ArticlePDF Available

Polley AC, Mulholland F, Pin C, Williams EA, Bradburn DM, Mills SJ, Mathers JC, Johnson ITProteomic analysis reveals field-wide changes in protein expression in the morphologically normal mucosa of patients with colorectal neoplasia. Cancer Res 66: 6553-6562

Authors:

Abstract and Figures

Models for the pathogenesis of colorectal cancer tend to focus on the localized lesion, with less attention paid to changes in normal-appearing mucosa. Here we used two-dimensional gel electrophoresis and mass spectrometry to define patterns of protein expression in morphologically normal colonic mucosa from 13 healthy subjects, 9 patients with adenomatous polyps, and 9 with cancer. Tumor samples were also compared with the normal mucosa. Systematic gel comparisons identified a total of 839 spots that differed significantly between one or more groups (P < 0.05). Principle component analysis indicated that the first three components accounted for approximately 37% of the total variation and provided clear evidence that flat mucosa from healthy subjects differed significantly from that of patients with polyps or cancer. Sixty-one proteins differed significantly between mucosa from healthy subjects and all other tissue types, and 206 differed significantly between healthy mucosa and polyp mucosa. Several of the proteins showing significant underexpression in tumor tissue were cytokeratins and other cytoskeletal components. In contrast, cytokeratins, including several isoforms of cytokeratin 8, were overexpressed in apparently normal mucosa from polyp and cancer patients compared with mucosa from healthy subjects. These findings indicate that protein expression in the apparently normal colonic mucosal field is modified in individuals with neoplastic lesions at sites distant from the lesion. Recognition and further characterization of this field effect at the molecular level may provide protein biomarkers of susceptibility to colorectal cancer and facilitate development of hypotheses for the role of diet and other environmental factors in its causation.
Content may be subject to copyright.
2006;66:6553-6562. Cancer Res
Abigael C.J. Polley, Francis Mulholland, Carmen Pin, et al.
Patients with Colorectal Neoplasia
Expression in the Morphologically Normal Mucosa of
Proteomic Analysis Reveals Field-Wide Changes in Protein
Updated version
http://cancerres.aacrjournals.org/content/66/13/6553
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/66/13/6553.full.html#ref-list-1
This article cites by 43 articles, 16 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/66/13/6553.full.html#related-urls
This article has been cited by 8 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Proteomic Analysis Reveals Field-Wide Changes in Protein
Expression in the Morphologically Normal Mucosa of
Patients with Colorectal Neoplasia
Abigael C.J. Polley,
1
Francis Mulholland,
1
Carmen Pin,
1
Elizabeth A. Williams,
2
D. Mike Bradburn,
3
Sarah J. Mills,
3
John C. Mathers,
2
and Ian T. Johnson
1
1
Institute of Food Research, Norwich Research Park, Norwich, United Kingdom;
2
Human Nutrition Research Centre, School of Clinical
Medical Sciences, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, United Kingdom; and
3
Wansbeck Hospital, Ashington, Northumberland, United Kingdom
Abstract
Models for the pathogenesis of colorectal cancer tend to focus
on the localized lesion, with less attention paid to changes in
normal-appearing mucosa. Here we used two-dimensional gel
electrophoresis and mass spectrometry to define patterns of
protein expression in morphologically normal colonic mucosa
from 13 healthy subjects, 9 patients with adenomatous polyps,
and 9 with cancer. Tumor samples were also compared with
the normal mucosa. Systematic gel comparisons identified a
total of 839 spots that differed significantly between one
or more groups (P < 0.05). Principle component analysis
indicated that the first three components accounted for
f37% of the total variation and provided clear evidence that
flat mucosa from healthy subjects differed significantly from
that of patients with polyps or cancer. Sixty-one proteins
differed significantly between mucosa from healthy subjects
and all other tissue types, and 206 differed significantly
between healthy mucosa and polyp mucosa. Several of the
proteins showing significant underexpression in tumor tissue
were cytokeratins and other cytoskeletal components. In
contrast, cytokeratins, including several isoforms of cytoke-
ratin 8, were overexpressed in apparently normal mucosa
from polyp and cancer patients compared with mucosa from
healthy subjects. These findings indicate that protein expres-
sion in the apparently normal colonic mucosal field is modi-
fied in individuals with neoplastic lesions at sites distant from
the lesion. Recognition and further characterization of this
field effect at the molecular level may provide protein bio-
markers of susceptibility to colorectal cancer and facilitate
development of hypotheses for the role of diet and other
environmental factors in its causation. (Cancer Res 2006;
66(13): 6553-62)
Introduction
The central paradigm for the pathogenesis of colorectal cancer is
the adenoma-carcinoma sequence, a complex stepwise series of
changes in cellular proliferation and differentiation, driven by a
progressive accumulation of genetic abnormalities, leading to
malignancy via adenomatous polyps (1). It is increasingly recog-
nized that this model is probably inadequate in that other
molecular abnormalities, including modified epigenetic marks, also
contribute and may define alternative pathways to neoplasia (2).
However, most current mechanistic models focus almost exclusively
on the localized lesion, with much less attention paid to pathologic
changes occurring in the normal-appearing mucosa from which
such lesions emerge. Physiologic anomalies in the flat mucosa,
including abnormal cell proliferation (3, 4), apoptosis (5), and gene
expression (6), have previously been reported but the nature, dura-
tion, and causes of such putative field effects are poorly defined.
The physiologic state of a complex tissue is reflected in the full
complement of proteins expressed by its constituent cells. The
pattern of expressed proteins thus constitutes a ‘library’’ of
information about the functional status and health of the tissue.
The development of new methods for protein extraction, display,
and analysis has led to the emergence of a new field of clinical
proteomics, in which these techniques are harnessed to identify
biomarkers of cancer and other diseases (7), but there are few
studies on the differential expression of proteins during early
colorectal carcinogenesis.
Tracy et al. (8) described reproducible patterns of protein
expression associated with the normal mucosa and specific
differences associated with tumors. Similarly, Anderson et al. (9)
compared hepatic and colorectal tumors with their normal host
tissues, and with each other, and identified characteristic patterns
of protein expression that could be used to distinguish primary and
secondary tumors from their adjacent uninvolved tissues. Several
other groups have reported similar findings and the development
of proteomic technology has enabled a more comprehensive
analysis of protein expression (10, 11). Among the most recent of
these studies is that of Friedman et al. (12), who employed two-
dimensional difference gel electrophoresis to compare tumor
samples and macroscopically normal mucosa. Using this approach,
Friedman’s group observed 52 proteins for which statistically
significant differences in abundance were detectable within the
mucosa/tumor pairs from the same patients.
Although many different techniques for the collection and
fractionation of tissues have been employed, virtually all previous
studies have compared tumors with flat mucosa from the same
individual. Whereas this approach provides a comparison of ana-
tomically normal and neoplastic tissues against the same genetic
background, it does not address the possibility that the apparently
normal mucosa of healthy individuals differs from that of
those with neoplastic lesions. The possibility of precancerous field
changes that render the mucosa more vulnerable to the emergence
of localized lesions has long been recognized but seldom explored.
In the present study, we used proteomic techniques to test the
Note: Present address for E. A Williams: Centre for Human Nutrition, Northern
General Hospital, University of Sheffield, Sheffield S5 7AU, United Kingdom.
Requests for reprints: Ian T. Johnson, Institute of Food Research, Norwich
Research Park, Norwich, NR4 7UA, United Kingdom. Phone: 01603-255330; Fax: 01603-
255288; E-mail: ian.johnson@bbsrc.ac.uk.
I2006 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-06-0534
www.aacrjournals.org
6553
Cancer Res 2006; 66: (13). July 1, 2006
Research Article
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
hypothesis that colonic mucosa from disease-free patients would
exhibit patterns of protein expression distinct from the mucosa of
patients with adenomatous polyps or cancer. Tumors from the
cancer patients were also compared with each group of normal
mucosal samples.
Materials and Methods
Patients and biopsies. Volunteers were either patients with previously
diagnosed colorectal cancer or outpatients with no known major pathology,
typically presenting for investigation of symptoms, including abnormal
bowel habit or rectal bleeding, and undergoing flexible sigmoidoscopy or
colonoscopy as a diagnostic procedure. All patients were recruited from
the gastroenterology outpatient and surgical lists of the Wansbeck General
Hospital, Ashington, Northumberland, United Kingdom. Ethical approval
for the project was received from the Northumberland Local Research
Ethics Committee (project reference NLREC2/2001). Patients were con-
tacted in advance and sent an information leaflet, and those patients
consenting to the study were advised to attend endoscopy or theatre as
expected. Experimental biopsies were collected from the rectum of the
endoscopy patients, in addition to those obtained for diagnostic purposes.
For the cancer patients, samples of normal rectal mucosa (>10 cm from
tumor margin) and tumor tissue were collected at surgery. All samples
were immediately snap frozen in liquid nitrogen and transferred to a 80jC
freezer. Medical notes for each volunteer were reviewed 6 to 8 weeks after
the procedure and the findings of the pathology report and the conclusions
of the responsible consultant were recorded. The final groups were
composed of 13 ‘‘normal’’ individuals [9 females (mean age, 54.8 years;
SD, 8.4 years) and 4 males (mean age, 68 years; SD, 3.6 years)] showing no
evidence of neoplasia; 9 ‘‘polyp’’ patients [5 females (mean age, 66.6 years;
SD, 15.4 years) and 4 males (mean age, 55.8 years; SD, 4.3 years)] in whom
adenomatous polyps were detected at endoscopy; and 10 ‘‘cancer’’ patients.
The latter group was composed of six females (mean age, 64.5 years; SD,
8.4 years) and four males (mean age, 66.5 years; SD, 11.6 years), most of
whom had moderately differentiated adenocarcinomas of the recto-
sigmoidal region (Table 1). Two-dimensional gel electrophoresis was done
on 41 samples; one mucosal biopsy from a cancer patient was lost.
