorimetric labels such as colloidal gold; magnetic labels such as those sold under the mark DYNABEADS™; biotin; dioxigenin; or haptens and proteins for which antisera or monoclonal antibodies are available. The label can be directly incorporated into the polynucleotide, or it can be attached to a molecule which hybridizes or binds to the polynucleotide. The labels may be coupled to the isolated polynucleotides by any means known to those of skill in the art. In various embodiments, the isolated polynucleotides are labeled using nick translation, PCR, or random primer extension (see, e.g., Sambrook et al. supra). Methods for detecting the label include, but are not limited to spectroscopic, photochemical, biochemical, immunochemical, physical or chemical techniques.


The electrophoresis used herein is 2-dimensional differential gel electrophoresisgel (2D-DIGE), which works efficiently to identify the quantity of breast cancer biomarker expression.


2-DE is currently a key technique in profiling thousands of proteins within biological samples and plays a role complementary to LC/MS-based proteomic analysis. However, reliable quantitative comparisons between gels and gel-to-gel variations remain the primary challenge in 2-DE analysis. A significant improvement in the gel-based analysis of protein quantitation and detection was achieved by the introduction of 2D-DIGE, which can co-detect numerous samples in the same 2-DE. This approach minimizes gel-to-gel variations and compares the relative amount of protein features across different gels using an internal fluorescent standard. Moreover, the 2D-DIGE technique has the advantages of a broader dynamic range, higher sensitivity, and greater reproducibility than traditional 2-DE. This innovative technology relies on the pre-labeling of protein samples with fluorescent dyes (Cy2, Cy3 and Cy5) before electrophoresis. Each dye has a distinct fluorescent wavelength, allowing multiple experimental samples with an internal standard to be simultaneously separated in the same gel. The internal standard, which is a pool of an equal amount of the experimental protein samples, helps provide accurate normalization data and increase statistical confidence in relative quantitation among gels.


Whether or not comparisons of normal cell lines with cancer cell lines actually reflect common changes associated with cancer and can be successfully developed into clinically useful biomarkers or therapeutic targets remains debatable. Thus, a direct comparison of cancer tissue with normal tissue is the best theoretical method of obtaining protein expression signatures during tumor progression. However, a direct comparison of clinical samples increases the amount of false positives due to the heterogeneity of tumor specimens, which interferes with the identification of tumor-specific markers. For this reason, well-characterized model cell lines established from normal and tumor tissue are recognized as more informative in cancer proteomics research.


In the field of breast cancer research, MCF-10A, MCF-7 and MDA-MB-231 are widely used to represent normal luminal epithelial cells, non-invasive breast cancer cells derived from the luminal duct and invasive breast cancer cells derived from the same tissue, respectively. The present invention compares the proteomic profiles of total cellular proteins and secreted proteins of this cell model system using 2D-DIGE to quantitatively identify biomarkers in breast cancer, wherein the biomarkers reflect the progression of tumorigenesis.


The results show differentially expressed protein profiles across normal and transformed breast cell lines, ranging from extracellular secreted proteins and intracellular proteins. The 2D-DIGE strategy is powerful enough to identify numerous breast cancer signatures and offers a complementary role to LC/MS-based proteomic analysis. Even though the global coverage of protein mixtures identified by LC-MS based analysis is generally higher than that of 2-DE based analysis, 2-DE based analysis offers some distinct advantages, such as direct protein quantification at protein isoform levels instead of peptide levels to reduce analytical variations.


The examples below are non-limiting but merely representative of various aspects and features of the present invention.


EXAMPLE 1
Chemicals and Reagents

Generic chemicals were purchased from Sigma-Aldrich (St. Louis, USA), while reagents for 2D-DIGE were purchased from GE Healthcare (Uppsala, Sweden). All primary antibodies were purchased from Abcam (Cambridge, UK) and anti-mouse, anti-goat and anti-rabbit secondary antibodies were purchased from GE Healthcare (Uppsala, Sweden). All chemicals and biochemicals used in the present invention were of analytical grade.


Cell Lines and Cell Cultures

The breast epithelial cell line MCF-10A was from National Health Research Institute, Taiwan. The breast cancer cell lines MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 were purchased from American Type Culture Collection (ATCC), Manassas, Va. MCF-10A was maintained in Dulbecco's Modified Eagle's medium and F-12 medium (DMEM/F-12) supplemented with 5% horse serum, L-glutamine (2 mM), streptomycin (100 μg/mL), penicillin (100 IU/mL), epidermal growth factor (20 ng/ml) (all from Gibco-Invitrogen Corp., UK), insulin (10 μg/ml) (Sigma) and hydrocortisone (0.5 μg/ml) (Sigma). MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 were maintained in Dulbecco's Modified Eagle's medium (DMEM) supplemented with 10% (v/v) fetal calf serum (FCS), L-glutamine (2 mM), streptomycin (100 μg/mL), and penicillin (100 IU/mL) (all from Gibco-Invitrogen Corp., UK). All cells were incubated at 37° C. and 5% CO2.


Sample Preparation for Proteomic Analysis

Cells in normal growth medium at ˜80% confluence were used for proteomic analysis. For total cellular protein analysis, cells were washed in chilled 0.5×PBS and scraped in 2-DE lysis buffer containing 4% w/v CHAPS, 7M urea, 2M thiourea, 10 mM Tris-HCl, pH8.3, 1 mM EDTA. Lysates were homogenized by passage through a 25-gauge needle 10 times, insoluble material was removed by centrifugation at 13000 rpm for 30 min at 4° C., and protein concentrations were determined using Coomassie Protein Assay Reagent (BioRad). For secreted protein analysis, approximately 1.25×108 cells were seeded into twenty-five 175-cm2 cell culture plates for each cell line. After 2 days of incubation, the DMEM or DMEM/F-12 media were discarded, and the cells were rinsed three times with PBS. Subsequently, 375 ml of serum-free DMEM or DMEM/F-12 media were added for an additional 30 hours. The media were collected and filtered with 0.45 μm microfilters to remove cell debris and then concentrated 1000 fold with 10-kDa molecular mass cutoff concentrators (Millipore). The concentrated media were then precipitated by adding 1 volume of 100% TCA (at −20° C.) to 4 volumes of sample and incubated for 10 min. at 4° C. The precipitated protein was then recovered by centrifugation at 13000 rpm for 10 min., and the resulting pellet was washed twice with ice-cold acetone. Air-dried pellets were resuspended in 2-DE lysis buffer for protein quantification.


EXAMPLE 2
2D-DIGE and Gel Image Analysis

Before performing 2D-DIGE, protein samples were labeled with N-hydroxy succinimidyl ester-derivatives of the cyanine dyes Cy2, Cy3 and Cy5. Briefly, 150 μg of protein sample was minimally labeled with 375 pmol of either Cy3 or Cy5 for comparison on the same 2-DE. To facilitate image matching and cross-gel statistical comparison, a pool of all samples was also prepared and labeled with Cy2 at a molar ratio of 2.5 pmol Cy2 per μg of protein as an internal standard for all gels. Thus, the triplicate samples and the internal standard could be run and quantify on multiple 2-DE. The labeling reactions were performed in the dark on ice for 30 min and then quenched with a 20-fold molar ratio excess of free L-lysine to dye for 10 min. The differentially Cy3- and Cy5-labeled samples were then mixed with the Cy2-labeled internal standard and reduced with dithiothreitol for 10 min. IPG buffer, pH3-10 nonlinear (2% (v/v), GE Healthcare) was added and the final volume was adjusted to 450 μl with 2D-lysis buffer for rehydration. The rehydration process was performed with immobilized non-linear pH gradient (IPG) strips (pH3-10, 24 cm) which were later rehydrated by CyDye-labeled samples in the dark at room temperature overnight (at least 12 hours). Isoelectric focusing was then performed using a Multiphor II apparatus (GE Healthcare) for a total of 62.5 kV-h at 20° C. Strips were equilibrated in 6M urea, 30% (v/v) glycerol, 1% SDS (w/v), 100 mM Tris-HCl (pH8.8), 65 mM dithiothreitol for 15 min and then in the same buffer containing 240 mM iodoacetamide for another 15 min. The equilibrated IPG strips were transferred onto 26×20-cm 12.5% polyacrylamide gels casted between low fluorescent glass plates. The strips were overlaid with 0.5% (w/v) low melting point agarose in a running buffer containing bromophenol blue. The gels were run in an Ettan Twelve gel tank (GE Healthcare) at 4 Watt per gel at 10° C. until the dye front had completely run off the bottom of the gels. Afterward, the fluorescence 2-DE was scanned directly between the low fluorescent glass plates using an Ettan DIGE Imager (GE Healthcare). This imager is a charge-coupled device-based instrument that enables scanning at different wavelengths for Cy2-, Cy3-, and Cy5-labeled samples. Gel analysis was performed using DeCyder 2-D Differential Analysis Software v7.0 (GE Healthcare) to co-detect, normalize and quantify the protein features in the images. Features detected from non-protein sources (e.g. dust particles and dirty backgrounds) were filtered out. Spots displaying a ≧1.5 average-fold increase or decrease in abundance with a p-value <0.05 were selected for protein identification.


Protein Staining

Colloidal coomassie blue G-250 staining was used to visualize CyDye-labeled protein features in 2-DE. Bonded gels were fixed in 30% v/v ethanol, 2% v/v phosphoric acid overnight, washed three times (30 min each) with ddH2O and then incubated in 34% v/v methanol, 17% w/v ammonium sulphate, 3% v/v phosphoric acid for 1 hr., prior to adding 0.5 g/liter coomassie blue G-250. The gels were then left to stain for 5-7 days. No destaining step was required. The stained gels were then imaged on an ImageScanner III densitometer (GE Healthcare).


