ArticlePDF Available

CpG Island Methylator Phenotype in Colorectal Cancer

Authors:

Abstract and Figures

Aberrant methylation of promoter region CpG islands is associated with transcriptional inactivation of tumor-suppressor genes in neoplasia. To understand global patterns of CpG island methylation in colorectal cancer, we have used a recently developed technique called methylated CpG island amplification to examine 30 newly cloned differentially methylated DNA sequences. Of these 30 clones, 19 (63%) were progressively methylated in an age-dependent manner in normal colon, 7 (23%) were methylated in a cancer-specific manner, and 4 (13%) were methylated only in cell lines. Thus, a majority of CpG islands methylated in colon cancer are also methylated in a subset of normal colonic cells during the process of aging. In contrast, methylation of the cancer-specific clones was found exclusively in a subset of colorectal cancers, which appear to display a CpG island methylator phenotype (CIMP). CIMP+ tumors also have a high incidence of p16 and THBS1 methylation, and they include the majority of sporadic colorectal cancers with microsatellite instability related to hMLH1 methylation. We thus define a pathway in colorectal cancer that appears to be responsible for the majority of sporadic tumors with mismatch repair deficiency.
Comparison of detection of aberrant methylation by using Southern blotting, MCA, or bisulfite-PCR analysis. (A) Methylation of MINT2 detected by Southern blot analysis. Genomic DNA from normal colon mucosa of an 18-year-old person and the CRC cell line Caco2 was digested with restriction endonucleases (H, HindIII; HS, HindIII SmaI; HX, HindIII XmaI), electrophoresed, blotted, and hybridized with MINT2 probe. Caco2 DNA fails to digest completely with SmaI, indicating methylation of one or both SmaI sites, but cuts down with XmaI, ruling out polymorphisms at the SmaI sites. (B) Semiquantitative feature of MCA. DNAs from the Caco2 cell line and normal colon were mixed in various proportions before MCA, and methylation of MINT2 was analyzed by dot-blot hybridization (Upper). % refers to the relative proportion of Caco2 DNA. The signal intensity of each dot was determined by PhosphorImager densitometry, and it increased linearly relative to the proportion of tumor cells in the mix. (C) Detection of MINT2 methylation by MCA. We blotted 0.1 g of MCA products from colorectal tumors and corresponding normal colon mucosa in duplicate onto a nylon filter and hybridized them with a MINT2 probe. Tumors 391, 467, 874, 709, and 351 are methylated at this site. Sample numbers are shown above each lane. N, normal colon; T, colon tumor. (D) Detection of methylation by bisulfite-PCR. Genomic DNA was treated with bisulfite and amplified with primers specific to MINT2. Twenty percent of the PCR products were digested with BstUI and electrophoresed in 3% agarose gels. Arrows indicate bands reflecting methylation of the BstUI site. BstUI cleaves only the methylated alleles, yielding 115-and 88-bp bands (seen in a CRC cell line, RKO, and tumors 391, 467, 874, and 709). The coexistence of methylated and unmethylated bands reflects the fact that all these tumors are not microdissected and contain various amounts of contaminating normal tissues. Sample numbers are shown above. N, normal colon; T, colon tumor.
… 
Methylation of the MINT clones in CRC and mucosa. (A) Examples of MINT clone methylation in colorectal cancer. MCA products from colon tumors (T) and corresponding normal colon mucosa (N) were blotted on a nylon membrane and hybridized with one of the MINT clones (indicated on the left), as well as a p16 probe. MINT6, 24, and 32 are examples of type A methylation, whereas MINT2, 12, and p16 are examples of type C methylation. Tumors 850 and 874 are CIMP, whereas tumors 835, 872, and 733 are CIMP (see text). Also shown is the Caco2 cell line (right). (B) Examples of age-related methylation in normal colon as detected by MCA using MINT5, 8, and 21 as probes. MCA products from normal colon mucosa from patients of various ages (indicated at the top in years) were hybridized with the probe indicated on the left. The signal intensity for each sample was determined by densitometry, and a ratio of mucosaCaco2 (right) is indicated below each sample as a percentage. In each case, methylation is more prominent in DNA from older individuals. (C) Southern blot analysis of clones showing type A methylation in normal colon. DNA from normal colon mucosa from patients of various ages (top) was digested with HindIII and the methylation-sensitive enzyme SmaI and probed with MINT 5 (Left) and MINT22 (Right). Arrows indicate the methylated alleles. The relative proportion of methylated alleles was determined by densitometry and is indicated below each lane. (D) Bisulfite-PCR analysis of clones showing type A and type C methylation in normal colon. DNA from normal colon mucosa from patients of various ages (top) was bisulfite treated, amplified by PCR, and digested with restriction enzymes specific for sites that are created after bisulfite treatment (if the CpG sites are methylated). The arrows indicate the methylated alleles. Percentage methylation was determined by densitometry and is indicated below each lane. In type A loci, the percentage of methylated alleles from older people (70) was significantly higher than that of younger people (30) (6.4 1.1 vs. 22.9 1.2, P 0.000001 for MINT23; 1.3 0.3 vs. 16.1 4.0, P 0.01 for MINT32). In type C loci no methylation was detected in normal colon mucosa. The loci analyzed and restriction enzymes used are indicated below each gel.