Protein extraction. Mucosal biopsies and samples of tumor were
thawed, weighed, and extracted without further manipulation. Resected
mucosa was allowed to thaw and subsamples of mucosal tissue (10-15 mg)
were scraped from the underlying muscle layers using a glass microscope
slide. The tissue was extracted using Bio-Rad ReadyPrep Sequential
Extraction Kit (Bio-Rad, Hemel Hempstead, United Kingdom) with the
following additions to Reagent 1 just before use: MgCl
2
(3 mmol/L),
protease inhibitor cocktail (Sigma, Poole, United Kingdom; 2.5 AL/mL),
DNase I (RNase-free; 5 units/mL), and RNase A (5 Ag/mL). Modified
Reagent 1 (25 AL) was added and each tissue sample was hand homo-
genized. A further 175 AL of Reagent 1 were added and the whole volume
was sonicated for 10 minutes at room temperature, centrifuged to obtain a
pellet, and the supernatant was removed. The volume equivalent to 100 Ag
protein was determined using a Bio-Rad Protein Assay according to the
instructions of the manufacturer, with bovine g globulin as standard. The
extraction supernatants were stored at 80jC.
Two-dimensional gel electrophoresis. For isoelectric focusing in the
first dimension, a sample volume equivalent to 100 Ag protein was added to
a rehydration mix, containing urea (7 mol/L), thiourea (2 mol/L), CHAPS
(2%), bromophenol blue, DTT (18.2 mmol/L), and IPG buffer (0.5%, pH 4-7;
GE Healthcare, Little Chalfont, United Kingdom) to make a final volume of
450 AL. The whole volume was transferred into a well of the Immobiline
DryStrip re-swelling tray and IPG strips (24 cm, pH 4-7; GE Healthcare) were
rehydrated overnight at 20jC. Each strip was then transferred to a ceramic
strip-holder and submerged in DryStrip Cover Fluid (f3.5 mL). The iso-
electric focusing was run on an Ettan IPGphor bed (GE Healthcare) with
a gradient of 500 V for 1 hour (500 V-h), 4,000 V for 1.5 hours (6,000 V-h),
and a ‘‘step-n-hold’’ of 8,000 V for 6.75 hours (51,600 V-h). After completion
of isoelectric focusing, the strips were stored at 80jC.
The second-dimension protein separation was carried out on 1-mm-thick
10% gels, prepared in 28
23-cm gel-plate cassettes. Focused strips were
rinsed free of excess mineral oil, conditioned in modified Tris Acetate
Equilibration Buffer (Genomic Solutions, Huntingdon, United Kingdom),
treated first with 8 mg/mL DTT in equilibration buffer (9 mL; 30 minutes
with gentle shaking), transferred to 25 mg/mL iodoacetamide in equilibration
buffer (9 mL; 30 minutes with gentle shaking), and placed in a gel cassette
before transfer to the gel tank. The top reservoir contained the cathode
buffer (200 mmol/L Tris base, 200 mmol/L Tricine, 14 mmol/L SDS; Sigma)
and the bottom reservoir contained the anode buffer (25 mmol/L Tris-acetate
buffer, pH 8.3). Electrophoresis conditions were set to give an upper voltage of
500 V, power of 20,000 mW/gel, and a total run time of f3.5 hours.
Gel imaging and analysis. After electrophoresis, the gels were fixed and
stained using SYPRO Ruby Protein Gel Stain (Bio-Rad) and imaged using
ProXPRESS Proteomics Imaging System and Perkin-Elmer imaging
software. Images were saved as TIF files and analysis was carried out
using ProteomWeaver analysis software (Definiens, Munich, Germany).
Four gel groups were established representing the anatomically normal
mucosa from 13 healthy patients (‘‘healthy mucosa’’), 9 polyp patients
(‘‘polyp mucosa’’), and 9 cancer patients (‘‘cancer mucosa’’), and samples
of tumor from 10 cancer patients (‘‘tumor tissue’’). Each gel underwent
automatic spot detection and manual editing before automatic spot
matching both within and between groups. Manual matching was done
where necessary. Base-paired normalization was done on all the gels before
examination of spot volume data. Average gels were constructed using spots
that were detectable in a minimum of 50% of the gels in each group.
Selected spots were picked from a gel using the ProPick spot-picking robot
(Genomic Solutions) and gel plugs were transferred to a modified 96-well
microtitre plate.
Protein analysis. In-gel trypsin digestion was carried out using a
ProGest Protein Digester (Genomic Solutions). After preincubation, the
digestions were carried out at 37jC for 3 hours using 50 ng of sequencing
Table 1. Characteristics of cancer patients from whom mucosal and tumor biopsies were obtained in the present study
Patient no. Sex Age (y) Tumor site Duke’s stage Histology
1 F 51 Rectum C Poorly differentiated adenocarcinoma
2 F 66 Hepatic flexure B Histopathology report incomplete
3 F 60 Hepatic flexure B Moderately differentiated adenocarcinomas
4 F 72 Sigmoid colon C1 Moderately differentiated adenocarcinoma
5 F 64 Sigmoid colon/rectum B Moderately differentiated adenocarcinoma
6 F 74 Hepatic flexure B Moderately differentiated adenocarcinoma
7 M 66 Ascending colon B Moderately differentiated adenocarcinoma
8 M 60 Sigmoid colon/rectum C1 Moderately differentiated adenocarcinoma
9 M 83 Sigmoid colon/rectum A Moderately differentiated adenocarcinoma
10 M 57 Sigmoid colon/rectum B Moderately differentiated adenocarcinoma
Cancer Research
Cancer Res 2006; 66: (13). July 1, 2006
6554
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
grade porcine trypsin (5 AL/well; Promega, Southampton, United Kingdom).
The digests were analyzed using a Reflex III MALDI/ToF (Bruker Ltd.,
Coventry, United Kingdom) with Scout 384 ion source, fitted with a nitrogen
laser (wavelength, 337 nm) to desorb/ionize the matrix/analyte material
from the sample substrate. All spectra were acquired in a positive-ion
reflector set at the following variables: 25 kV acceleration voltage, 28.7 kV
reflection voltage, 20.9 kV ion source acceleration voltage, and 1.65 kV
reflector-detector voltage. Calibration was carried out using a set of peptide
standards having an approximate concentration of 1 pmol/AL from spots
adjacent to the samples. In some cases, quadrupole time-of-flight mass
spectrometry was done using a Micromass quadrupole time-of-flight
electrospray fitted with a Waters Cap LC system.
Peptide masses obtained from the matrix-assisted laser desorption/
ionization time-of-flight mass spectrometry were searched (on the basis of
mass) against the MSDB protein database using the Mascot peptide mass
fingerprint program from Matrix Science.
4
The search parameters were as
Figure 1. Two-dimensional protein map
based on the average gel for the healthy
mucosa. A, arrows, positions and spot
numbers for all the identified proteins
listed in Table 2. B, a similar map of
cancer mucosa is shown together with
spot numbers and positions for proteins
identified as cytokeratins. C, coordinates
for each tissue sample on the first
three principal components, following
principal component analysis carried
out on 839 proteins that differed
significantly across the 41 samples of
tissue analyzed. Each sample is
numbered to indicate its origin from
healthy mucosa (1), polyp mucosa (2 ),
cancer mucosa (3), or tumor tissue (4 ).
4
http://www.matrixscience.com.
Proteomic Analysis of Colorectal Mucosa
www.aacrjournals.org
6555
Cancer Res 2006; 66: (13). July 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
follows: (a) tryptic digest was assumed to have a maximum number of one
missed cleavage; (b) peptide masses were stated to be monoisotopic; (c)
methionine residues were assumed to be partially oxidized; (d) the
carbamidomethylation of cysteine residues was considered; (e) the mass
tolerance was kept at 75 ppm; and ( f ) the taxonomy group searched was
Homo sapiens. The results give a Probability Based Mowse Score (13), equal
to 10
Log(P), where P is the probability that the observed match is a
random event. Protein scores >63 are considered statistically significant
(P < 0.05) under the above parameters.
Statistical analysis. Gels were normalized in accordance with
ProteomWeaver software protocols using base-paired normalization. The
spot-density data were transferred to Excel spreadsheets for statistical
analysis using a nonparametric inference approach. Statistical filtering
retained only those spots for which average spot-density was >0.07
(arbitrary units), the within-group frequency was >50%, or which were
present in more than 6 of the 41 gels. To test the null hypothesis of equal
distribution of protein expression between groups, a nonparametric one-
way ANOVA (Kruskal-Wallis) was done on the ranked data (14). The null
hypothesis was rejected for any protein where the rank differed significantly
(P < 0.05) in at least one of the four groups. For all proteins showing
evidence of unequal expression between groups, principal component
analysis was carried out on the correlation matrix of the ranks of the 41
independent samples. F tests were used to assess the significance of
differences between groups and combinations of groups (15). Models were
set up to determine whether a particular protein differed significantly
between independent subsets of the four groups of observations. Differ-
ences were considered significant when the P value associated with the F
statistic was <0.05.
Results
Figure 1A is a two-dimensional protein map based on the
average gel for the healthy mucosa. The equivalent map for cancer
mucosa is shown in Fig. 1B. Gel-analysis software identified a total
of 6,494 unique spots across all the 41 gels and statistical filtering
yielded 1,910 spots of potential interest. Among these, the Kruskal-
Wallis test identified a total of 839 spots for which the average
density in one set of gels differed significantly (P < 0.05) from that
in at least one other set. For these proteins, principle component
analysis was carried out on the correlation matrix of the ranks of
the 41 independent samples; the first three principal components
were found to account for 37% of the total variability. Principal
component 1, which was highly correlated with 166 proteins
(correlation coefficient > 0.6) and represented f21% of the total
variation, indicated that the greatest differences in the ranks of
proteins occurred between the tumor tissues and the three sets of
morphologically normal mucosa. A total of 72 of the 839 proteins
Table 2. Characteristics of 26 identified proteins differentially expressed in tumor tissue compared with healthy mucosa, polyp
mucosa, and cancer mucosa
Spot no. Protein name Score Accession
no.