In-Gel Digestion

Excised post-stained gel pieces were washed three times in 50% acetonitrile, dried in a SpeedVac for 20 min., reduced with 10 mM dithiothreitol in 5 mM ammonium bicarbonate pH 8.0 (Ammonium bicarbonate) for 45 minutes at 50° C. and then alkylated with 50 mM iodoacetamide in 5 mM Ammonium bicarbonate for 1 hr. at room temperature in the dark. The gel pieces were then washed three times in 50% acetonitrile and vacuum-dried before reswelling with 50 ng of modified trypsin (Promega) in 5 mM Ammonium bicarbonate. The pieces were then overlaid with 10 μl of 5 mM Ammonium bicarbonate and trypsinized for 16 hr at 37° C. Supernatants were collected, peptides were further extracted twice with 5% trifluoroacetic acid in 50% acetonitrile and the supernatants were pooled. Peptide extracts were vacuum-dried, resuspended in 5 μl ddH2O, and stored at −20° C. prior to MS analysis.


EXAMPLE 3
Protein Identification by MALDI-TOF MS

Extracted proteins were cleaved with a proteolytic enzyme to generate peptides, then a peptide mass fingerprinting (PMF) database search following MALDI TOF mass analysis was employed for protein identification. Briefly, 0.5 μl of tryptic digested protein sample was first mixed with 0.5 μl of a matrix solution containing α-cyano-4-hydroxycinammic acid at a concentration of 1 mg in 1 ml of 50% acetonitrile (v/v)/0.1% trifluoroacetic acid (v/v), spotted onto an anchorchip target plate (Bruker Daltonics) and dried. The peptide mass fingerprints were acquired using an Autoflex III mass spectrometer (Bruker Daltonics) in reflector mode. The algorithm used for spectrum annotation was SNAP (Sophisticated Numerical Annotation Procedure). This process used the following detailed metrics: Peak detection algorithm: SNAP; Signal to noise threshold: 25; Relative intensity threshold: 0%; Minimum intensity threshold: 0; Maximal number of peaks: 50; Quality factor threshold: 1000; SNAP average composition: Averaging; Baseline subtraction: Median; Flatness: 0.8; MedianLevel: 0.5. The spectrometer was also calibrated with a peptide calibration standard (Bruker Daltonics) and internal calibration was performed using trypsin autolysis peaks at m/z 842.51 and m/z 2211.10. Peaks in the mass range of m/z 800-3000 were used to generate a peptide mass fingerprint that was searched against the Swiss-Prot/TrEMBL database (v57.12) with 513877 entries using Mascot software v2.2.06 (Matrix Science, London, UK). The following parameters were used for the search: Homo sapiens; tryptic digest with a maximum of 1 missed cleavage; carbamidomethylation of cysteine, partial protein N-terminal acetylation, partial methionine oxidation and partial modification of glutamine to pyroglutamate and a mass tolerance of 50 ppm. Identification was accepted based on significant MASCOT Mowse scores (p<0.05), spectrum annotation and observed versus expected molecular weight and pI on 2-DE.


EXAMPLE 4
Immunoassay

Immunoblotting was used to validate the differential expression of mass spectrometry identified proteins. Cells were lysed with a lysis buffer containing 50 mM HEPES pH 7.4, 150 mM NaCl, 1% NP40, 1 mM EDTA, 2 mM sodium orthovanadate, 100 μg/mL AEBSF, 17 μg/mL aprotinin, 1 μg/mL leupeptin, 1 μg/mL pepstatin, 5 μM fenvalerate, 5 μM BpVphen and 1 μM okadaic acid prior to protein quantification with Coomassie Protein Assay Reagent (BioRad).


30 μg of protein samples were diluted in Laemmli sample buffer (final concentrations: 50 mM Tris pH 6.8, 10% (v/v) glycerol, 2% SDS (w/v), 0.01% (w/v) bromophenol blue) and separated by 1D-SDS-PAGE following standard procedures. After electroblotting separated proteins onto 0.45 μm Immobilon P membranes (Millipore), the membranes were blocked with 5% w/v skim milk in TBST (50 mM Tris pH 8.0, 150 mM NaCl and 0.1% Tween-20 (v/v)) for 1 hour. Membranes were then incubated in primary antibody solution in TBS-T containing 0.02% (w/v) sodium azide for 2 hours. Membranes were washed in TBS-T (3minutes, 10 times) and then probed with the appropriate horseradish peroxidase-coupled secondary antibody (GE Healthcare). After further washing in TBS-T, immunoprobed proteins were visualized using an enhanced chemiluminescence method (Visual Protein Co.).


For immunofluorescence staining, cells were plated onto coverslips (VWR international) for overnight incubation. The cells were fixed with PBS containing 4% (v/v) paraformaldehyde for 25 minutes, washed three times with PBS, and followed by permeabilization in PBS containing 0.2% (v/v) Triton X-100 for 10 minutes. Coverslips were rinsed and blocked in PBS containing 5% (w/v) BSA for 10 minutes before incubation with primary antibodies diluted in 2.5% BSA/PBS for 1 hour. After three washings with PBS, samples were incubated with the appropriate fluorescently labelled secondary antibodies diluted in 2.5% BSA/PBS for 1 hour. Coverslips were then washed three times with PBS and at least twice with ddH2O before mounting in Vectashield mounting medium (Vector Lab). Coverslip edges were sealed with nail polish onto glass slides (BDH) and then dried in the dark at 4° C. For image analysis, cells were imaged using a Zeiss Axiovert 200M fluorescent microscope (Carl Zeiss Inc., Germany). The laser intensities used to detect the same immunostained markers from different cell lines were identical, and none of the laser intensities used to capture images was saturated.


EXAMPLE 5
Optimization of Cell Conditions for Secreted Protein Analysis

For secretomic analysis, MCF-10A, MCF-7 and MDA-MB-231 were grown on cell culture dishes and the confluency of cells was checked prior to incubation in serum-free culture media to ensure that no other exogenous proteins were present. To minimize cell autolysis induced by starvation and to maximize secreted protein concentration in the media, the starvation time of each cell line was optimized. Through immunoblotting, the LDH and β-tubulin levels were detected in the 1000-fold concentrated serum-free media starting at 48˜60 hours and at 60˜72 hours, respectively (FIG. 1). LDH and β-tubulin are both cytoplasmic proteins and their levels in the media represent the amount of cell death taking place in cell culture.


Accordingly, a starvation period of 30 hours was chosen for further 2D-DIGE based secretomic analysis.


EXAMPLE 6

DIGE and MALDI-TOF Analysis of Secretomes among MCF-10A, MCF-7 and MDA-MB-231 Cells


Proteins secreted from each cell type were enriched from the serum-free medium followed by labeling with CyDyes for 2D-DIGE analysis. The secretomic profiling of MCF-10A, MCF-7 and MDA-MB-231 were visualized using a fluorescence scanner and the images were superimposed using ImageQuant software (FIG. 2). To investigate the potential involvement of secreted proteins in tumorigenesis and metastasis for human breast cancer, biological variation analysis of spots showing greater than 1.5-fold change in expression with a t-test score of P value<0.05 were visually checked before confirming the alterations for protein identification. MALDI-TOF MS identification revealed 50 unique differentially expressed proteins across MCF-10A, MCF-7 and MDA-MB-231 (Table 1). Of the proteins identified, 42 were differentially expressed between MCF-7/MCF-10A, 44 of them were differentially expressed between MDA-MB-231/MCF-10A, and 37 proteins were differentially expressed between MDA-MB-231 and MCF-7. In the three cell lines investigated, 39% of the total proteins identified were extracellular and plasma membrane-anchored proteins (FIG. 3A). Most of the identified proteins were involved in signaling transduction, redox-regulation and metabolism (FIG. 3B). To our knowledge, 14 out of these identified spots, including IFIT3, have not been reported in any breast cancer related studies. Consequently, these proteins have the potential to be breast cancer markers. As expected, this 2D-DIGE experiment also identified a number of reported breast cancer markers, including Cathepsin D (Zhang, Y. G; D U, J.; Tian, X. X.; Zhong, Y. F.; Fang, W. G. Chin Med. J. (Engl.) 2007, 120, 1597) and IGFBP4 (Mita, K.; Zhang, Z.; Ando, Y.; Toyama, T.; Hamaguchi, M.; Kobayashi, S.; Hayashi, S.; Fujii, Y.; Iwase, H.; Yamashita, H. Jpn. J. Clin. Oncol. 2007, 37, 575).


Using the LC-MS/MS strategy, Kulasingam and Diamandis analyzed and compared the expressions of extracellular and membrane-bound proteins in conditioned media of three breast cells corresponding to the normal control cells and cell lines derived from stage 2 and stage 4 patients, respectively (Kulasingam, V.; Diamandis, E. P. Mol. Cell Proteomics. 2007, 6, 1997). Kulasingam's experiment identified 1062 differentially expressed proteins across these three cell lines. A comparison between Kulasingam's result and 2D-DIGE secretomic of the present invention shows that 25 out of 50 identified differentially expressed secreted proteins coincide with Kulasingam's study, indicating that both LC-MS/MS and 2D-DIGE are potential tools for discovering breast cancer markers with reasonable reproducibility. Importantly, 25 out of 50 identified proteins were not reported in Kulasingam's study or any other studies, demonstrating that 2D-DIGE plays a powerful complementary role in the assumed biomarker discovery (Table 1a & 1b).









TABLE 1a







Alphabetical list of identified differentially expressed secreted


proteins of breast cells MCF-10A, MCF-7, and MDA-MB-231.


