… 
Content may be subject to copyright.
Proc. Natl. Acad. Sci. USA
Vol. 96, pp. 8681–8686, July 1999
Medical Sciences
CpG island methylator phenotype in colorectal cancer
MINORU TOYOTA,NITA AHUJA,MUTSUMI OHE-TOYOTA,JAMES G. HERMAN,STEPHEN B. BAYLIN,
AND JEAN-PIERRE J. ISSA*
The Johns Hopkins Oncology Center, 424 North Bond Street, Baltimore, MD 21231
Communicated by John W. Littlefield, Johns Hopkins University School of Medicine, Baltimore, MD, May 26, 1999 (received for review
February 19, 1999)
ABSTRACT Aberrant methylation of promoter region
CpG islands is associated with transcriptional inactivation of
tumor-suppressor genes in neoplasia. To understand global
patterns of CpG island methylation in colorectal cancer, we
have used a recently developed technique called methylated
CpG island amplification to examine 30 newly cloned differ-
entially methylated DNA sequences. Of these 30 clones, 19
(63%) were progressively methylated in an age-dependent
manner in normal colon, 7 (23%) were methylated in a
cancer-specific manner, and 4 (13%) were methylated only in
cell lines. Thus, a majority of CpG islands methylated in colon
cancer are also methylated in a subset of normal colonic cells
during the process of aging. In contrast, methylation of the
cancer-specific clones was found exclusively in a subset of
colorectal cancers, which appear to display a CpG island
methylator phenotype (CIMP). CIMP tumors also have a
high incidence of p16 and THBS1 methylation, and they
include the majority of sporadic colorectal cancers with
microsatellite instability related to hMLH1 methylation. We
thus define a pathway in colorectal cancer that appears to be
responsible for the majority of sporadic tumors with mis-
match repair deficiency.
In the development of colorectal cancers (CRCs), a series of
tumor-suppressor genes such as APC, p53, and genes on
chromosome 18q (DCC, SMAD2, and DPC4SMAD4) are
inactivated by mutations and chromosomal deletions (1, 2). A
subset of CRCs also show a characteristic mutator phenotype
which causes microsatellite instability (MSI) and mutations at
other gene loci such as TGF
RII (3) and BAX (4). This
phenotype usually results from inactivation of mismatch repair
(MMR) genes such as hMSH2 and hMLH1 (5). Another
molecular defect described in CRC is CpG island (CGI)
methylation. CGIs are short sequences rich in the CpG dinu-
cleotide and can be found in the 5 region of about half of all
human genes (6). Methylation of cytosine within 5 CGIs is
associated with loss of gene expression and has been seen in
physiological conditions such as X chromosome inactivation
and genomic imprinting (7). Aberrant methylation of CGIs has
been detected in genetic diseases such as the fragile-X syn-
drome (8), in aging cells (9), and in neoplasia (10, 11). About
half of the tumor-suppressor genes that have been shown to be
mutated in the germ line of patients with familial cancer
syndromes have also been shown to be aberrantly methylated
in some proportion of sporadic cancers, including Rb, VHL,
p16, hMLH1, and BRCA1 (10, 11). Tumor-suppressor gene
methylation in cancer is usually associated with (i) lack of gene
transcription and (ii) absence of coding region mutation. Thus
it has been proposed that CGI methylation serves as an
alternative mechanism of gene inactivation in cancer.