Theo. pI Theo. MW Act. pI Act. MW Expression
in tumor
117903 h-Actin 157 P60709 5.55 40,536 5.12 42,093 1.104
17654 a1-Antitrypsin precursor 235 P01009 5.37 46,878 4.98 59,532 0.788
3385 Apolipoprotein A-I precursor 162 P02647 5.56 30,759 5.24 24,471 3.031
9315 Calgizzarin 70 P31949 6.56 11,847 5.86 12,048 2.032
9669 Creatine kinase 261 P12277 5.34 42,902 5.43 42,655 2.571
12691 Cytokeratin 9 76 P35527 5.14 62,178 4.81 50,192 0.07
18566 Cytokeratin 19 268 P08727 5.04 44,065 4.87 45,588 0.298
17043 Cytokeratin 19 260 P08727 5.04 44,065 4.82 45,484 0.275
16185 Cytokeratin 19 223 P08727 5.04 44,065 4.78 45,640 0.449
64231 Cytokeratin 19 66 P08727 5.04 44,065 4.92 44,917 1.012
7046 Glycyl-tRNA synthetase 148 P41250 6.61 83,828 5.95 77,814 0.597
17391 IgG Fc binding protein 59 O95784 5.56 81,017 5.31 132,499 0.147
16796 IgG Fc binding protein 52 O95784 5.56 81,017 5.22 135,256 0.113
86052 Maspin 76 P36952 5.72 42,586 5.75 42,325 0.395
17865 Nucleoside diphosphate kinase A 164 P15531 5.42 17,309 5.57 19,758 1.154
114132 Protein disulfide isomerase 46 P07237 4.76 57,480 4.77 58,722 0.301
145857 Serum albumin, human 147 1AO6_A 5.73 68,126 5.45 52,323 0.207
13756 Serum albumin, human 91 1A06A 5.63 67,690 5.43 41,401 0.539
16253 Human serum albumin
complexed with myristic acid
131 1BJ5 5.73 68,126 5.77 24,841 0.447
5764 Stathmin 107 P16949 5.77 17,161 5.64 18,761 0.412
10280 Transthyretin (prealbumin)
complex with thyroxine (T4)
93 2ROX_A 5.33 12,996 5.48 16,838 1.144
8115 Tropomyosin a4 chain
(tropomyosin 4; TM30-pl)
63 P67936 4.67 28,487 4.68 31,786 1.928
9264 Ubiquitin thiolesterase 125 P15374 4.48 26,337 4.79 28,835 0.414
17404 Vinculin (metavinculin) 165 P18206 5.51 124,161 5.56 127,631 0.104
18975 14-3-3 h 120 P31946 4.76 28,179 4.76 28,296 4.11
17784 21K tumor protein 93 P13693 4.84 19,697 4.84 25,485 1.723
Abbreviations: Theo, theoretical data based on whole sequence in database; Act, actual gel pI and MW data from calibrated gel; Score, probability-based
Mowse score (see ref. 15); pI, isoelectric point.
Cancer Research
Cancer Res 2006; 66: (13). July 1, 2006
6556
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
differed significantly between tumor tissue and all three sets of
mucosal tissues. The second principal component accounted for
f10% of the total variation and was highly correlated with 56
proteins, 35 of which differed significantly among the three sets of
normal mucosal samples. The third component accounted for
about 6% of the total variation and was most highly correlated with
the polyp mucosa. Figure 1C illustrates the coordinates for each
tissue sample on the first three principal components. Each sample
is numbered to indicate its origin from healthy mucosa (1), polyp
mucosa (2), cancer mucosa (3), or tumor tissue (4). The separation
of the tumor samples and the three sources of mucosa into discreet
clusters is clearly apparent, with some evidence of a further
segregation within group 3.
Differential expression of proteins in tumor tissue com-
pared with flat mucosa. Use of the F test for between-group
comparisons indicated differential expression of 588 proteins in
tumor tissue compared with healthy mucosa, 536 compared with
polyp mucosa, and 520 compared with cancer mucosa. A total of
291 proteins differed significantly between tumor tissue and all
three groups of flat mucosal samples; of these, 90 were underex-
pressed in tumor tissue and 201 were overexpressed. A large
proportion of these proteins were so weakly expressed that analysis
was impractical, but 26 were positively identified by mass
spectrometry and are listed in Table 2.
Many of the spots showing differential expression among the four
groups of tissues were identified as cytoskeletal proteins. Among
these, we noted two separate spots identified as h-actin. This
protein is widely regarded as a ‘‘housekeeping gene,’’ characterized
by stable expression across a range of cell types, although variations
in expression in colorectal cancer (10) and other tissues (16) have
previously been reported. Most of the variation in h-actin
expression was associated with a weakly expressed spot (117903)
which was more prominent in tumor tissue (Table 2). Figure 2
indicates that the only detectable difference between spots 3476
and 117903 was the absence in the latter of the peptide of mass
1,516.75 Da. This corresponds to the peptide sequence QEYDESG-
PSIVHR, found at the COOH-terminal of the protein, which was the
fourth highest intensity peptide of the 14 used to derive the identity.
As all the other tryptic peptides were detected, it seems probable
that it was genuinely absent from spot 117903 and that a limited
COOH-terminal clipping accounted for the small shifts in molecular
mass and charge observed on the gel.
Other cytoskeletal proteins showing evidence of differential
expression in tumor tissue included cytokeratin 9 (CK9), four
isoforms of cytokeratin 19 (CK19), and vinculin. The latter is involved
in the attachment of actin-based microfilaments to the inner surface
of the plasma membrane (17). Conversely, h-actin and tropomyosin
(TM30-pl) both showed strong evidence of overexpression in tumor
tissue. Other proteins showing evidence of significant under-
expression included a1-antitrypsin precursor, creatine kinase,
protein disulfide isomerase, immunoglobulin G (IgG) Fc binding
protein, and the putative plasma proteins transthyretin, human
Table 2. Characteristics of 26 identified proteins differentially expressed in tumor tissue compared with healthy mucosa, polyp
mucosa, and cancer mucosa (Cont’d)
Expression in
healthy mucosa
Fold change Expression in
polyp mucosa
Fold change Expression in
tumor mucosa
Fold change P
0.141 8 0.125 9 0.12 9 9.7
10
5
1.853 -2.3 1.692 -2.1 2.997 -3.8 8.8
10
4
6.876 -2.3 5.381 -1.8 7.257 -2.4 2.3
10
3
0.594 3.4 0.56 3.6 0.765 2.7 5.5
10
7
9.158 -3.6 14.158 -5.51 8.004 -3.1 7.47
10
5
0.365 -5.2 0.809 -11.6 0.322 -4.6 1.9
10
3
1.45 -4.9 2.005 -6.7 1.76 -5.9 1.99
10
5
1.408 -5.1 2.064 -7.5 1.42 -5.2 6.24
10
5
1.467 -3.3 1.868 -4.2 1.03 -2.3 1.08
10
3
1.869 -1.8 2.24 -2.2 2.469 -2.4 8.05
10
3
0.226 2.6 0.188 3.2 0.199 3 6.8
10
5
0.667 -4.5 0.952 -6.5 0.547 -3.7 4.47
10
6
0.467 -4.1 0.649 -5.7 0.339 -3 1.6
10
6
0.058 6.8 0.09 4.4 0.188 2.1 2.3
10
4
0.462 2.5 0.424 2.7 0.494 2.3 5.3
10
7
0.738 -2.5 0.806 -2.7 0.551 -1.8 1.0
10
3
0.821 -4 0.568 -2.7 0.639 -3.1 7.9
10
3
1.888 -3.5 1.613 -3 1.336 -2.5 4.63
10
4
1.285 -2.9 1.054 -2.4 1.083 -2.4 2.2
10
3
0.135 3.1 0.143 2.9 0.181 2.3 2.1
10
3
1.839 -1.6 1.661 -1.5 2.677 -2.3 1.6
10
3
1.127 1.7 0.991 1.9 1.024 1.9 4.3
10
4
0.241 1.7 0.252 1.6 0.217 1.9 3.1
10
4
0.68 -6.5 0.516 -5 0.621 -6 1.37
10
6
2.628 1.6 2.658 1.5 2.568 1.6 1.3
10
3
0.867 2 0.873 2 1.044 1.7 1.3
10
5
Proteomic Analysis of Colorectal Mucosa
www.aacrjournals.org
6557
Cancer Res 2006; 66: (13). July 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
serum albumin and APO A1. S100A11 (calgizzarin), stathmin, 14-3-3
h, glycyl-tRNA synthetase, and ubiquitin thiolesterase all showed
evidence of increased expression in tumor compared with
morphologically normal mucosa.
Differential expression of proteins in mucosa of patients
with and without neoplasia. The expression of 61 proteins
differed significantly between healthy mucosa and all of the other
tissue types (polyp mucosa, cancer mucosa, and tumor tissue).
Moreover, a total of 206 proteins differed significantly between
healthy mucosa and polyp mucosa. Sixteen positively identified
proteins with expression in polyp mucosa differing significantly
compared with healthy mucosa are listed in Table 3. In contrast
with tumor tissue, in which there was reduced expression of
cytokeratins, three proteins identified as CK8 and two identified
as CK9 were overexpressed in polyp mucosa compared with
healthy mucosa. Two isoforms of a1-antitrypsin precursor were
identified; one was markedly increased in expression and the other
was significantly reduced, an effect which may reflect differing post-
translational modification of this protein in polyp mucosa. Of the
seven identified proteins that were underexpressed in polyp mucosa
compared with healthy mucosa (Table 3), two proteins, desmin
and transgelin, are cytoskeleton-associated proteins. Other proteins
showing evidence of underexpression in polyp mucosa included
calvasculin, 14-3-3 e, and 70K thyroid antigen fragment (Ku protein).