Putative
Reported by


Accession

MCF7/
MDA231/
MDA231/
Breast
Kulasingam


Code
Protein name
MCF10A#
MCF10A#
MCF7#
Markers##
et al###
















P63104
14-3-3 protein zeta/delta
−2.25
−4.28
−1.85

K


O95861
3′(2′),5′-bisphosphate nucleotidase 1
1.16
2.89
2.56


O95336
6-phosphogluconolactonase
1.05
1.95
1.91

K


P60709
Actin
2.57
1.17
−2.14

K


P60709
Actin
2.21
−1.04
−2.25

K


P15121
Aldose reductase
3.14
8.14
2.66

K


P06733
Alpha-enolase
−2.57
1.86
4.94

K


P08758
Annexin A5
−1.03
3.28
3.47

K


Q8N1M1
Bestrophin-3/BEST3
3.6
2.62
−1.34
A


P00918
Carbonic anhydrase 2
−4.72
−6.11
−1.25
B


P46527
Cyclin-dependent kinase inhibitor
1.65
3.41
2.07



1B/Cyclin-dependent kinase



inhibitor p27/p27Kip1


Q9C0G6
Dynein heavy chain 6
5.07
2.87
−1.71
A


Q9NPC3
E3 ubiquitin-protein ligase
4.92
5.8
1.22



CCNB1IP1


Q93070
Ecto-ADP-ribosyltransferase 4/
1.75
1.92
1.14
B



CD297/ART4


P04075
Fructose-bisphosphate aldolase A
1.89
1.15
−1.61

K


P21266
Glutathione S-transferase Mu 3
4.18
−1.09
−4.46


P09211
Glutathione S-transferase P
−5.67
−4.47
1.3

K


P09211
Glutathione S-transferase P
−13.19
−13.28
1.02

K


P04406
Glyceraldehyde-3-phosphate
−1.71
−1.12
1.57

K



dehydrogenase


Q9HC38
Glyoxalase domain-containing
−1.19
1.71
2.09



protein 4


Q8IUY3
GRAM domain-containing protein
2.33
2.85
1.25
A



2/GRAMD2


Q8IUY3
GRAM domain-containing protein
6.8
6.97
1.06
A



2/GRAMD2


Q8IUY3
GRAM domain-containing protein
3.81
3.47
−1.06
A



2/GRAMD2


P04792
Heat shock protein beta-1/HSP27
3.43
−1.5
−5.02

K


P04792
Heat shock protein beta-1/HSP27
2.39
−2.82
−6.57

K


P29218
Inositol monophosphatase
−2.41
1.1
2.72

K


P22692
Insulin-like growth factor-binding
6.64
29.51
4.56

K



protein 4/IGFBP4


P22692
Insulin-like growth factor-binding
8.64
25.19
2.99

K



protein 4/IGFBP4


O14879
Interferon-induced protein with
−9.31
−7.91
1.22
A



tetratricopeptide repeats 3/IFIT3/



ISG60


P30740
Leukocyte elastase inhibitor/
−4.73
−3.25
1.49

K



Serpin B1


Q14168
MAGUK p55 subfamily member
2.66
13.13
5.06



2/MPP2


Q14168
MAGUK p55 subfamily member
1.31
2.53
1.98



2/MPP2


P35240
Merlin/Neurofibromin-2
4.77
1.54
−3.03


P26038
Moesin
−3
1.95
6.04

K


O75380
NADH dehydrogenase
2.09
2.16
1.07



[ubiquinone] iron-sulfur protein 6


P62937
Peptidyl-prolyl cis-trans isomerase
−1.88
−1.2
1.61

K



A/cyclophilin A


Q06830
Peroxiredoxin-1
2.11
1.29
−1.6

K


P32119
Peroxiredoxin-2
2.06
1.84
−1.09

K


P30086
Phosphatidylethanolamine-binding
1.52
1.26
−1.17

K



protein 1/PEBP1/Raf kinase



inhibitor protein


P18669
Phosphoglycerate mutase 1
−1.2
4.81
5.9
B


Q01814
Plasma membrane
−2.96
−6.05
−1.99



calcium-transporting ATPase 2/



PMCA2/ATP2B2/Plasma



membrane calcium ATPase



isoform 2


P05121
Plasminogen activator inhibitor 1/
−10.93
−8.8
1.29

K



PAI1/SERPINE1


P25786
Proteasome subunit alpha type-1
−1.05
1.56
1.68
A


P25787
Proteasome subunit alpha type-2
−1.49
1.05
1.61

K


P49720
Proteasome subunit beta type-3
−1.45
1.03
1.52
A


P31150
Rab GDP dissociation inhibitor
12.45
7.78
−1.55

K



alpha/GDI1


P50395
Rab GDP dissociation inhibitor
1.21
1.68
1.44

K



beta/GDI2


Q9H5N1
Rab GTPase-binding effector
−15.11
−8.81
1.78
A



protein 2


Q8WUD1
Ras-related protein Rab-2B
1.2
−1.77
−2.07
A


Q13228
Selenium-binding protein 1/
1.38
−1.52
−2.03
B



SELENBP1


P36952
Serpin B5
−16.8
−21.59
−1.25


P04179
Superoxide dismutase [Mn],
−4.36
−1.72
2.6



mitochondrial


P55072
Transitional endoplasmic
−1.31
1.36
1.85

K



reticulum ATPase/



Valosin-containing protein/VCP


Q8N912
Transmembrane protein
1.88
1.91
1.05
A



C14orf180


Q9P1Q0
Vacuolar protein
−18.84
−25.44
−1.31
A



sorting-associated protein 54/



Hepatocellular carcinoma protein



8/Tumor antigen SLP-8p/



VPS54


O75083
WD repeat-containing protein 1/
−1.26
1.76
2.3

K



Actin-interacting protein 1






#The everage ratio of differentially expressed (p < 0.05) proteins after 2D-DIGE analysis across MCF-10A, MCF-7, and MDA-MB-231 were calculated considering 3 replica gels.




##Identified protiens which have not been reported in any cancer research are marked “A”, while proteins which have been reported in cancer research but not in breast cancer research are marked “B”.




###Poreins in this list have been reported in Kulasing et al's experiment.














TABLE 1b







Alphabetical list of identified differentially expressed secreted


proteins obtained after MALTI-TOF mass spectrometry analysis.