The causes and global patterns of CGI methylation in human
cancers remain poorly defined. We have previously reported
that aging could play a factor in this process because methyl-
ation of several CGIs could be detected in an age-related
manner in normal colon mucosa as well as in CRC (9). In
addition, aberrant methylation of CGIs has been associated
with the MSI phenotype in CRC (12) as well as specific
carcinogen exposures (13). However, an understanding of
aberrant methylation in CRC has been somewhat limited by
the small number of CGIs analyzed to date. We have used a
recently developed PCR-based methylation screening tech-
nique, methylated CpG island amplification (MCA), to deter-
mine the methylation status of multiple CGIs in a relatively
large number of samples. In this report, we analyzed the
methylation status of 30 new loci and 3 known tumor-
suppressor genes in a panel of 50 primary CRCs and 15 colonic
adenomas. We find that (i) the majority of CGI methylation
events in CRCs are related to incremental hypermethylation in
normal colon as an age-related phenomenon; and (ii) virtually
all the other methylation events occur in a distinct subset of
CRCs and adenomas which appear therefore to have a new
phenotype, which we termed CpG island methylator pheno-
type (CIMP). These data shed additional light on the global
patterns of CGI methylation in human cancer and delineate a
distinct pathway involving tumor-suppressor gene hypermeth-
ylation in the evolution of CRC.
MATERIALS AND METHODS
Samples and Cell Lines. Fifty CRCs and 15 colorectal
adenoma samples were obtained from The Johns Hopkins
Hospital. All patients gave informed consent prior to specimen
collection according to institutional guidelines. CRCs used in
this study were characterized previously for MSI status (12).
Presence or absence of MSI was determined according to strict
criteria, requiring band shifts at both dinucleotide and mono-
nucleotide tracts. Of these, 43 cancers were randomly selected
and 7 cancers had previously been shown to display MSI. Nine
of 43 (21%) randomly selected samples showed MSI. There-
fore, in total, 16 samples were MSI positive and 34 samples
were MSI negative.
Genes Examined. In this study, we examined 30 of 33
differentially methylated CpG islands cloned by MCA as
described (MINT1–33; ref. 14). The remaining three clones
were not examined because of high background or size too
small. In addition, we examined the methylation status of p16,
THBS1, and hMLH1 by MCA (p16) or bisulfite-PCR (THBS1
and hMLH1).
Detection of Aberrant Methylation by MCA. MCA was
performed essentially as described (14). A detailed protocol is
available at http:兾兾www.med.jhu.edumethylationMCA.
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
PNAS is available online at www.pnas.org.
Abbreviations: CRC, colorectal cancer; MSI, microsatellite instability;
CGI, CpG island; MCA, methylated CpG island amplification; CIMP,
CpG island methylator phenotype.
*To whom reprint requests should be addressed. e-mail: jpissa@jhmi.
edu.
8681
html. Presence or absence of methylation in cancers was
determined by comparing the signals in the tumor vs. normal
lanes. Each sample was blotted in duplicate. Each filter
included mixtures of positive control (Caco2) and a negative
control (normal colon mucosa from an 18-year-old individual).
All 30 MINT clones, as well as p16 (15) were examined by
MCA.
Bisulfite-PCR. Bisulfite treatment and PCR reactions were
performed as described previously (16, 17). A detailed proto-
col is available at http:兾兾www.med.jhu.edumethylation
protocols.html. In total, 12 MINT clones (MINT 1, 2, 4, 6, 7,
11, 12, 23, 25, 29, 31, 32), as well as THBS1, and hMLH1 were
examined for methylation status in this way. Primer sequences,
conditions for PCR, and restriction enzymes used are available
at http:兾兾 www.med.jhu.edumethylationprimers.htm.
Southern Blot Analysis. Five micrograms of DNA was
digested with 20–100 units of restriction enzymes as specified
by the manufacturer (New England Biolabs). DNA fragments
were separated by agarose gel electrophoresis and transferred
to a nylon membrane (Zeta-probe, Bio-Rad). Filters were
hybridized with
32
P-labeled probes and washed at 65°C with 2
SSC0.1% SDS for 10 min twice, and 0.1 SSC0.1% SDS for
20 min. Filters were then exposed to a phosphor screen for
24–72 hr and analyzed by using a PhosphorImager (Molecular
Dynamics).