There was further evidence of increased expression of cytoker-
atins in cancer mucosa compared with healthy mucosa (Fig. 1B;
Table 4). Seven proteins positively identified as cytokeratins (CK8,
CK9, and CK20) were overexpressed but a2-actin and other
proteins associated with the cytoskeleton (desmin, transgelin, and
vimentin) were all underexpressed. As in polyp mucosa, there
was evidence for modified expression of members of the serpin
superfamily of protease inhibitors. One isoform of a1-antitrypsin
precursor (24831) was increased by 4.7-fold, but again this was
associated with a reduction in a second isoform of the same
protein (163130). There was increased expression of the related
proteins, elastase inhibitor and maspin. Other proteins listed in
Table 4 are associated with the regulation of a variety of cellular
functions including proliferation and apoptosis.
Discussion
ThemultivariateanalysisillustratedinFig.1C provides
confirmation that colorectal tumor tissue can be distinguished
from morphologically normal mucosa on the basis of protein
expression. This is not unexpected, given the many histologic,
metabolic, and cytokinetic differences between normal and
neoplastic tissues, and our findings are consistent with a number
of other proteomics studies in which tumor tissue has been
Figure 2. Mass spectrometric data (mass and intensity) and sequences identified by peptide mass fingerprinting for two spots (117903 and 3476) identified as
h-actin. The relative positions and intensities of the spots are shown in detail from Fig. 1A . The two spots apparently differ because of a single peptide (highlighted in
3476) that is absent in 117903.
Cancer Research
Cancer Res 2006; 66: (13). July 1, 2006
6558
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
compared with flat mucosa from the same patients (9, 11, 12).
However, it is also clear that the three sources of morphologically
normal mucosa do not form a single coherent group; of particular
interest is the fact that the cluster of samples derived from patients
without polyps or cancer is clearly separated from both sets of
patients with neoplastic lesions. This observation contradicts
the conventional, but largely unexamined, assumption that the
morphologically normal mucosa of cancer patients is also
functionally normal. However, individuals with a history of multiple
colorectal polyps or cancer are well known to be at a greater risk
of developing another cancer at sites remote from the original
lesion. This implies that the whole mucosal field of such individuals
has undergone functional changes that make it persistently
vulnerable to neoplasia. Chen et al. (6) established that, for a
panel of genes, the expression profiles measured in polyp-free
mucosa differed significantly between APC
min
and wild-type mice
and between human patients with and without colorectal cancer.
A later study from the same group reported consistent differences
in gene expression in subjects with a family history of cancer
compared with those without (18). Our present results are
consistent with these findings and show that the changes in gene
expression associated with increased vulnerability to colorectal
cancer encompass a larger number of genes than previously
reported and extend to the level of protein expression. Although it
was not our primary intention to conduct a comprehensive
analysis of all the proteins showing differential expression in polyp
and cancer mucosa, some of our observations may shed light on
the functional changes taking place in the mucosal field of
individuals at increased risk of bowel cancer.
Epithelial cells contain 20 different cytokeratins, numbered 1 to
8 (type II CKs) and 9 to 20 (type I CKs); cytokeratins 1 to 6 are
characteristic of squamous epithelia whereas the columnar
epithelia of the gastrointestinal tract typically contain CKs 7, 8,
and 18 to 20 (19). In the present study, we observed differential
expression of seven isoforms of CK8, four of CK19, and one each of
CK9 and CK20. All four isoforms of CK19 were underexpressed in
tumor tissue compared with morphologically normal mucosa,
regardless of source, and one of the spots identified as CK9 was
absent from tumor samples (Table 2). In contrast, four CK8
isoforms and both CK9s were overexpressed in polyp mucosa
relative to normal mucosa (Table 3) and seven CK8 isoforms, one of
CK9 and one of CK20, were overexpressed in cancer mucosa
relative to healthy mucosa (Table 4). The increased abundance of
cytokeratins in cancer mucosa is highlighted in Fig. 1B. Overall,
these findings indicate that expression of CK8 increases in the
morphologically normal mucosa as the adenoma-carcinoma
sequence progresses, with multiple isoforms differing slightly in
mass and charge.
In this complex situation, the increased abundance of certain
isoforms may be balanced by reductions in others so that total
amounts of protein remain the same. A full characterization of the
subtle changes in peptide structure and abundance that we have
identified presents a considerable technical challenge, which is
beyond the scope of the present study, although further work to
identify their structure and cellular localization is certainly
warranted.
Apart from the maintenance of normal epithelial architecture,
cytokeratins have functional roles in epithelial physiology. CK8-null
mice develop chronic inflammation of the colon (20) and have
impaired electrolyte and fluid transport (21). Mutations affecting
the structure of CK8 in humans also lead to epithelial fragility in
the gut and may play a role in some types of inflammatory bowel
disease (22). The colitis of CK8-null mice is due to a primary
epithelial defect rather than an immune deficiency, and colonic
bacteria are required for its development (23). Thus, CK-8 may be
directly involved in the defense of the mucosal epithelium against
proinflammatory stimuli that contribute to neoplastic change (24).
a1-Antitrypsin [a1 proteinase inhibitor (API)] is a member of
the serine-protease superfamily (25) and an anti-inflammatory
protein, the main function of which is to prevent degradation of
connective tissues by opposing the activity of endogenous
proteases (26). In the present study, four distinct spots were
Table 3. Characteristics of 15 proteins differentially expressed in polyp mucosa compared with healthy mucosa
Spot no. Protein name Score Accession
no.
Theo.
pI
Theo.
MW
Act.
pI
Act.
MW
Expression
in healthy
mucosa
Expression
in polyp
mucosa
Fold
change
P
3476 h-Actin 178 P60709 5.29 42,052 5.25 42,893 51.2 71.73 1.4 1.3
10
2
24831 a1-antitrypsin precursor 85 P01009 5.37 46,878 5.02 58,483 0.629 2.04 3.1 2.1
10
3
163130 a1-Antitrypsin precursor 168 P01009 5.37 46,878 5.03 58,483 2.045 0.596 3.5 6.6
10
5
18889 Calcyclin 88 P06703 5.33 10,230 5.05 9,671 6.29 8.32 1.3 2.2
10
2
12155 Calvasculin 49 P26447 5.85 11,949 5.19 10,727 0.208 0.103 23.1
10
2
68702 Cytokeratin 8 75 P05787 5.52 53,510 5.04 51,491 0.167 0.534 3.2 9.1
10
3
64844 Cytokeratin 8 116 P05787 5.52 53,510 5.10 52,913 0.195 0.686 3.5 1.9
10
3
65803 Cytokeratin 9 82 P35527 5.14 62,178 5.06 62,008 0.026 0.135 5.2 1.7
10
2
12691 Cytokeratin 9 76 P35527 5.14 62,178 4.81 50,192 0.365 0.81 2.2 4.5
10
2
63453 Desmin 70 P17661 5.21 53,429 4.86 48,541 0.794 0 3.2
10
10
2887 Fibrinogen g chain,
isoform g-A precursor
79 P02679 5.70 49,465 5.63 49,356 0.553 0.36 1.5 3.8
10
2
17391 IgG Fc binding protein 59 O95784 5.56 81,017 5.31 132,499 0.667 0.952 1.4 2.1
10
2
17076 Transgelin-2
(SM22-a homologue)
59 P37802 8.45 22,417 5.22 18,242 0.205 0.067 31.6
10
2
19586 14-3-3 e 53 P62258 4.63 29,326 4.68 30,803 0.158 0.072 2.2 7.0
10
3
72917 70K thyroid
antigen fragment
66 P12956 6.23 69,953 5.14 32,121 0.169 0 2.1
10
5
Proteomic Analysis of Colorectal Mucosa
www.aacrjournals.org
6559
Cancer Res 2006; 66: (13). July 1, 2006
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
positively identified as API precursor, which is a 52-kDa poly-
peptide that subsequently becomes glycosylated to form the
55-kDa secreted protein. One spot (24831) was significantly
overexpressed in both polyp mucosa and cancer mucosa relative
to healthy mucosa, but expression in tumor tissue was not
significantly different from healthy mucosa. A second isoform
(17654) was overexpressed in cancer mucosa only and two others
were unchanged or underexpressed. Overall, the data suggest that
modified expression of API isoforms in the morphologically normal
mucosal field occurs during development of neoplasia. API has
previously been shown to be synthesized by hepatocytes, macro-
phages, and intestinal epithelial cells (27), and gastric cancer has
been reported to be associated with high levels of API in the gastric
juice (28). It is also detectable in the feces of both healthy subjects
and patients with inflammatory bowel disease (29), and in vitro
evidence suggests that it is secreted in response to certain
proinflammatory cytokines (30). There are also reports suggesting
that high levels of API expression in sporadic colorectal tumors are
associated with poor prognosis (31). A second serpin, elastase
inhibitor, also showed evidence of increased expression in cancer
mucosa compared with healthy mucosa. BLAST searches con-
firmed that both proteins were members of the serpin superfamily
but a comparison of their sequences established that they were
products of different genes, with 31.4% homology across the amino
acid sequence.
A third member of the serine protease family, maspin, was
shown to be overexpressed in tumor and cancer mucosa relative to
healthy mucosa. Maspin is an unusual serpin in that it lacks
Table 4. Characteristics of 37 identified proteins differentially expressed in cancer mucosa compared with healthy mucosa
Spot no. Protein name Score Accession
no.
Theo.
pI
Theo.
MW
Act.
pI
Act.