Accession


No. Match.
seq cov

Subcellular



code
pI
MW
Peptides
(%)
Score
location*
functional class*

















P63104
4.73
27899
17/43 
52
137/56 
Cytoplasm
Siganl transduction


O95861
5.46
33713
8/12
27
76/56
Cytoplasm
Biosynthesis


O95336
5.7
27815
9/44
41
60/56
Cytoplasm
Metabolism


P60709
5.29
42052
7/12
23
65/56
Cytoplasm
Cytoskeleton


P60709
5.29
42052
7/15
22
65/56
Cytoplasm
Cytoskeleton


P15121
6.51
36230
7/24
25
57/56
Cytoplasm
Metabolism


P06733
7.01
47481
10/20 
26
70/56
Cytoplasm
Metabolism


P08758
4.94
35971
10/17 
26
77/56
Plasma membrane
Signal transduction/Ca









regulation


Q8N1M1
6.13
76457
9/22
14
66/56
Plasma membrane
Transport/channel


P00918
6.87
29285
7/31
29
62/56
Cytoplasm
Metabolism


P46527
6.54
22288
6/28
32
83/56
Nucleus
Cell cycle


Q9C0G6
5.72
479761
19/31 
5
66/56
Cytoplasm
Cytoskeleton


Q9NPC3
8.59
31923
6/23
24
60/56
Nucleus
Cell cycle


Q93070
9.31
36197
5/49
15
57/56
Plasma membrane
Biosynthesis


P04075
8.3
39851
15/35 
47
103/56 
Cytoplasm
Metabolism


P21266
5.37
26998
7/11
25
58/56
Cytoplasm
Redox regulation


P09211
5.43
23569
6/15
38
56/56
Cytoplasm
Redox regulation


P09211
5.43
23569
7/15
41
73/56
Cytoplasm
Redox regulation


P04406
8.57
36201
6/17
20
59/56
Mito
Metabolism


Q9HC38
5.4
35170
6/15
20
58/56
Mito
Redox regulation


Q8IUY3
8.73
40908
6/26
13
60/56
Plasma membrane
Unknown


Q8IUY3
8.73
40908
9/56
25
63/56
Plasma membrane
Unknown


Q8IUY3
8.73
40908
7/29
19
61/56
Plasma membrane
Unknown


P04792
5.98
22826
4/26
25
69/56
Cytoplasm
Protein folding


P04792
5.98
22826
6/14
30
72/56
Cytoplasm
Protein folding


P29218
5.16
30568
6/32
23
64/56
Cytoplasm
Biosynthesis


P22692
6.81
29113
10/42 
37
123/56 
Secreted
Siganl transduction


P22692
6.81
29113
8/23
28
105/56 
Secreted
Siganl transduction


O14879
5.12
56691
6/16
16
66/56
Plasma membrane
Siganl transduction


P30740
5.9
42829
14/34 
33
126/56 
Cytoplasm
Protease inhibitor


Q14168
6.28
64887
7/24
14
60/56
Plasma membrane
Siganl transduction


Q14168
6.28
64887
7/24
14
57/56
Plasma membrane
Siganl transduction


P35240
6.11
69874
10/29 
15
70/56
Plasma membrane
Cell motility/signal









transduction


P26038
6.08
67892
14/34 
19
96/56
Plasma membrane
Cell motility/









cytoskeleton


O75380
8.59
14045
6/52
33
58/56
Mito
Electron transport


P62937
7.68
18229
10/25 
64
112/56 
Cytoplasm
Protein folding


Q06830
8.27
22324
13/33 
56
139/56 
Cytoplasm
Redox Regulation


P32119
5.66
22049
6/24
35
75/56
Cytoplasm
Redox Regulation


P30086
7.01
21158
8/33
54
97/56
Cytoplasm
Siganl transduction


P18669
6.67
28900
8/27
35
70/56
Cytoplasm
Metabolism


Q01814
5.66
137987
7/16
7
71/56
Plasma membrane
Transport


P05121
6.68
45088
8/14
14
72/56
Secreted
Protease inhibitor


P25786
6.15
29822
6/26
23
58/56
Proteasome
Proteins degradation


P25787
6.92
255996
6/22
33
70/56
Proteasome
Proteins degradation


P49720
6.14
23219
7/20
38
67/56
Proteasome
Proteins degradation


P31150
5
51177
8/32
25
65/56
Plasma membrane
Siganl transduction


P50395
6.11
51087
5/38
18
58/56
Plasma membrane
Siganl transduction


Q9H5N1
4.76
63690
8/26
13
62/56
Plasma membrane
Membrane trafficking


Q8WUD1
7.68
24427
5/20
25
68/56
Plasma membrane
Siganl transduction


Q13228
5.93
52928
11/23 
20
94/56
Plasma membrane
Transport


P36952
5.72
42568
6/22
18
61/56
Secreted
Protease inhibitor


P04179
8.35
24878
6/27
30
65/56
Mito
Redox Regulation


P55072
5.14
899550
25/41 
34
197/56 
Cytoplasma
Vascular transport


Q8N912
11.2
18382
4/38
25
58/56
Plasma membrane
Unknown


Q9P1Q0
6.1
111545
7/24
11
64/56
Endosome
Vascular transport


O75083
6.17
66836
18/32 
40
197/56 
Cytoplasm
Cytoskeleton





*The subcellular locations and functional classes of identified proteins were obtained from the Uniprot website.






EXAMPLE 7
DIGE and MALDI-TOF Analyses of the Total Cell Proteomes Among MCF-10A, MCF-7 and MDA-MB-231 Cells

To identify the altered abundances of proteins and relate them to the tumorigenesis of breast cancer, the proteomic profiles of MCF-10A, MCF-7 and MDA-MB-231 were analyzed. Triplicates of the three different cell lysates were compared using 2D-DIGE to obtain an overview of breast cell tumorigenesis. Image analysis using DeCyder v7.0 clearly defined more than 2500 protein spots (FIG. 4). To reduce the intrinsic variability derived from protein samples and gel-to-gel variation, only those protein spots that appeared in all of the triplicate gel images were used for statistical analysis.


Furthermore, biological variation analysis of spots showing greater than 1.5-fold change in expression with a t-test score of less than 0.05 were visually checked before confirming the alterations for protein identification. MALDI-TOF MS identification revealed 133 unique differentially expressed proteins across MCF-10A, MCF-7, and MDA-MB-231 (Table 2). Of the 133 proteins identified, 107 of them had differential expressions between MCF-7/MCF-10A, 63 were differentially expressed between MDA-MB-231/MCF-10A and 96 had differential expressions between MDA-MB-231 and MCF-7. Almost half of the total proteins identified in this breast cell model were cytosolic proteins (FIG. 5A), and most of the identified proteins were involved in signaling transduction, metabolism, protein folding, and cell motility (FIG. 5B). According to the comparison table, 51 of these identified spots, including Calumenin, have not been reported in any breast cancer related studies. As such, these proteins might have the potential to be putative breast cancer markers. As expected, some well-known breast cancer markers, such as 14-3-3 proteins, annexins, calmodulin, AGR-2, Galectin-1 and ROCK2, were also identified in this 2D-DIGE experiment, lending credence to the reliability of early phase biomarker detection using this experimental strategy. In a previous study, Nagaraja et.al. used traditional 2-DE with post-stains (silver stain and coomassie blue stain) to reveal 26 differentially expressed proteins among transformed breast cells with different levels of invasiveness and normal cells which were the same cell lines used in the present study (Nagaraja, G. M.; Othman, M.; Fox, B. P.; Alsaber, R.; Pellegrino, C. M.; Zeng, Y.; Khanna, R.; Tamburini, P.; Swaroop, A.; Kandpal, R. P. Oncogene 2006, 25, 2328). Their study showed no evidence of visualizing protein spots with sensitive strategies, and protein expression changes were not quantifiable because no broader linear-ranged methods and statistical analysis were employed. Only 6 out of those 26 proteins coincide with statistical 2D-DIGE data of the present invention, which implies that differences might have derived from artificial variations or from results with no statistical analysis (Table 2).









TABLE 2







Alphabetical list of identified differentially expressed total


cellular proteins across MCF-10A, MCF-7 and MDA-MB-231 breast


cells.



