RESULTS
Methylation Analysis of Multiple CpG Islands by Using
MCA and Bisulfite-PCR. We have recently developed a PCR-
based method termed MCA (14). In MCA, the methylation
status of CRC and normal colon mucosa samples can be
determined by the presence or absence of a hybridization
signal. Using MCA coupled with representational difference
analysis, we have isolated 33 sequences (MINT1–33) differ-
entially methylated in Caco2, a CRC cell line, and have
confirmed the MCA results by Southern blot analysis. These
included 29 CGIs, and several fragments were identical to
known gene sequences. Thus, they appear to be fairly repre-
sentative of hypermethylation events in cancer. The cloning of
a large number of CGIs from a CRC cell line allowed us to
study the global patterns of hypermethylation in this neoplasm.
To determine the methylation status of these clones in
primary tissues, we have used MCA for 30 of the 33 clones (3
clones could not be accurately studied because of high back-
ground (MINT19 and MINT29) or small size (MINT33). As
shown in Fig. 1, MCA provides semiquantitative methylation
data and yields results that are essentially identical to Southern
blot analysis or bisulfite-PCR. In 12 cases (MINT1, 2, 4, 6, 7,
11, 12, 23, 25, 29, 31, 32), MCA results were confirmed by
bisulfite-PCR for all samples, and the two techniques were
concordant in 98% of the cases (examples in Figs. 1 and 2).
Of the 30 clones analyzed, 26 (87%) were also found to be
methylated in some primary CRCs (examples in Figs. 1–3).
The four clones methylated only in the cell line Caco2 were (i)
MINT14, a LINE element; (ii) MINT14 and MINT18, se-
quences that had a very low CpG frequency and did not qualify
as CpG Islands; and (iii) MINT16 (intron 1 of
-Tubulin). The
other 26 clones all qualified as CpG islands on the basis of the
criteria 200 bp, GC content 50%, and CpGGpC 0.6
(18). Thus, almost all (26 of 27) nonrepetitive CpG islands
recovered from the Caco2 cell line were also methylated in
some primary tumors.
Two Types of Methylation in CRC. By examining the
methylation status of several known genes in colorectal tu-
mors, we have previously demonstrated that some genes tend
to be methylated in an age-dependent manner in normal colon
(9). In the present study of 26 CpG islands, we find that
hypermethylation patterns in CRC fell into two distinct cate-
gories. A majority of the clones (1926, 73%) were found to be
frequently methylated (average 75%, ranging from 30% to
100% of the tumors) in the tumors tested, and a slight amount
of methylation was also detected in normal colon mucosa (Fig.
2A). For all of these clones, the normal colon mucosa obtained
from young patients showed less methylation compared with
the normal mucosa from older patients (Fig. 2 B–D). For this
age-related methylation, identical results were obtained by
MCA, Southern blot analysis, and bisulfite-PCR analysis,
suggesting that it is not related to the technique used to study
methylation. Thus, the majority of CGIs hypermethylated in
CRC are methylated in normal colon mucosa as well, in an
age-related manner. We propose to name this methylation
type A for aging-specific methylation.
The remaining 7 clones were methylated exclusively in CRC:
no methylation was observed in normal colon by any technique
(see Figs. 1 and 2), and their frequency of methylation was
significantly lower than type A methylation (ranging from 10%
to 50%, see below). We propose to name this type of meth-
ylation type C for cancer-specific. There was no significant
FIG. 1. Comparison of detection of aberrant methylation by using
Southern blotting, MCA, or bisulfite-PCR analysis. (A) Methylation of
MINT2 detected by Southern blot analysis. Genomic DNA from
normal colon mucosa of an 18-year-old person and the CRC cell line
Caco2 was digested with restriction endonucleases (H, HindIII; HS,
HindIII SmaI; HX, HindIII XmaI), electrophoresed, blotted, and
hybridized with MINT2 probe. Caco2 DNA fails to digest completely
with SmaI, indicating methylation of one or both SmaI sites, but cuts
down with XmaI, ruling out polymorphisms at the SmaI sites. (B)
Semiquantitative feature of MCA. DNAs from the Caco2 cell line and
normal colon were mixed in various proportions before MCA, and
methylation of MINT2 was analyzed by dot-blot hybridization (Upper).