MW
Expression
in healthy
mucosa
Expression
in cancer
mucosa
Fold
change
P
19076 a2-Actin 104 P62736 5.23 42,381 5.30 43,815 2.997 1.46 2.1 9.9
10
3
17372 a2-Actin 167 P62736 5.23 42,381 5.22 43,570 12.4 7.3 1.7 1.1
10
2
3476 h-Actin 178 P60709 5.29 42,052 5.25 42,893 51.23 63.95 1.3 4.3
10
2
24831 a1-Antitrypsin precursor 85 P01009 5.37 46,878 5.02 58,483 0.629 2.97 4.7 3.1
10
5
163130 a1-Antitrypsin precursor 168 P01009 5.37 46,878 5.03 58,483 2.045 0.969 2.1 3.0
10
3
50128 ATPase h-chain 98 P06576 5.26 56,525 5.09 54,474 0 0.209 2.5
10
4
18889 Calcyclin 88 P06703 5.33 10,230 5.05 9,671 6.29 9.21 1.5 3.4
10
3
12155 Calvasculin 49 P26447 5.85 11,949 5.19 10,727 0.208 0.034 6.1 2.5
10
4
3013 Calpain 69 P04632 5.05 28,469 4.95 27,853 0.187 0.31 1.7 1.1
10
2
66557 Cytokeratin 8 102 P05787 5.52 53,510 5.06 48,199 0 0.357 8.1
10
4
68702 Cytokeratin 8 75 P05787 5.52 53,510 5.04 51,491 0.167 0.869 5.2 8.5
10
4
64844 Cytokeratin 8 116 P05787 5.52 53,510 5.10 52,913 0.195 0.888 4.6 2.0
10
3
68667 Cytokeratin 8 136 P05787 5.52 53,510 5.21 50,928 0.168 0.918 5.5 2.3
10
3
88139 Cytokeratin 8 130 P05787 5.52 53,510 5.28 52,781 0.076 0.709 9.3 2.5
10
3
162428 Cytokeratin 8 154 P05787 5.52 53,510 5.13 48,714 0.018 0.079 4.4 2.9
10
3
163121 Cytokeratin 8 101 P05787 5.52 53,510 5.06 47,359 0.322 0.743 2.3 3.0
10
2
65803 Cytokeratin 9 82 P35527 5.14 62,178 5.06 62,008 0.026 0.125 4.8 2.5
10
2
80605 Cytokeratin 20 126 P35900 5.60 48,599 5.38 49,415 0.154 0.581 3.8 3.8
10
3
63453 Desmin 70 P17661 5.21 53,429 4.86 48,541 0.794 0.274 2.9 1.1
10
5
10872 Fatty acid-binding protein 132 P07148 6.60 14,256 6.02 13,502 5.002 12.452 2.5 4.3
10
3
2887 Fibrinogen g chain,
isoform g-A precursor
79 P02679 5.70 49,465 5.63 49,356 0.553 0.219 2.5 2.2
10
4
90505 Hemopexin 61 P02790 6.55 52,385 5.65 65,289 0 0.295 9.4
10
4
11118 Leukocyte elastase inhibitor 153 P30740 5.90 42,829 5.94 42,750 0.289 0.847 2.9 1.0
10
3
66836 Mannose-6-phosphate isomerase 76 P34949 5.63 47,065 5.57 44,917 0.301 0.191 1.6 9.9
10
3
86052 Maspin 76 P36952 5.72 42,586 5.75 42,325 0.058 0.188 3.3 2.3
10
2
146954 Peroxiredoxin 2 (thiol-specific
antioxidant protein;
natural killer
cell-enhancing factor B)
82 P32119 5.67 21,918 5.24 24,131 0.259 2.033 7.8 7.4
10
3
11873 Rho GDi, ly 88 P52566 5.10 23,031 5.10 28,896 0.952 0.58 1.6 7.5
10
3
16534 Sarcomeric tropomyosin n;TPM1-n 67 AAT68294 4.65 32,688 4.70 40,372 2.171 1.298 1.7 0.4
10
2
18169 Secernin 2 54 AAH17317 5.44 46,989 5.54 44,512 0.692 0.347 22.3
10
3
18889 S100A6 (calcyclin) 88 P06703 5.33 10,230 5.05 9,671 6.29 9.21 1.5 3.4
10
3
12155 S100A4 (calvasculin) 94 P26447 5.85 11,949 5.19 10,727 0.208 0.034 6.1 2.5
10
4
17076 Transgelin-2 (SM22-a homologue) 59 P37802 8.45 22,417 5.22 18,242 0.205 0.016 12.8 1.8
10
4
13202 Vimentin 58 P08670 5.06 53,545 4.73 46,273 0.259 0.064 42.5
10
2
72917 70K thyroid antigen frag 66 P12956 6.23 69,953 5.14 32,121 0.169 0.056 34.2
10
3
19586 14-3-3 e 53 P62258 4.63 29,326 4.68 30,803 0.158 0 1.9
10
4
18349 14-3-3 protein ~/y (protein kinase C
inhibitor protein 1, KCIP-1)
199 P63104 4.73 27,899 4.72 28,504 5.42 4.68 1.2 3.1
10
2
17784 21K tumor protein 93 P13693 4.84 19,697 4.84 25,485 0.867 1.04 1.2 1.2
10
2
Cancer Research
Cancer Res 2006; 66: (13). July 1, 2006
6560
www.aacrjournals.org
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
protease inhibitory activity and is classified as a tumor suppressor
gene. It has been shown to induce apoptosis in tumor cells via Bax
(32) and Bcl-2 (33), but in prostate cancer the tumor suppressor
function is associated with increased cell adhesion, thereby
suppressing cell mobility and malignant behavior (34). Over-
expression of maspin in colorectal tumor tissue has previously
been described (35) and a recent report suggests that this is
particularly associated with microsatellite instability (36). The
present study seems to be the first in which the expression of
maspin in morphologically normal mucosa has been described.
Several other cytoplasmic proteins with diverse regulatory
functions were shown to be differentially regulated in polyp and
cancer mucosa. The S100 proteins are calcium-activated signaling
molecules involved in cell proliferation, differentiation, and
cytoskeletal dynamics. In the present study, S100A11 (calgizzarin)
was overexpressed in tumor tissue compared with healthy mucosa,
polyp mucosa, and cancer mucosa. Three S100 proteins, including
S100A11, were previously identified by Chaurand et al. (37) as
tumor-specific markers of colorectal neoplasia. Stulik et al. (11)
also identified overexpression of S100A11 in their proteomic
analysis of human colorectal tumors. We observed statistically
significant overexpression of S100A6 (calcyclin) in both polyp
mucosa and cancer mucosa compared with healthy mucosa (Tables
2 and 3) whereas S1004A (calvasculin) was underexpressed in these
tissues. Stulik et al. (11) reported a statistically significant
correlation between the development of colorectal neoplasia and
the expression of different isoforms of S1006A (38), and Bronckart
et al. (39) also reported characteristic patterns of expression of
S100 proteins, including reduced expression of S100A4, associated
with colorectal dysplasia and neoplasia. A high expression of
S100A4 has been reported to be positively associated with risk of
metastasis of colorectal tumors (40) and of many other tumor types
(41). The present study shows that S100A4 is expressed in healthy
mucosa, albeit at a low level, and that reduced expression was
associated with the development of neoplasia.
Overall, the present study provides evidence for progressive
changes in protein expression patterns in the colonic mucosal field,
associated with the development of neoplastic lesions at distant
sites. This observation has a number of important implications.
The use of proteomic techniques to characterize tumors is
becoming increasingly common and many groups have described
studies in which tumor tissue was compared with supposedly
normal mucosa obtained from the same patient. The present study
shows that this is a misleading strategy if it is assumed that the
paired mucosal sample is equivalent to the healthy mucosa of
disease-free individuals. This is well illustrated by the example of
liver fatty acid binding protein, which was reported recently by
Lawrie et al. (42) to be reduced consistently in colorectal tumor
tissue compared with adjacent normal colon. This observation is
consistent with our own study in which the level of liver fatty acid
binding protein expression in cancer mucosa was 3.9-fold higher
than in tumor tissue (P = 0.0004). However, the level in cancer
mucosa was also f2.5-fold higher compared with healthy mucosa
(P = 0.004) whereas there was no statistically significant difference
between expression levels in tumor tissue and healthy mucosa.
The existence of characteristic patterns of protein expression
associated with enhanced vulnerability to neoplasia provides
opportunities for development of biomarkers of increased risk.
Kinzler and Vogelstein (43) proposed that the adverse effects of
certain dietary factors on the risk of colorectal cancer were
attributable not to food-borne carcinogens but to hypothetical
effects they described as chronic ‘‘irritation,’’ causing the colorectal
mucosa to enter a perpetual state of tissue regeneration. Under
these conditions, the survival of cells carrying preneoplastic
mutations would be favored and the vulnerability to cancer would
increase. Since Kinzler and Vogelstein’s review, evidence for the
importance of apoptosis as a protective mechanism against
carcinogenesis has grown (5, 44), but little direct biological evidence
for the putative state of chronic tissue regeneration has emerged.
The results of the present study do not provide conclusive evidence
that the mucosal field defect involves a heightened state of tissue
repair but the observed changes in cytoskeletal proteins and serine
protease inhibitors may be consistent with this hypothesis. Some
of the differences in protein expression that we have observed may
reflect altered gene expression occurring as a consequence of
aberrant CpG island methylation (2, 45). Recognition and further
characterization of the field effect will provide a framework on
which to build hypotheses for future research on the role of diet as
an etiologic factor in the development of colorectal cancer.
Acknowledgments
Received 2/10/2006; revised 4/6/2006; accepted 4/25/2006.
Grant support: Food Standards Agency (UK) and the Biotechnology and Biological
Sciences Research Council.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank Drs. Mike Naldrett and Andrew Bottrill for the MALDI-T of analysis and
Wendy Bal, Julie Coaker, and Catherine Lamb for technical support.