Reported







Putative
by


Accession

MCF7/
MDA231/
MDA231/
Breast
Nagaraja


Code
Protein name
MCF10A#
MCF10A#
MCF7#
Markers##
et al###





P62258
14-3-3 protein epsilon
−3.37
1.3
4.55


P63104
14-3-3 protein zeta/delta
−1.95
1.13
2.26


P61254
60S ribosomal protein L26
9.72
−2.69
−22.09


P11021
78 kDa glucose-regulated protein
1.73
−1.63
−2.59


Q6XD76
Achaete-scute homolog 4/ASCL4
1.18
1.74
1.47
B


P21399
Aconitate hydratase
1.24
1.62
1.31
A


Q9Y6K8
Adenylate kinase isoenzyme 5
−2.07
−1.07
2.01


O95994
AGR2/Anterior gradient protein 2 homolog
31.03
1.75
−15


P15121
Aldose reductase
−1.18
2.3
2.73


P06733
Alpha-enolase
1.3
1.62
1.24


Q9H4A4
Aminopeptidase B
−1.56
−2.43
−1.55
B


P04083
Annexin A1
−3.96
−1.03
3.97


P07355
Annexin A2
−5.67
−2.37
2.47


P07355
Annexin A2
−8.1
−3.56
2.35


P12429
Annexin A3
1.61
−3.86
−6.21
B


P09525
Annexin A4
−1.39
−3.91
−2.82


P09525
Annexin A4
−1.1
−4.59
−4.19


P08758
Annexin A5
−2.84
1.65
4.83


P08758
Annexin A5
−1.81
−1.11
1.63


P08133
Annexin A6
2.28
1.39
−1.64


P14868
Aspartyl-tRNA synthetase
−3.18
−3.65
−1.15


P06576
ATP synthase subunit beta
−1.26
−1.66
−1.32


O75531
Barrier-to-autointegration factor/Breakpoint
3.09
−3.18
−9.49
A



cluster region protein 1


P31939
Bifunctional purine biosynthesis protein PURH/
2.37
1.14
−2.08
A



IMP cyclohydrolase/ATIC


P62158
Calmodulin
2.92
−2.15
−6.08


P62158
Calmodulin
3.85
−2.6
−9.67


P04632
Calpain small subunit 1/Capn4/
−1.59
−1.55
1.02



Calcium-dependent protease small subunit/



Calpain regulatory subunit


P27797
Calreticulin
2.16
−1.36
−2.84

N


O43852
Calumenin
2.03
−1.16
−2.26
B


P00918
Carbonic anhydrase 2
−1.54
−9.13
−5.94
B


P07339
Cathepsin D
8.45
−2.1
−17.7


P07339
Cathepsin D
9.91
−9.22
−40.11


P07339
Cathepsin D
5.19
−1.48
−53.69


P07339
Cathepsin D
7.8
−5.32
−88.32


P29373
Cellular retinoic acid-binding protein 2/
2.37
−5.13
−12.14



CRABP2


O00299
Chloride intracellular channel protein 1/CLIC1
−1.14
1.6
1.82


O00299
Chloride intracellular channel protein 1/CLIC1
−1.13
1.37
1.55


Q9Y696
Chloride intracellular channel protein 4/CLIC4
−5.72
−2.25
2.54


Q9Y696
Chloride intracellular channel protein 4/CLIC4
−4.09
−1.87
2.19


O75390
Citrate synthase
−2.09
−2.68
−1.24


O14579
Coatomer subunit epsilon (COPE)
2.17
1.13
−1.86


P23528
Cofilin-1
1.33
−3.8
−5.06


A6NKD9
Coiled-coil domain-containing protein/
5.12
6.54
1.28
A



CCDC85C


Q14993
Collagen alpha-1(XIX) chain/Collagen alpha-1
3.23
3.84
1.23


Q15828
Cystatin-M
2.34
1.76
−1.33


Q04695
Cytokeratin 1
1.54
1.34
−1.15


P19012
Cytokeratin 15
−6.26
−11.21
−1.73


P05787
Cytokeratin 8
17.75
1.67
−10.66


Q9UBS4
DnaJ homolog subfamily B member 11
2.28
−1.06
−2.34


P29692
Elongation factor 1-delta
1.29
1.56
1.21


P26641
Elongation factor 1-gamma
1.74
1.85
1.06


P13639
Elongation factor 2
1.62
1.11
−1.46


P30040
Endoplasmic reticulum protein ERp29
4.31
1.23
−2.92


P30040
Endoplasmic reticulum protein ERp29
4.89
1.62
−3.39


P14625
Endoplasmin
2.41
1.08
−1.89


O94905
Erlin-2/SPFH 2
1.94
−1.53
−2.87
A


P63241
Eukaryotic translation initiation factor 5A-1
−1.69
1.28
2.53


P63241
Eukaryotic translation initiation factor 5A-1
−2.54
−1.04
2.23


P47756
F-actin-capping protein subunit beta
1.53
−1.38
−2.1
B


P47756
F-actin-capping protein subunit beta
1.2
−1.81
−2.18
B


Q02790
FK506-binding protein 4/Peptidyl-prolyl
2.83
−1.07
−3.03



cis-trans isomerase/FKBP52


P30043
Flavin reductase
11.67
5.46
−2.14
B


P09467
Fructose-1,6-bisphosphatase 1
4.57
1.19
−3.83
A


P09467
Fructose-1,6-bisphosphatase 1
12.62
−1.12
−14.19
A


P04075
Fructose-bisphosphate aldolase A
1.66
−1.31
−2.18
B


P04075
Fructose-bisphosphate aldolase A
1.8
−1.29
−2.32
B


P09382
Galectin-1
5.99
−1.18
−6.83

N


P11413
Glucose-6-phosphate 1-dehydrogenase/G6PD
4.39
−1.05
−4.62


P14314
Glucosidase 2 subunit beta/PRKCSH/80K-H
2.91
−1.05
−2.96



protein


P21266
Glutathione S-transferase Mu 3
16.67
2.54
−6.57


P21266
Glutathione S-transferase Mu3
46.76
2.56
−18.29


P09211
Glutathione S-transferase P
−27.61
−11.61
2.38


P09211
Glutathione S-transferase P
−17.35
−8.71
2.06


P62993
Growth factor receptor-bound protein 2 (Grb2)
3.19
1.63
−1.96


Q12931
Heat shock protein 75 kDa/TNFR-associated
1.83
−1.09
−1.99
B



protein 1/TRAP1


P04792
Heat shock protein beta-1/HSP 27
4.22
−4.51
−19.01

N


P04792
Heat shock protein beta-1/HSP 27
7.97
−3.26
−25.98

N


P04792
Heat shock protein beta-1/HSP 27
7.62
−3.7
−28.21

N


P68871
Hemoglobin subunit beta
2.9
2.44
−1.15


P61978
Heterogeneous nuclear ribonucleoprotein K
−2.66
−2.36
1.17


P22626
Heterogeneous nuclear ribonucleoproteins
−2.74
−2.52
1.13
B



A2/B1


Q13547
Histone deacetylase 1
12.52
11.23
−1.08


Q9Y4L1
Hypoxia up-regulated protein 1
2.05
−1.12
−1.94


Q04760
Lactoylglutathione lyase/Glyoxalase 1/
1.52
1.64
1.08



Aldoketomutase


P09960
Leukotriene A-4 hydrolase/LTA4H
−1.6
−1.28
1.25
B


P07195
L-lactate dehydrogenase B chain
−7.5
−1.09
6.89


P07195
L-lactate dehydrogenase B chain
−3.25
−1.09
2.98


P40926
Malate dehydrogenase
1.32
−1.92
−2.53


O00264
Membrane-associated progesterone receptor
3.27
−1.43
−4.51



component 1/PGRMC1


O75121
Microfibrillar-associated protein 3 like/
−1.58
9.38
14.78
B



MFAP3L


Q15691
Microtubule-associated protein RP/EB family
−1.36
1.44
1.97
B



member 1 (End-binding protein 1)


P26038
Moesin
−2.33
3.51
8.46


P60660
Myosin light polypeptide 6
5.49
−1.79
−9.48


O14950
Myosin regulatory light chain MRLC2
1.84
1.8
1.16


P19105
Myosin regulatory light chain MRLC3
−3.9
−1.93
2.09


P30084
Myosin-IXa
−1.03
−2.08
−1.95


Q14697
Neutral alpha-glucosidase AB/Glucosidase II
2.34
−1.22
−2.32



subunit alpha


Q14697
Neutral alpha-glucosidase AB/Glucosidase II
1.87
−1.28
−2.75



subunit alpha


Q9NXR1
Nuclear distribution protein nudE homolog 1/
7.78
6.44
−1.17
A



NDE1


P15531
Nucleoside diphosphate kinase A/NDP kinase A
−1.85
1.07
2.04

N


P20472
Parvalbumin alpha
16.77
10.64
−1.52
B


O00151
PDZ and LIM domain protein 1/LIM domain
−3.96
−1.46
2.8
B



protein CLP-36/Elfin/CLP36


P62937
Peptidyl-prolyl cis-trans isomerase A/
−1.34
−1.65
−1.24

N



cyclophilin A


P62937
Peptidyl-prolyl cis-trans isomerase A/
−1.41
−1.76
−1.25

N



cyclophilin A


Q96BP3
Peptidylprolyl isomerase domain and WD
−4.68
−3.06
1.58
A



repeat-containing protein 1


Q13162
Peroxiredoxin-4
1.74
1.19
−1.46


Q13162
Peroxiredoxin-4
4.92
−1.11
−5.29


P30041
Peroxiredoxin-6
1.4
−1.23
−1.73


P30041
Peroxiredoxin-6
1.09
−1.67
−1.82


Q9Y617
Phosphoserine aminotransferase/PSAT
−5.37
−1.26
4.25
B


P13797
Plastin-3/T plastin
−3.88
−1.67
2.32
A


P07737
Profilin-1
−1.35
1.2
1.62


P07737
Profilin-1
−1.29
1.16
1.5


Q8WUM4
Programmed cell death 6-interacting protein/
1.88
−1.07
−2.02
B



ALG-2-interacting protein 1/Hp95/PDCD6IP


P35232
Prohibitin
1.08
1.63
1.51


Q15185
Prostaglandin E synthase 3/PGES
1.63
1.18
−1.38


Q06323
Proteasome activator complex subunit 1
1.73
−1.3
−2.24
A


Q9UL46
Proteasome activator complex subunit 2
1.36
−1.22
−1.66
B


Q9UL46
Proteasome activator complex subunit 2
1.58
−1.25
−1.98
B



(PSME2)


P61289
Proteasome activator complex subunit 3
−1.11
1.48
1.64



(PSME3)


Q9Y2B0
Protein canopy homolog 2/CNPY2/
1.85
−3.25
−5.82
A



MIR-interacting saposin-like protein


Q6XLA1
Protein CASC2, isoform 3/Cancer
−2.97
1.09
3.34
B



susceptibility candidate gene 2 protein, isoform 3


P07237
Protein disulfide-isomerase A1/PDI
3.33
−1.75
−5.63


P30101
Protein disulfide-isomerase A3/ERp57
−2
−1.41
1.42


Q15084
Protein disulfide-isomerase A6/PDIA6/
−1.12
−1.66
−1.49
A



Protein disulfide isomerase P5


Q9HCY8
Protein S100A14
−5.11
−6.92
−1.31


Q6PI26
Protein SHQ1
−1.27
−4.27
−3.36
A


P14618
Pyruvate kinase isozymes M1/M2/Pyruvate
−1.77
−1.54
1.15



kinase 2/3/



PKM2/THBP1


P50395
Rab GDP dissociation inhibitor beta/GDI2
1.53
1.19
−1.29
B


Q8WUD1
Ras-related protein Rab-2B
−5.45
−13.37
−2.07
A


Q14257
Reticulocalbin-2/ERC-55/RCN2
1.5
−1.78
−2.59
B


O94788
Retinal dehydrogenase 2/Aldehyde
6.02
−1.12
−6.51



dehydrogenase family 1 member A2/



ALDH1A2


P42331
Rho GTPase-activating protein 25
1.54
1.11
−1.17
A


Q13017
Rho GTPase-activating protein 5/p190-B/
1.93
1.31
−1.47
B



ARHGAP5/RhoGAP 5


O75116
Rho-associated protein kinase 2/ROCK2
5.16
−1.56
−8.07


O75116
Rho-associated protein kinase 2/ROCK2
8.04
−2.56
−20.57


P13489
Ribonuclease inhibitor/RNase inhibitor
−1.54
−1.68
−1.09
A


Q13228
Selenium-binding protein 1/SELENBP1
3.04
−1.1
−3.34
B


Q13228
Selenium-binding protein 1/SELENBP1
6.76
−1.12
−7.56
B


Q9NVA2
Septin-11
2.15
−1.9
−3.98
A


Q92599
Septin-8
2.36
1.05
−2.17
A


Q8TDX7
Serine/threonine-protein kinase Nek7
−2.14
−1.53
1.66
A


Q00536
Serine/threonine-protein kinase PCTAIRE-1
4.12
−1.28
−5.28
A


P36952
Serpin B5
−10.78
−11.05
−1.02


P55854
Small ubiquitin-related modifier 3/SUMO3
−2.61
−1.83
1.48
B


P31948
Stress-induced-phosphoprotein 1/STIP1
−2.2
1.07
2.42
B


Q8NBS9
Thioredoxin domain-containing protein 5/
5.71
−1.11
−6.13
B



ERp46/TXNDC5


Q8NBS9
Thioredoxin domain-containing protein 5/
5.8
−1.53
−8.61
B



ERp46/TXNDC5


Q9UI15
Transgelin-3
5.15
2.84
−1.53


P60174
Triosephosphate isomerase/TPI
1.61
−1.05
−1.68

N


P60174
Triosephosphate isomerase/TPI
1.5
−1.45
−2.18


P67936
Tropomyosin alpha-4 chain/Tropomyosin-4
−1.72
1.38
2.46


P61088
Ubiquitin-conjugating enzyme E2/Ubc13
−2.28
1.29
3.04
A


Q5VYV7
UPF0492 protein C20orf94
−4.17
−2.6
1.66
A


P08670
Vimentin
−3.55
4.14
15.21


P08670
Vimentin
−3.77
2.65
10.32


P21796
Voltage-dependent anion-selective channel
−1.65
−2.13
−1.29
B



protein 1/VDAC1


P45880
Voltage-dependent anion-selective channel
−1.55
−1.62
−1.05
B



protein 2/VDAC2


Q2VY69
Zinc finger protein 284
−8.32
−2.7
3.19


Q8N7K0
Zinc finger protein 433
7.92
−1.02
−8.04
B


Q6ZNA1
Zinc finger protein 836
2.2
1.21
−1.53











#The everage ratio of differentially expressed (p < 0.05) proteins after 2D-DIGE analysis



across MCF-10A, MCF-7, and MDA-MB-231 were calculated considering 3 replica gels.



##Identified protiens which have not been reported in any cancer research are marked



“A”, while proteins which have been reported in cancer research but not in breast cancer


research are marked “B”.



###Poreins in this list have been reported in Nagaraja et al's experiment.








Alphabetical list of identified differentially expressed total


cellular proteins obtained after MALDI-TOF mass spectrometry


analysis.
















Accession


No. Match.
seq cov

Subcellular




Code
pI
MW
Peptides
(%)
Score
location
functional class*







P62258
4.63
29326
8
20%
67/56
Cytoplasma
Sgnal Transduction



P63104
4.73
27899
8
28%
74/56
Cytoplasma
Sgnal Transduction



P61254
10.55
17248
6
31%
56/56
Cytoplasma
Biosynthesis



P11021
5.07
72402
11
20%
98/56
ER
Biosynthesis



Q6XD76
9.23
19469
4
31%
59/56
Nucleus
Gene Regulation



P21399
6.23
98850
10
15%
92/56
Mito
TCA cycle



Q9Y6K8
5.38
22358
5
35%
63/56
Cytoplasma
Sgnal Transduction



O95994
9.03
20024
5
34%
63/56
Secreted
Unknown



P15121
6.51
36230
7
33%
82/56
Cytoplasma
Metabolism



P06733
7.01
47481
16
47%
151/56 
Cytoplasma
Metabolism



Q9H4A4
5.51
73234
10
20%
98/56
Secreted
Protein catabolism



P04083
6.57
38918
9
33%
105/56 
Cell Membrane
Signal transduction/










Ca regulation



P07355
7.57
38808
10
34%
92/56
Cell Membrane
Signal transduction/










Ca regulation



P07355
7.57
38808
14
41%
158/56 
Cell Membrane
Signal transduction/










Ca regulation



P12429
5.63
36524
16
45%
151/56 
Cytoplasma
Signal transduction/










Ca regulation



P09525
5.84
36088
6
26%
61/56
Cell Membrane
Signal transduction/










Ca regulation



P09525
5.84
36088
10
34%
109/56 
Cell Membrane
Signal transduction/










Ca regulation



P08758
4.94
35971
9
35%
127/56 
Cell Membrane
Signal transduction/










Ca regulation



P08758
4.94
35971
11
42%
142/56 
Cell Membrane
Signal transduction/










Ca regulation



P08133
5.42
76168
12
20%
104/56 
Cell Membrane
Signal transduction/










Ca regulation



P14868
6.11
57499
7
21%
61/56
Cytoplasma
Biosynthesis



P06576
5.26
56525
9
23%
98/56
Mito
Sgnal Transduction



O75531
5.81
10280
6
55%
76/61
Nucleus
Gene Regulation



P31939
6.27
65089
11
23%
78/56
Cytoplasma
Biosynthesis



P62158
4.09
16827
5
39%
81/56
Cytoplasma
Signal transduction/










Ca regulation



P62158
4.09
16827
5
39%
78/56
Cytoplasma
Signal transduction/










Ca regulation



P04632
5.05
28469
5
28%
64/56
Cytoplasma
Signal Transduction/









Cell membrane
Cytoskeleton










remodelling/Ca










regulation



P27797
4.29
48283
7
16%
71/56
ER
Signal transduction/










Ca regulation



O43852
4.47
37198
9
35%
129/56 
ER
Signal transduction/










Ca regulation



P00918
6.87
29285
6
26%
69/56
Cytoplasma
Metabolism



P07339
6.1
45037
5
14%
63/56
Lysosome
Protein degradation



P07339
6.1
45037
5
16%
59/56
Lysosome
Protein degradation



P07339
6.1
45037
6
16%
74/56
Lysosome
Protein degradation



P07339
6.1
45037
7
16%
57/56
Lysosome
Protein degradation



P29373
5.42
15854
6
46%
66/56
Cytoplasma
Transport



O00299
5.09
27248
8
43%
114/56 
Cell Membrane
Transport



O00299
5.09
27248
9
44%
98/56
Cell Membrane
Transport



Q9Y696
5.45
28982
6
32%
64/56
Cell Membrane
Transport



Q9Y696
5.45
28982
6
26%
69/56
Cell Membrane
Transport



O75390
8.45
51908
4
10%
58/56
Mito
TCA cycle



O14579
4.97
34688
15
50%
181/56 
Golgi
Vascular transport



P23528
8.22
18719
5
37%
56/56
Cytoplasma
Cell motility/Ca










regulation



A6NKD9
6.48
45467
7
16%
58/56
Cytoplasma
Unknown



Q14993
8.57
115947
7
9%
61/56
Secreted
Cell-cell interaction



Q15828
8.31
16785
4
34%
61/56
Secreted
Other



Q04695
4.97
48361
8
18%
68/56
Cytoplasma
Cytoskeleton



P19012
4.71
49365
9
25%
106/56 
Cytoplasma
Cytoskeleton



P05787
5.52
53671
14
31%
108/56 
Cytoplasma
Cytoskeleton



Q9UBS4
5.81
40774
6
19%
69/56
ER
Protein folding



P29692
4.9
31217
6
28%
84/56
Cytoplasma
Biosynthesis



P26641
6.25
50429
10
36%
124/56 
Cytoplasma
Biosynthesis



P13639
6.41
96246
10
12%
67/56
Cytoplasma
Biosynthesis



P30040
6.77
29032
6
24%
69/56
ER
Protein folding



P30040
6.77
29032
7
24%
70/56
ER
Protein folding



P14625
4.76
92696
10
10%
89/56
ER
Protein folding



O94905
5.47
38044
9
27%
111/56 
Cell Membrane
Protein catabolism



P63241
5.08
17049
8
59%
109/56 
Nucleus
Biosynthesis



P63241
5.08
17049
4
50%
56/56
Nucleus
Biosynthesis



P47756
5.36
31613
9
34%
97/56
Cytoskeleton
Cell motility



P47756
5.36
31613
7
27%
74/56
Cytoskeleton
Cell motility



Q02790
5.35
52057
12
26%
94/56
Cytoskeleton
Protein folding



P30043
7.13
22219
6
44%
82/56
Cytoplasma
Redox regulation



P09467
6.54
37190
12
40%
128/56 
Cytoplasma
Metabolism



P09467
6.54
37190
14
42%
123/56 
Cytoplasma
Metabolism



P04075
8.3
39851
6
28%
60/56
Cytoplasma
Metabolism



P04075
8.3
39851
6
28%
80/56
Cytoplasma
Metabolism



P09382
5.34
15048
5
52%
82/56
Cytoplasma
Cell-cell interaction



P11413
6.39
59675
13
20%
100/56 
Cytoplasma
Metabolism



P14314
4.33
60357
13
17%
87/56
ER
Metabolism



P21266
5.37
26998
8
40%
100/56 
Cytoplasma
Redox regulation



P21266
5.37
26998
7
37%
88/56
Cytoplasma
Redox regulation



P09211
5.43
23569
10
57%
123/56 
Cytoplasma
Redox regulation



P09211
5.43
23569
5
31%
60/56
Cytoplasma
Redox regulation



P62993
5.89
25304
7
26%
91/56
Golgi
Sgnal Transduction



Q12931
8.3
80345
10
18%
64/56
Mito
Protein folding/Cell










survival



P04792
5.98
22826
9
40%
92/56
Cytoplasma
Protein folding



P04792
5.98
22826
6
30%
68/56
Cytoplasma
Protein folding



P04792
5.98
22826
8
36%
78/56
Cytoplasma
Protein folding



P68871
6.75
16102
6
53%
83/56
Cytoplasma
Oxygen transport



P61978
5.39
51230
5
13%
74/56
Nucleus
Gene Regulation



P22626
8.97
37464
7
34%
85/56
Nucleus
Gene Regulation



Q13547
5.31
55638
7
22%
60/56
Nucleus
Gene Regulation



Q9Y4L1
5.16
111494
11
13%
77/56
ER
Redox regulation



Q04760
5.12
20992
6
26%
67/56
Cytoplasma
Metabolism



P09960
5.8
69868
9
19%
84/56
Cytoplasma
Metabolism



P07195
5.71
36900
6
17%
58/56
Cytoplasma
Metabolism



P07195
5.71
36900
6
21%
64/56
Cytoplasma
Metabolism



P40926
8.92
35965
7
26%
69/56
Mito
TCA cycle



O00264
4.56
21772
4
16%
58/56
Cell Membrane
Receptor



O75121
5.13
45750
7
13%
72/56
Cell membrane
Unknown



Q15691
5.02
30151
9
48%
143/56
Cytoplasma
Cytoskeleton



P26038
6.08
67892
7
12%
73/56
Cell Membrane
Cytoskeleton



P60660
4.56
17090
5
30%
63/56
Cytoplasma
Cell motility



O14950
4.71
19824
5
23%
559/56
Cytoplasma
Cell motility



P19105
4.67
19839
4
30%
67/56
Cytoplasma
Cell motility



P30084
4.95
294989
9
5%
58/56
Cytoplasma
Cell motility



Q14697
5.74
107263
26
32%
233/56 
Golgi
Metabolism



Q14697
5.74
107263
16
20%
164/56 
Golgi
Metabolism



Q9NXR1
5.2
38842
7
21%
57/56
Cytoplasma
Cell cycle



P15531
5.83
17309
6
42%
89/56
Cytoplasma
Biosynthesis



P20472
4.98
12051
6
55%
62/56
Nucleus
Cell motility/Ca










regulation



O00151
6.56
36505
8
27%
97/56
Cytoplasma
Cell motility/Ca










regulation



P62937
7.68
18229
9
53%
91/56
Cytoplasma
Protein folding



P62937
7.68
18229
6
36%
68/56
Cytoplasma
Protein folding



Q96BP3
6.7
74098
6
12%
68/56
Splicesome
Protein folding



Q13162
5.86
30749
6
24%
58/56
Cytoplasma
Redox regulation



Q13162
5.86
30749
7
32%
80/56
Cytoplasma
Redox regulation



P30041
6
25133
10
51%
116/56 
Cytoplasma
Redox regulation



P30041
6
25133
7
31%
98/56
Cytoplasma
Redox regulation



Q9Y617
7.56
40796
6
16%
62/56
Cytoplasma
Metabolism



P13797
5.52
70904
7
11%
62/56
Cytoplasma
Cytoskeleton



P07737
8.44
15216
5
47%
62/56
Cytoplasma
Cell motility



P07737
8.44
15216
5
47%
63/56
Cytoplasma
Cell motility



Q8WUM4
6.13
96590
9
14%
61/56
Cytoplasma
Vascular transport



P35232
5.57
29843
7
37%
82/56
Mito
Gene Regulation



Q15185
4.35
18971
6
35%
71/56
Cytoplasma
Sgnal Transduction



Q06323
5.78
28876
12
39%
98/56
Proteasome
Protein degradation



Q9UL46