Proteomic Analysis of Colorectal Mucosa
www.aacrjournals.org
6561
Cancer Res 2006; 66: (13). July 1, 2006
References
1. Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic
alterations during colorectal-tumor development. N
Engl J Med 1988;319:525–32.
2. Feinberg AP, Ohlsson R, Henikoff S. The epigenetic
progenitor origin of human cancer. Nat Rev Genet 2006;
7:21–33.
3. Mills SJ, Shepherd NA, Hall PA, et al. Proliferative
compartment deregulation in the non-neoplastic colonic
epithelium of familial adenomatous polyposis. Gut 1995;
36:391–4.
4. Gerdes H, Gillin JS, Zimbalist E, et al. Expansion of the
epithelial cell proliferative compartment and frequency
of adenomatous polyps in the colon correlate with the
strength of family history of colorectal cancer. Cancer
Res 1993;53:279–82.
5. Martin C, Connelly A, Keku TO, et al. Nonsteroidal
anti-inflammatory drugs, apoptosis, and colorectal
adenomas. Gastroenterology 2002;123:1770–7.
6. Chen LC, Hao CY, Chiu YS, et al. Alteration of gene
expression in normal-appearing colon mucosa of
APC(min) mice and human cancer patients. Cancer
Res 2004;64:3694–700.
7. Wulfkuhle JD, Liotta LA, Petricoin EF. Proteomic
applications for the early detection of cancer. Nat Rev
Cancer 2003;3:267–75.
8. Tracy RP, Wold LE, Currie RM, et al. Patterns for
normal colon mucosa and colon adenocarcinoma
compared by two-dimensional gel electrophoresis. Clin
Chem 1982;28:915–9.
9. Anderson KM, Baranowski J, Olson L, et al. The
distribution of acidic coomassie blue-stained proteins
from uninvolved human liver, hepatoma, normal colon,
primary colon cancer, and colon metastases to the liver,
determined by two-dimensional protein electrophoresis.
J Surg Oncol 1983;24:184–91.
10. Alfonso P, Nunez A, Madoz-Gurpide J, et al.
Proteomic expression analysis of colorectal cancer by
two-dimensional differential gel electrophoresis. Pro-
teomics 2005;5:2602–11.
11. Stulik J, Koupilova K, Osterreicher J, et al. Protein
abundance alterations in matched sets of macroscopi-
cally normal colon mucosa and colorectal carcinoma.
Electrophoresis 1999;20:3638–46.
12. Friedman DB, Hill S, Keller JW, et al. Proteome
analysis of human colon cancer by two-dimensional
difference gel electrophoresis and mass spectrometry.
Proteomics 2004;4:793–811.
13. Perkins DN, Pappin DJ, Creasy DM, et al. Probability-
based protein identification by searching sequence
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Cancer Research
Cancer Res 2006; 66: (13). July 1, 2006
6562
www.aacrjournals.org
databases using mass spectrometry data. Electrophore-
sis 1999;20:3551–67.
14. Zar ZH. Biostatistical analysis. 4th ed. New Jersey:
Prentice-Hall; 1999.
15. Brown D, Rothery P. Models in biology: mathematics,
statistics and computing. Chichester: John Wiley and
Sons; 1994.
16. Glare EM, Divjak M, Bailey MJ, et al. h-Actin and
GAPDH housekeeping gene expression in asthmatic
airways is variable and not suitable for normalising
mRNA levels. Thorax 2002;57:765–70.
17. Pokutta S, Weis WI. The cytoplasmic face of cell
contact sites. Curr Opin Struct Biol 2002;12:255–62.
18. Hao CY, Moore DH, Wong P, et al. Alteration of gene
expression in macroscopically normal colonic mucosa
from individuals with a family history of sporadic colon
cancer. Clin Cancer Res 2005;11:1400–7.
19. Moll R. Molecular diversity of cytokeratins: signifi-
cance for cell and tumor differentiation. Acta Histochem
Suppl 1991;41:117–27.
20. Baribault H, Penner J, Iozzo RV, et al. Colorectal
hyperplasia and inflammation in keratin 8-deficient
FVB/N mice. Genes Dev 1994;8:2964–73.
21. Toivola DM, Krishnan S, Binder HJ, et al. Keratins
modulate colonocyte electrolyte transport via protein
mistargeting. J Cell Biol 2004;164:911–21.
22. Owens DW, Wilson NJ, Hill AJ, et al. Human keratin
8 mutations that disturb filament assembly observed in
inflammatory bowel disease patients. J Cell Sci 2004;117:
1989–99.
23. Habtezion A, Toivola DM, Butcher EC, et al. Keratin-
8-deficient mice develop chronic spontaneous Th2
colitis amenable to antibiotic treatment. J Cell Sci
2005;118:1971–80.
24. Clevers H. At the crossroads of inflammation and
cancer. Cell 2004;118:671–4.
25. Silverman GA, Bird PI, Carrell RW, et al. The serpins
are an expanding superfamily of structurally similar but
functionally diverse proteins. Evolution, mechanism of
inhibition, novel functions, and a revised nomenclature.
J Biol Chem 2001;276:33293–6.
26. Sun Z, Yang P. Role of imbalance between neutrophil
elastase and a1-antitrypsin in cancer development and
progression. Lancet Oncol 2004;5:182–90.
27. Perlmutter DH, Daniels JD, Auerbach HS, et al.
The a1-antitrypsin gene is expressed in a human
intestinal epithelial cell line. J Biol Chem 1989;264:
9485–90.
28. Lee K, Kye M, Jang JS, et al. Proteomic analysis
revealed a strong association of a high level of a1-
antitrypsin in gastric juice with gastric cancer. Proteo-
mics 2004;4:3343–52.
29. Mizon C, Becuwe C, Balduyck M, et al. Qualitative
study of fecal a1-proteinase inhibitor in normal subjects
and patients with Crohn’s disease. Clin Chem 1988;34:
2268–70.
30. Faust D, Raschke K, Hormann S, et al. Regulation of
a1-proteinase inhibitor release by proinflammatory
cytokines in human intestinal epithelial cells. Clin Exp
Immunol 2002;128:279–84.
31. Karashima S, Kataoka H, Itoh H, et al. Prognostic
significance of a-1-antitrypsin in early stage of colorec-
tal carcinomas. Int J Cancer 1990;45:244–50.
32. Liu J, Yin S, Reddy N, et al. Bax mediates the
apoptosis-sensitizing effect of maspin. Cancer Res 2004;
64:1703–11.
33. Zhang W, Shi HY, Zhang M. Maspin overexpression
modulates tumor cell apoptosis through the regulation
of Bcl-2 family proteins. BMC Cancer 2005;5:50–61.
34. Abraham S, Zhang W, Greenberg N, et al. Maspin
functions as tumor suppressor by increasing cell
adhesion to extracellular matrix in prostate tumor cells.
J Urol 2003;169:1157–61.
35. Song SY, Lee SK, Kim DH, et al. Expression of maspin
in colon cancers: its relationship with p53 expression
and microvessel density. Dig Dis Sci 2002;47:1831–5.
36. Bettstetter M, Woenckhaus M, Wild PJ, et al. Elevated
nuclear maspin expression is associated with micro-
satellite instability and high tumour grade in colorectal
cancer. J Pathol 2005;205:606–14.
37. Chaurand P, DaGue BB, Pearsall RS, et al. Profiling
proteins from azoxymethane-induced colon tumors at
the molecular level by matrix-assisted laser desorp-
tion/ionization mass spectrometry. Proteomics 2001;1:
1320–6.
38. Stulik J, Osterreicher J, Koupilova K, et al. Differential
expression of the Ca
2+
binding S100A6 protein in
normal, preneoplastic and neoplastic colon mucosa.
Eur J Cancer 2000;36:1050–9.
39. Bronckart Y, Decaestecker C, Nagy N, et al.
Development and progression of malignancy in human
colon tissues are correlated with expression of specific
Ca(2+)-binding S100 proteins. Histol Histopathol 2001;
16:707–12.
40. Takenaga K, Nakanishi H, Wada K, et al. Increased
expression of S100A4, a metastasis-associated gene, in
human colorectal adenocarcinomas. Clin Cancer Res
1997;3:2309–16.
41. Garrett SC, Varney KM, Weber DJ, et al. S100A4,
a mediator of metastasis. J Biol Chem 2006;281:
677–80.
42. Lawrie LC, Dundas SR, Curran S, et al. Liver fatty acid
binding protein expression in colorectal neoplasia. Br J
Cancer 2004;90:1955–60.
43. Kinzler KW, Vogelstein B. Lessons from hereditary
colorectal cancer. Cell 1996;87:159–70.
44. Johnson IT. Mechanisms and possible anticarcino-
genic effects of diet related apoptosis in colorectal
mucosa. Nutr Res Rev 2001;14:229–56.
45. Belshaw NJ, Elliott GO, Williams EA, et al. Methyl-
ation of the ESR1 CpG island in the colorectal mucosa is
an ‘‘all or nothing’’ process in healthy human colon, and
is accelerated by dietary folate supplementation in the
mouse. Biochem Soc Trans 2005;33:709–11.
Research.
on June 3, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
... An important limitation of our study is associated with the use of normal mucosa samples. Although they were taken at least 20 cm away from the tumor and showed no microscopic abnormalities, genetic and protein aberrations may already be present in morphologically normal mucosa [92,93]. However, these samples can still be used as corresponding control samples to overcome differences in the physiological and genetic background. ...