5.44
27515
7
30%
67/56
Proteasome
Protein degradation



Q9UL46
5.44
27515
7
33%
83/56
Proteasome
Protein degradation



P61289
5.69
29602
8
36%
72/56
Proteasome
Protein degradation



Q9Y2B0
4.81
20981
10
53%
161/56 
ER
Gene Regulation



Q6XLA1
8.53
12065
4
21%
64/56
Peroxisome
Gene Regulation



P07237
4.76
57480
6
12%
66/56
ER
Protein folding



P30101
5.98
57146
10
23%
84/56
ER
Protein folding



Q15084
4.95
48490
7
24%
77/56
ER
Protein folding



Q9HCY8
5.16
11826
5
63%
92/56
Cytoplasma
Signal transduction/










Ca regulation



Q6PI26
4.7
65712
6
14%
56/56
Cytoplasma
Unknown



P14618
7.96
58470
10
23%
101/56 
Cytoplasma
Metabolism



P50395
6.11
51087
15
39%
158/56 
Cell Membrane
Sgnal Transduction



Q8WUD1
7.68
24427
5
29%
72/56
Cell Membrane
Sgnal Transduction



Q14257
4.26
36911
6
25%
76/56
ER
Signal transduction/










Ca regulation



O94788
5.79
57144
7
14%
61/56
Cytoplasma
Biosynthesis



P42331
5.83
72955
6
11%
62/56
Cell Membrane
Cell motility



Q13017
6.18
173834
8
6%
63/56
Cell Membrane
Cell motility



O75116
5.75
161952
9
5%
59/56
Cytoplasma
Cell motility



O75116
5.75
161952
14
11%
66/56
Cytoplasma
Cell motility



P13489
4.71
51766
9
30%
115/56 
Cytoplasma
Sgnal Transduction



Q13228
5.93
52928
8
15%
71/56
Cell Membrane
Transport



Q13228
5.93
52928
9
15%
83/56
Cell Membrane
Transport



Q9NVA2
6.36
49652
9
27%
110/56 
Nucleus
Cell cycle



Q92599
5.89
56234
9
22%
104/56 
Nucleus
Cell cycle



Q8TDX7
8.49
34985
7
22%
66/56
Cytoplasma
Cell cycle



Q00536
7.23
55909
7
15%
68/56
Cytoplasma
Cell cycle



P36952
5.72
42568
7
28%
81/56
Secreted
Protease inhibitor



P55854
5.32
11687
5
37%
81/56
Cytoplasma
Protein degradation



P31948
6.4
63227
9
20%
62/56
Cytoplasma
Protein folding



Q8NBS9
5.63
48283
7
22%
68/56
ER
Redox regulation



Q8NBS9
5.63
48283
9
26%
124/56 
ER
Redox regulation



Q9UI15
6.84
22629
6
27%
63/56
Cytoplasma
Unknown



P60174
6.45
26938
12
66%
142/56 
Cytoplasma
Metabolism



P60174
6.45
26938
7
34%
95/56
Cytoplasma
Metabolism



P67936
4.67
28619
7
22%
80/56
Cytoplasma
Cell motility/Ca










regulation



P61088
6.13
17184
6
41%
70/56
Nucleus
Gene Regulation



Q5VYV7
9.5
46094
5
17%
58/56
Unknown
Unknown



P08670
5.06
53676
6
15%
70/56
Cytoplasma
Cytoskeleton



P08670
5.06
53676
16
32%
93/56
Cytoplasma
Cytoskeleton



P21796
8.62
30868
8
40%
110/56 
Mito
Transport



P45880
7.49
32060
6
31%
84/56
Mito
Transport



Q2VY69
8.77
71198
8
11%
59/56
Nucleus
Gene Regulation



Q8N7K0
9.38
79872
11
24%
75/56
Nucleus
Gene Regulation



Q6ZNA1
9.39
111011
6
8%
56/56
Nucleus
Gene Regulation











*The subcellular locations and functional classes of identified proteins were


obtained from the Uniprot website.






EXAMPLE 8
Validation of Characterized Breast Cancer Related Proteins Through Immunoblotting and Immunofluorescence

The secrometic experiment of the present invention indentified some of the well-characterized breast cancer related cytosolic proteins such as Cyclophilin A, 14-3-3 delta and peroxiredoxin 2 in culture media. It wa essential to validate the levels of these cytosolic proteins in the medium from independent experiments. To this end, the expression level of cyclophilin A, 14-3-3 delta and peroxiredoxin 2 from the culture media of MDA-MB-231, MCF-7 and MCF-10A were validated with immunoblotting. The results indicated that both the proteomic and immunoblot analysis showed cyclophilin A and 14-3-3 delta down-regulated in MCF-7 in comparison to the levels in MCF-10A. In contrast, peroxiredoxin 2 showed up-regulation in MCF-7 in comparison to the levels in MCF-10A. Comparing the secreted protein levels between MCF-10A and MDA-MB-231 indicates that the peroxiredoxin 2 and 14-3-3 delta expression levels increased in MDA-MB-231 and MCF-10A, respectively; however, the cyclophilin A level showed no significant change (FIG. 6 A˜C). This observation confirmed that cyclophilin A, 14-3-3delta and peroxiredoxin 2 were differentially secreted across the breast cells.


Immunoblot and immunofluorescence analyses were carried out to further confirm the differential protein levels observed in the total cellular proteins (profilin, cathepsin D, annexin 2, protein disulfide isomerase A1 and HDAC1) across MDA-MB-231, MCF-7 and MCF-10A (FIG. 6 D˜H). These proteins have been reported to play important roles in cytoskeleton regulation, proteolysis, calcium regulation, protein disulfide bond rearrangement and chromatin assembly during tumorigenesis. The results of the immunoblotting indicated that cathepsin D and PDI showed up-regulation in MCF-7 cells but down-regulation in MDA-MB-231 compared to the two protein expressions in MCF-10A. The expression levels of the profilin and annexin 2 proteins showed down-regulation in MCF-7 but no significant changes in MDA-MB-231 compared to the levels in MCF-10A. These immunoblotting results demonstrated a positive correlation with the 2D-DIGE results (FIG. 6 D˜G). In addition to immunoblotting, validation was also performed with immunofluorescent analysis. FIG. 6H shows that most of the HDAC1 signal was distributed within the nucleus, which is consistent with the subcellular location of HDAC1 in cells. As expected, the fluorescent intensity with the same exposure indicates that HDAC1 showed increased expressions in MCF-7 and MDA-MB-231 compared to its expression in MCF-10A. Altogether, the results from immunoblotting and immunofluorescent agreed with the results from 2D-DIGE data.


EXAMPLE 9
Validation of Unreported Identified Putative Tumorigenesis Markers Through Immunoblotting and Immunofluorescence

The cellular proteomic and secretomic analyses above reveal a number of identified proteins may be breast cancer markers (Tables 1 and 2). To verify this observation, immunoblotting and immunofluorescence were used to validate these differentially expressed proteins including bestrophin 3, MPP2, parvalbumin, PdLIM1, IFIT3 and BANF1 as these proteins showed relatively significant changes (>3 fold) in comparison with most of the unreported identified proteins across MCF-10A, MCF-7 and MDA-MB-231. The immunoblotting analysis of concentrated serum-free media shows that more bestrophin 3 was secreted in the cell lines of MCF-7 and MDA-MB-231 than MCF-10A, while MPP2 was only detected in MDA-MB-231. Notably, the bestrophin 3 blotting result did not completely agree with the 2D-DIGE data, where levels in MCF-7 were higher than MB-231 (FIG. 7A). Using immunofluorescent staining, the robust increase of parvalbumin signal in both the MCF-7 and MDA-MB-231 cells was first confirmed after comparison with the signal in MCF-10A. Parvalbumin was primarily localized in the nucleus, which coincided with the DAPI stained nucleus. Further investigation of parvabumin expression in other breast cancer cell lines indicated that parvabumin was over-expressed in MDA-MB-453, a line of non-invasive breast cancer cells, and slightly up-regulated in MDA-MB-361, an adenocarcinoma with metastatic ability (FIG. 7B). These results implied that parvabumin might have the potential to be a breast cancer marker. In contrast, PdLIM1, a cytosolic protein, was down-regulated in all breast cancer lines: MCF-7, MDA-MB-231, MDA-MB-453 and MDA-MB-361 (FIG. 7B). In addition, IFIT3, a plasma membrane protein, was down-regulated in transformed cells, especially in MCF-7 and MDA-MB-231, and was consistent with the proteomic data from 2D-DIGE (FIG. 7B). Interestingly, BANF1, a major nucleus-located protein, was distributed in the cytoplasma of the MCF-10A cells, but was confined within the nucleus in MCF-7, MDA-MB-231 and MDA-MB-453 cells; in addition, BANF1 was distributed within the cytoplasma and nucleus in MDA-MB-361 (FIG. 7B). These results indicated that the BANF1 levels were different between normal breast cells and breast cancer cells, and that the subcellular locations of the protein may account for tumorigenesis.


With the basis of a Swiss-Prot search and KEGG pathway analysis, numerous potential biological functions of the identified proteins across MCF-10A, MCF-7 and MDA-MB-231 were determined. The information should be useful for studying the mechanisms of breast cancer tumorigenesis and metastasis.



FIG. 8 compared the expression profiles of the identified differentially expressing proteins in these 3 cell lines. Proteins known to regulate cell cycle are found to be upregulated in both MCF-7 and MDA-MB-231 (FIG. 8A), and are associated with the promotion of tumorigenesis. In addition, the expression of proteins linked to redox-regulation increased in the MCF-7 cells in comparison to the levels in MCF-10A (FIG. 8B). Induced expression of these proteins may account for cancer development and progression. Proteomic analysis also reveals that proteins involved in carbohydrate metabolism are significantly over-expressed in MCF-7 cells (FIG. 8C). This demonstrates that cancer cells rely heavily on glycolysis to obtain ATP for proliferation and tumorigenesis in the presence of adequate oxygen levels; this mechanism has been implicated in numerous cancer therapies. FIGS. 8D˜F show the downregulated profiles of proteins in both MCF-7 and MDA-MB-231 cells. These proteins are involved in calcium regulation, vascular transport and protease inhibition. Calcium-binding proteins, such as annexin 1, whose function is modulated by an estrogen receptor, have been reported to show decreased expression in correlation with breast cancer development and progression. The S100 protein family is a family of low molecular weight calcium-binding proteins that is responsible for the regulation of protein phosphorylation, intracellular calcium homostasis, the dynamics of cytoskeleton constituents and cell proliferation. S100A14 was identified as downregulated in MCF-7 and MDA-MB-231, suggesting its potential role in breast cancer. Howerever, proteins involved in vascular transport, including Rab GTPase-binding effector protein and vacuolar protein sorting-associated protein 54, were decreased in expression in MCF-7 and MDA-MB-231 (FIG. 8F). This may be explained by a previous report indicating that the downregulation of Rab5 GDP/GTP exchange factor enhances receptor tyrosine kinase signaling and promotes the growth factor-directed migration of tumor cells (Hu, H.; Milstein, M.; Bliss, J. M.; Thai, M.; Malhotra, G; Huynh, L. C.; Colicelli, J. Mol. Cell Biol. 2008, 28, 1573). However, there are few studies on tumorigenesis regarding the roles of the Rab GTPase-binding effector protein and the vacuolar protein sorting-associated protein 54. Serpin is a group of proteins able to inhibit protease and block the growth, invasion, and metastatic properties of breast tumors. Hence, serpin families function as tumor suppressors in cancer research. The downregulation of serpin is well-correlated with the progression of breast cancer and the present observations in MCF-7 and MDA-MB-231 cells (FIG. 8F).


Other differentially expressed proteins of interest across MCF-10A, MCF-7 and MDA-MB-231 include cathepsin D, bestrophin-3 and interferon-induced protein with tetratricopeptide repeats 3 (IFIT3). Cathepsin D, a lysosomal aspartic protease, is over-expressed in estrogen receptor positive breast cancer cells and is generally of good prognostic value in comparison with estrogen receptor negative breast cancer in clinical studies. The present invention indicates that cathepsin D is highly expressed in MCF-7, both in total cellular proteins or in secreted fraction. In contrast, cathepsin D is significantly down-regulated in MDA-MB-231 cells compared with MCF-7.


Results of the proteomic experiments display good correlation with secrmetic experiments.


Bestrophin-3, a cGMP-dependent calcium-activated chloride channel, has not been reported to be associated with cancer and shows upregulation in MCF-7 and MDA-MB-231 in the present inveniton. Nevertheless, the related study in bestrophin-1 showed the protein improves intracellular Ca2+ signaling and increases cell growth rate in colonic carcinoma cells. The proliferation of the cells was significantly suppressed by bestrophin-1 RNA interference treatment (Spitzner, M.; Martins, J. R.; Soria, R. B.; Ousingsawat, J.; Scheidt, K.; Schreiber, R.; Kunzelmann, K. J. Biol. Chem. 2008, 283, 7421). This indicated that bestrophin-3 may be a potential target for breast cancer therapy. IFIT3 plays a key role in the antiproliferative activity of the interferon-related signaling pathway through inducing expression of cell cycle inhibitors, p21 and p27 proteins (Xiao, S.; Li, D.; Zhu, H. Q.; Song, M. G; Pan, X. R.; Jia, P. M.; Peng, L. L.; Dou, A. X.; Chen, G. Q.; Chen, S. J.; Chen, Z.; Tong, J. H. Proc. Natl. Acad. Sci. U.S.A 2006, 103, 16448). The 2D-DIGE results in this invention showed that IFIT3 is downregulated in both MCF-7 and MDA-MB-231 cells, implying that breast cancer cells may maintain a high level of proliferative activity by downregulating the expression of IFIT3.


Based upon the data presented here, the present invention infers that proteins selected from biomarker library plays key role in the regulation of breast cancer progression. Combining the data of 2D-DIGE and immunoassay, it concludes that the increasing or decreasing of biomarker expression relative to the expression of normal tissue results in an increased likelihood of breast cancer development.

Claims
  • 1. A breast cancer biomarker library, comprising bestrophin-3, carbonic anhydrase 2, dynein heavy chain 6, ecto-ADP-ribosyltransferase 4, GRAM domain-containing protein 2, interferon-induced protein with tetratricopeptide repeat 3, phosphoglycerate mutase 1, proteasome subunit alpha type-1, proteasome subunit alpha type-3, rab GTPase-binding effector protein 2, Ras-related protein Rab-2B, selenium-binding protein 1, transmembrane protein C14orf180, vascular protein sorting-associated protein 54, achaete-scute homologue 4, aconitate hydratase, aminopeptidase B, annexin A3, barrier-to-autointegration factor, bifunctional purine biosynthesis, calumenin, carbonic anhydrase 2, coiled-coil domain-containing protein, erlin-2, F-actin-capping protein subunit beta, flavin reductase, fructose-1,6-biphosphatase 1, fructose-biphosphate ldolase A, heat shock protein 75 kDa, heterogeneous nuclear ribonucleoproteins A2/B 1, leukotriene A-4 hydrolase, microfibrillar-associated protein 3 like, microtubule-associated protein RP, nuclear distribution protein nudE homologue 1, parvalbumin alpha, PDZ and LIM domain protein 1, peptidylprolyl isomerase domain and WD repeat-containing protein 1, phosphoserine aminotransferase, plastin-3, programmed cell death 6-interacting protein, proteasome activator complex subunit 1, proteasome activator complex subunit 2, protein canopy homologue 2, protein CASC2, protein disulfide-isomerase A6, protein SHQ1, Rab GDP dissociation inhibitor beta, reticulocalbin-2, Rho GTPase-activating protein 25, Rho GTPase-activating protein 5, ribonuclease inhibitor, selenium-binding protein 1, septin-11, septin-8, serine/threonine-protein kinase Nek7, serine/threonine-protein kinase PCTAIRE-1, small ubiqutin-related modifier 3, stress-induced phosphoprotein 1, thioredoxin domain-containing protein 5, ubiquitin-conjugating enzyme E2, UPF0492 protein C20orf94, voltage-dependent anion-selective channel protein and zinc finger protein 433.
  • 2. A method of predicting an increased likelihood of developing breast cancer progression of a subject comprising: (c) detecting the expression of at least one biomarker in a sample from a subject with bioassays, wherein the biomarker is selected from the biomarker library of claim 1; and1(d) comparing the pattern of biomarker expression in (a) to a reference biomarker expression pattern from normal tissue, wherein at least one fold of increasing or decreasing biomarker expression relative to the reference indicating an increased likelihood of breast cancer development.
  • 3. The method of claim 2, wherein the breast cancer progression includes the presence or absence of breast tumor, the stage of breast cancer and the effectiveness of breast cancer treatment.
  • 4. The method of claim 3, wherein the stage of breast cancer includes invasive and non-invasive tumor pregression.
  • 5. The method of claim 2, wherein the subject is human or mammal.
  • 6. The method of claim 2, wherein the sample is selected from blood, serum, plasma, ductal lavage fluid, nipple aspiration fluid.
  • 7. The method of claim 2, wherein the bioassays comprises immunoassay, electrophoresis and mass spectrometry.
  • 8. The method of claim 7, wherein the immunoassay is measured using immunoblotting.
  • 9. The method of claim 8, wherein the immunoblotting is an antibody-based assay.
  • 10. The method of claim 2, while further comprises applying software for protein expression comparison between normal tissue and tumor tissue.
  • 11. The method of claim 2, wherein the expression pattern indicates the quantity of biomarker expression.
  • 12. The method of claim 2, wherein the increasing or decreasing of biomarker expression is 1.5 fold relative to the biomarker expression of normal tissue.