Article
Full-text available
Cancer stem cells (CSC) play one of the crucial roles in the pathogenesis of various cancers, including colorectal cancer (CRC). Although great efforts have been made regarding our understanding of the cancerogenesis of CRC, CSC involvement in CRC development is still poorly understood. Using bioinformatics and RNA-seq data of normal mucosa, colorectal adenoma, and carcinoma (n = 106) from GEO and TCGA, we identified candidate CSC genes and analyzed pathway enrichment analysis (PEI) and protein–protein interaction analysis (PPI). Identified CSC-related genes were validated using qPCR and tissue samples from 47 patients with adenoma, adenoma with early carcinoma, and carcinoma without and with lymph node metastasis and were compared to normal mucosa. Six CSC-related genes were identified: ANLN, CDK1, ECT2, PDGFD, TNC, and TNXB. ANLN, CDK1, ECT2, and TNC were differentially expressed between adenoma and adenoma with early carcinoma. TNC was differentially expressed in CRC without lymph node metastases whereas ANLN, CDK1, and PDGFD were differentially expressed in CRC with lymph node metastases compared to normal mucosa. ANLN and PDGFD were differentially expressed between carcinoma without and with lymph node metastasis. Our study identified and validated CSC-related genes that might be involved in early stages of CRC development (ANLN, CDK1, ECT2, TNC) and in development of metastasis (ANLN, PDGFD).
... One of the limitations of our study is related to normal samples, which were taken at least 20 cm away from the tumour and showed no microscopic abnormalities. However, genetic and protein aberrations may already be present in morphologically normal mucosa [69,70]. Despite certain limitations, these samples may be used as corresponding control samples to overcome differences in the genetic background. ...
Article
Full-text available
Significant progress has been made in the last decade in our understanding of the pathogenetic mechanisms of colorectal cancer (CRC). Cancer stem cells (CSC) have gained much attention and are now believed to play a crucial role in the pathogenesis of various cancers, including CRC. In the current study, we validated gene expression of four genes related to CSC, L1TD1, SLITRK6, ST6GALNAC1 and TCEA3, identified in a previous bioinformatics analysis. Using bioinformatics, potential miRNA-target gene correlations were prioritized. In total, 70 formalin-fixed paraffin-embedded biopsy samples from 47 patients with adenoma, adenoma with early carcinoma and CRC without and with lymph node metastases were included. The expression of selected genes and microRNAs (miRNAs) was evaluated using quantitative PCR. Differential expression of all investigated genes and four of six prioritized miRNAs (hsa-miR-199a-3p, hsa-miR-335-5p, hsa-miR-425-5p, hsa-miR-1225-3p, hsa-miR-1233-3p and hsa-miR-1303) was found in at least one group of CRC cancerogenesis. L1TD1, SLITRK6, miR-1233-3p and miR-1225-3p were correlated to the level of malignancy. A negative correlation between miR-199a-3p and its predicted target SLITRK6 was observed, showing potential for further experimental validation in CRC. Our results provide further evidence that CSC-related genes and their regulatory miRNAs are involved in CRC development and progression and suggest that some them, particularly miR-199a-3p and its SLITRK6 target gene, are promising for further validation in CRC.
... Additionally, high post-surgical levels of circulating caspase-cleaved K18 fragments (M30) were associated with earlier cancer recurrence, possibly indicating systemic residual tumor load similar to K20 (Ausch et al., 2009). Increased K8 expression is associated with tumor burden, as K8 expression increases in both polyp and cancer mucosa (Wauters et al., 1995;Polley et al., 2006), and K8 degradation fragments have been suggested to accumulate in CRC tissue (Nishibori et al., 1996). However, K8 downregulation in CRC, accompanied by a K20-negative phenotype, was linked to an aggressive phenotype and suggested to indicate epithelial to mesenchymal transition (Knösel et al., 2006). ...
Article
Full-text available
Keratin intermediate filament proteins are major cytoskeletal components of the mammalian simple layered columnar epithelium in the gastrointestinal tract. Human colon crypt epithelial cells express keratins 18, 19 and 20 as the major type I keratins, and keratin 8 as the type II keratin. Keratin expression patterns vary between species, and mouse colonocytes express keratin 7 as a second type II keratin. Colonic keratin patterns change during cell differentiation, such that K20 increases in the more differentiated crypt cells closer to the central lumen. Keratins provide a structural and mechanical scaffold to support cellular stability, integrity and stress protection in this rapidly regenerating tissue. They participate in central colonocyte processes including barrier function, ion transport, differentiation, proliferation and inflammatory signaling. The cell-specific keratin compositions in different epithelial tissues has allowed for the utilization of keratin-based diagnostic methods. Since the keratin expression pattern in tumors often resembles that in the primary tissue, it can be used to recognize metastases of colonic origin. This review focuses on recent findings on the biological functions of mammalian colon epithelial keratins obtained from pivotal in vivo models. We also discuss the diagnostic value of keratins in chronic colonic disease and known keratin alterations in colon pathologies. This review describes the biochemical properties of keratins and their molecular actions in colonic epithelial cells and highlights diagnostic data in colorectal cancer and inflammatory bowel disease patients, which may facilitate the recognition of disease subtypes and the establishment of personal therapies in the future.
... In our study, "normal" samples were taken at least 20 cm away from the tumour and they showed no microscopic abnormalities. However, genetic and protein aberrations may also be present in morphologically normal mucosa [62], although it seems highly unlikely that EMT is activated in such samples. We therefore believe that, despite certain limitations, these samples may be used as corresponding control samples to overcome differences in the genetic background. ...
Article
Full-text available
MicroRNAs of the miR-200 family have been shown experimentally to regulate epithelial-mesenchymal transition (EMT). Although EMT is the postulated mechanism of development and progression of colorectal cancer (CRC), there are still limited and controversial data on expression of miR-200 family and their target genes during CRC cancerogenesis. Our study included formalin-fixed paraffin-embedded biopsy samples of 40 patients (10 adenomas and 30 cases of CRC with corresponding normal mucosa). Expression of miR-141, miR-200a/b/c and miR-429 and their target genes (CDKN1B, ONECUT2, PTPN13, RND3, SOX2, TGFB2 and ZEB2) was analysed using quantitative real-time PCR. Expression of E-cadherin was analysed using immunohistochemistry. All miRNAs were down-regulated and their target genes showed the opposite expression in CRC compared to adenoma. Down-regulation of the miR-200 family at the invasive front in comparison to the central part of tumour was observed as well as a correlation of expression of miR-200b, CDKN1B, ONECUT2 and ZEB2 expression to nodal metastases. Expression of the miR-200 family and SOX2 also correlated with E-cadherin staining. These results suggest that the miR-200 family and their target genes contribute to progression of adenoma to CRC, invasive properties and development of metastases. Our results strongly support the postulated hypotheses of partial EMT and intra-tumour heterogeneity during CRC cancerogenesis.
... Screening of endoscopically normal appearing mucosa from patients with CRN and comparing it to mucosa of non-CRN patients may generate important new insights into identifying subjects with increased risk of developing CRN (risk stratification, predictive biomarker). As such, we and a few others have communicated altered expression of several genes and proteins in normal appearing mucosa from CRN patients [1][2][3][4]. ...
Article
Full-text available
Background: Investigations of colorectal carcinogenesis have mainly focused on examining neoplastic tissue. With our aim of identifying potentially cancer-predisposing molecular compositions, we chose a different approach by examining endoscopically normal appearing colonic mucosa of patients with and without colorectal neoplasia (CRN). Directed by this focus, we selected 18 genes that were previously found with altered expression in colorectal cancer affected mucosa. Methods: Biopsies of colonic mucosa were sampled from 27 patients referred for colonoscopy on suspicion of colorectal disease. Of these, 14 patients had present or previous CRN and the remaining 13 patients served as controls. Using qPCR and Western blot technique, we investigated mRNA and protein expressions. Expressions were investigated for selected kinases in the extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK), the phosphoinositide 3-kinase/Akt, and the Wnt/β-catenin pathways as well as for selected phosphatases and several entities associated with prostaglandin E2 (PGE2) signaling. Colonic mucosal contents of PGE2 and PGE2 metabolites were determined by use of ELISA. Results: We found up-regulation of ERK1, ERK2, Akt1, Akt2, PLA2G4A, prostanoid receptor EP3 and phosphatase scaffold subunit PPP2R1B mRNA expression in normal appearing colonic mucosa of CRN patients compared to controls. Conclusion: Present study supports that even normal appearing mucosa of CRN patients differs from that of non-CRN patients at a molecular level. Especially expression of ERK1 mRNA was increased (p = 0.007) in CRN group. ERK1 may therefore be considered a potential candidate gene as predictive biomarker for developing CRN. Further validation in larger cohorts are required to determine such predictive use in translational medicine and clinics.
Article
Background Colorectal cancer (CRC) has a very high incidence and lethality rate and is one of the most dangerous cancer types. Timely diagnosis can effectively reduce the incidence of colorectal cancer. Changes in para-cancerous tissues may serve as an early signal for tumorigenesis. Comparison of the differences in gene expression between para-cancerous and normal mucosa can help in the diagnosis of CRC and understanding the mechanisms of development. Objectives This study aimed to identify specific genes at the level of gene expression, which are expressed in normal mucosa and may be predictive of CRC risk. Methods A machine learning approach was used to analyze transcriptomic data in 459 samples of normal colonic mucosal tissue from 322 CRC cases and 137 non-CRC, in which each sample contained 28,706 gene expression levels. The genes were ranked using four ranking methods based on importance estimation (LASSO, LightGBM, MCFS, mRMR, and RF) and four classification algorithms (decision tree [DT], K-nearest neighbor [KNN], random forest [RF], and support vector machine [SVM]) were combined with incremental feature selection [IFS] methods to construct a prediction model with excellent performance. Result The top-ranked genes, namely, HOXD12, CDH1, and S100A12, were associated with tumorigenesis based on previous studies. Conclusion This study summarized four sets of quantitative classification rules based on the DT algorithm, providing clues for understanding the microenvironmental changes caused by CRC. According to the rules, the effect of CRC on normal mucosa can be determined.
Article
Background: Individuals with adenomatous colorectal polyps undergo repeated colonoscopy surveillance to identify and remove metachronous adenomas. However, many patients with adenomas do not develop recurrent adenomas. Better methods to evaluate who benefits from increased surveillance are needed. We evaluated the use of altered EVL methylation as a potential biomarker for risk of recurrent adenomas. Methods: Patients with ≥1 colonoscopy had EVL methylation (mEVL) measured with an ultra-accurate methylation-specific droplet digital PCR assay on normal colon mucosa. The association between EVL methylation levels and adenoma or colorectal cancer (CRC) was evaluated using three case/control definitions in three models: unadjusted (model 1), adjusting for baseline characteristics (model 2), and an adjusted model excluding patients with CRC at baseline (model 3). Results: Between 2001 and 2020, 136 patients were included; 74 healthy patients and 62 patients with a history of CRC. Older age, never smoking and baseline CRC were associated with higher levels of mEVL (p≤0.05). Each log10 difference in mEVL was associated with an increased risk of adenoma(s) or cancer at/after baseline for model 1(OR: 2.64 95% CI: 1.09-6.36), and adenoma(s) or cancer after baseline for models 1(OR: 2.01, 95% CI: 1.04-3.90) and model 2 (OR: 3.17, 95% CI: 1.30-7.72). Conclusion: Our results suggest that EVL methylation level detected in the normal colon mucosa has the potential to be a biomarker for monitoring the risk for recurrent adenomas. Impact: These findings support the potential utility of EVL methylation for improving the accuracy for assigning risk for recurrent colorectal adenomas and cancer.
Article
Full-text available
Patients with oral squamous cell carcinoma (OSCC) present significant alterations in their saliva proteome. We have used the shotgun Phage Display (PD) technology to identify candidate proteins that were upregulated in saliva of OSCC by selecting ligands to salivary proteins from a single-chain variable fragment (scFv) PD combinatorial library. After two selection cycles, the highly reactive clone scFv-D09 was able to distinguish saliva of OSCC patients from healthy subjects by enzyme-linked immunosorbent assay (ELISA) with sensitivity and specificity of 96.67%. Additionally, the scFv-D09 clone presented a positive immunostaining for invasive malignant epithelial cells in the connective tissue, keratin pearls in the OSCC, and ducts of salivary glands. We have further identified the target protein as the tropomyosin alpha-4 chain (TPM4) by two-dimensional polyacrylamide gel electrophoresis and mass spectrometry, and its binding to the scFV-D09 was demonstrated by bioinformatics. Briefly, we have identified TPM4 as upregulated salivary protein in patients with OSCC, which plays a central role in stabilizing cytoskeleton actin filaments, probably linked with tumor tissue remodeling. Long-term longitudinal studies are needed to validate TPM4 as a potential marker of a malignant process.
Chapter
Cancer is a major global public health problem. Among different environmental and lifestyle factors contributing to cancer risk, diet is a key one. On the one hand, obesity and increased consumption of red and processed meat, ethanol, sugar and saturated fatty acids are associated with increased cancer risk. On the other hand, consumption of micronutrients such as vitamin D, selenium, zinc, folate and bioactive compounds from fruits and vegetables is associated with decreased risk. Written by an influential, international team of experts, this book presents and discusses current topics on nutrition and cancer prevention. It covers both nutritional influences on different cancers plus specific chapters on the commonly occurring cancers. Nutritional genomics-based studies show that some dietary components modulate carcinogenesis through complex cellular and molecular mechanisms. A better understanding of these different cellular and molecular mechanisms is needed to establish efficient dietary recommendations for cancer prevention. This book will provide such an understanding, serving as an important book for all those working in nutritional health, food science and cancer research.
Article
Full-text available
Inflammatory bowel disease (IBD) is a chronic relapsing/remitting inflammatory illness of the gastrointestinal tract of unknown aetiology. Despite recent advances in decoding the pathophysiology of IBD, many questions regarding disease pathogenesis remain. Genome-wide association studies (GWAS) and knockout mouse models have significantly advanced our understanding of genetic susceptibility loci and inflammatory pathways involved in IBD pathogenesis. Despite their important contribution to a better delineation of the disease process in IBD, these genetic findings have had little clinical impact to date. This is because the presence of a given gene mutation does not automatically correspond to changes in its expression or final metabolic or structural effect(s). Furthermore, the existence of these gene susceptibility loci in the normal population suggests other driving prerequisites for the disease manifestation. Proteins can be considered the main functional units as almost all intracellular physiological functions as well as intercellular interactions are dependent on them. Proteomics provides methods for the large-scale study of the proteins encoded by the genome of an organism or a cell, to directly investigate the proteins and pathways involved. Understanding the proteome composition and alterations yields insights into IBD pathogenesis as well as identifying potential biomarkers of disease activity, mucosal healing, and cancer progression. This review describes the state of the art in the field with respect to the study of IBD and the potential for translation from biomarker discovery to clinical application.
Chapter
Full-text available
Article
Full-text available
There is now ample epidemiological evidence to show that the wide international variations in the incidence of both adenomatous polyps and colo-rectal cancer are linked to diet, but the mechanisms through which particular dietary constituents influence the onset of neoplasia are poorly understood. The crypt epithelial cells of the human gastrointestinal mucosa are amongst the most rapidly proliferating tissues in the body, and those of the colorectum are particularly vulnerable to neoplasia. Within the crypt, continuous division of basally localized stem cells gives rise to daughter cells that may divide once or twice again, before differentiating and migrating to the mucosal surface. The majority of nascent crypt epithelial cells differentiate, become senescent and are shed into the gut lumen, but a small proportion die by apoptosis soon after cell division. Various lines of evidence suggest that these pathways of programmed cell death provide a protective mechanism against induction of neoplasia by removing genetically damaged stem cells before they can divide further and give rise to precancerous lesions. There is evidence that the short-chain fatty acid butyrate and several different classes of food constituents, including some polyunsaturated fatty acids, flavonoids and glucosinolate breakdown products, can regulate the processes of cell proliferation and death in vitro, and in colorectal crypts in vivo. All three classes of food components suppress the emergence of aberrant crypt foci in animal models of carcinogenesis. The cellular mechanisms underlying these phenomena, and their possible significance for human health, are discussed.
Article
Maspin, a serine protease inhibitor (serpin), can suppress tumor growth and metastasis in vivo and tumor cell motility and invasion in vitro . This may occur through maspin-mediated inhibition of pericellular proteolysis. In a recent report, we provided evidence that maspin may also suppress tumor progression by enhancing cellular sensitivity to apoptotic stimuli. To our knowledge, maspin is the only proapoptotic serpin among all of the serpins implicated thus far in apoptosis regulation. The goal of the present study is to identify the specific target molecule(s), the modification of which by maspin renders tumor cells sensitive to chemotherapeutic agents. Our cellular, molecular, and biochemical studies demonstrate an essential role of Bax in the proapoptotic effect of maspin. First, Bax was up-regulated in maspin-transfected prostate and breast tumor cells, whereas the levels of other Bcl-2 family members including Bcl-2, Bcl-xl, and Bak remained unchanged. Second, on apoptosis induction, a greater amount of Bax was translocated from cytosol to mitochondria in maspin-transfected cells. After treatment with a Bax-silencing small interfering RNA, maspin-transfected cells became significantly more resistant to drug-induced apoptosis. Consistently, the release of cytochrome c and Smac/DIABLO from mitochondria was more responsive to apoptosis stimuli in maspin-transfected cells than in the mock-transfected cells. Third, the apoptosis induction of maspin-transfected cells was associated with increased activation of both caspase-8 and caspase-9. However, a caspase-9-specific inhibitor blocked the sensitization effect of maspin in a dose-dependent and time-dependent manner, demonstrating a rate-limiting role for caspase-9. In line with the central role of the Bax-mediated mitochondrial apoptotic pathway, maspin sensitized the apoptotic response of breast and prostate carcinoma cells to various drugs, ranging from death ligands to endoplasmic reticulum stress. The link between maspin and Bax up-regulation explains the loss of maspin-expressing tumor cells in invasive breast and prostate carcinomas. Our data reveal a novel mechanism for tumor suppressive maspin and suggest that maspin may be used as a modifier for apoptosis-based cancer therapy.
Article
Our current results, aimed at the detection of protein abundance alterations that could be associated with the process of colon tumorigenesis, are summarized. The matched sets of macroscopically normal colon mucosa and colorectal carcinoma were examined by a one- or two-dimensional electrophoretic approach and proteins were identified using immunoblotting or mass spectrometry. The following results were observed: The levels of liver fatty acid-binding protein, actin-binding protein/smooth muscle protein 22-alpha and cyclooxygenase 2 were downregulated in colorectal carcinoma compared to normal colon mucosa. Conversely, the expression of a novel variant of heat shock protein70 and several members of the S100 protein family of calcium-binding proteins (two isoforms of S100A9, S100A8, S100A11 and S100A6) were upregulated in transformed colon mucosa. Despite the variations of the levels of expression of given protein among analyzed samples, all quantitative changes were found to be statistically significant (Mann-Whitney test assuming p < or = 0.05). We conclude that the proteomic approach is useful for the study of complex biological events underlying the process of colorectal tumorigenesis.
Article
Several algorithms have been described in the literature for protein identification by searching a sequence database using mass spectrometry data. In some approaches, the experimental data are peptide molecular weights from the digestion of a protein by an enzyme. Other approaches use tandem mass spectrometry (MS/MS) data from one or more peptides. Still others combine mass data with amino acid sequence data. We present results from a new computer program, Mascot, which integrates all three types of search. The scoring algorithm is probability based, which has a number of advantages: (i) A simple rule can be used to judge whether a result is significant or not. This is particularly useful in guarding against false positives. (ii) Scores can be compared with those from other types of search, such as sequence homology. (iii) Search parameters can be readily optimised by iteration. The strengths and limitations of probability-based scoring are discussed, particularly in the context of high throughput, fully automated protein identification.