% refers to the relative proportion of Caco2 DNA. The signal intensity
of each dot was determined by PhosphorImager densitometry, and it
increased linearly relative to the proportion of tumor cells in the mix.
(C) Detection of MINT2 methylation by MCA. We blotted 0.1
gof
MCA products from colorectal tumors and corresponding normal
colon mucosa in duplicate onto a nylon filter and hybridized them with
a MINT2 probe. Tumors 391, 467, 874, 709, and 351 are methylated
at this site. Sample numbers are shown above each lane. N, normal
colon; T, colon tumor. (D) Detection of methylation by bisulfite-PCR.
Genomic DNA was treated with bisulfite and amplified with primers
specific to MINT2. Twenty percent of the PCR products were digested
with BstUI and electrophoresed in 3% agarose gels. Arrows indicate
bands reflecting methylation of the BstUI site. BstUI cleaves only the
methylated alleles, yielding 115- and 88-bp bands (seen in a CRC cell
line, RKO, and tumors 391, 467, 874, and 709). The coexistence of
methylated and unmethylated bands reflects the fact that all these
tumors are not microdissected and contain various amounts of con-
taminating normal tissues. Sample numbers are shown above. N,
normal colon; T, colon tumor.
8682 Medical Sciences: Toyota et al. Proc. Natl. Acad. Sci. USA 96 (1999)
difference in the GC content between type A and type
C clones (average, 57.1% for type A vs. 58.3% for type C,
P 0.36).
CIMP in CRC. We next analyzed in detail the methylation
status of the type A and type C MINT clones in a panel of 50
primary colorectal cancers and 15 adenomatous polyps. The
data obtained by MCA were verified by bisulfite-PCR for 12
clones, and the results obtained with the two techniques were
98% concordant. As mentioned above, all 19 type A clones
were frequently methylated in cancers (average 75%, ranging
from 30% to 100% of the tumors, see examples in Figs. 2 A and
3A and summary in Fig. 4). These frequencies are similar to
what we had previously observed for ER (9), MyoD, and N33
(19). When we considered the methylation status of the 7 type
C clones however, a remarkable pattern emerged (examples in
Figs. 1, 2A, and 3B, summarized in Fig. 4). The 50 cancers fell
into two distinct groups: (i) a group with a high level of type
C methylation, whereby all the tumors had methylation of 3 or
more loci simultaneously (average 5.1 loci per tumor) and (ii)
a group where methylation of any type C clone is extremely
rare (average 0.3 locus per tumor). In sharp contrast, type A
methylation was not significantly different between these two
groups of tumors (Figs. 2A,3A, and 4). Thus, the first group
of tumors appears to display a novel process, which we propose
to call CpG island methylator phenotype (CIMP). These
tumors would then be prone to transcriptional silencing linked
to promoter methylation, and they would have the potential to
inactivate multiple tumor-suppressor genes simultaneously.
We next studied the relationship of CIMP to known clini-
copathological factors in the progression of colorectal neo-
plasia. In this limited series, CIMP did not appear to correlate
with age, gender, or stage of the CRC. However a majority of
proximal (cecum, ascending colon) cancers were affected by
this pathway (1822, 82%), which was significantly different
from distal (descending, recto-sigmoid) cancers (1027, 37%;
P 0.003, Fisher’s exact t test). Furthermore, CIMP was also
found in preneoplastic adenomatous polyps. Overall, 15 ade-
nomas were studied, and 715 were CIMP. In 6 cases, both
FIG. 2. Methylation of the MINT clones in CRC and mucosa. (A) Examples of MINT clone methylation in colorectal cancer. MCA products
from colon tumors (T) and corresponding normal colon mucosa (N) were blotted on a nylon membrane and hybridized with one of the MINT clones
(indicated on the left), as well as a p16 probe. MINT6, 24, and 32 are examples of type A methylation, whereas MINT2, 12, and p16 are examples
of type C methylation. Tumors 850 and 874 are CIMP, whereas tumors 835, 872, and 733 are CIMP (see text). Also shown is the Caco2 cell
line (right). (B) Examples of age-related methylation in normal colon as detected by MCA using MINT5, 8, and 21 as probes. MCA products from
normal colon mucosa from patients of various ages (indicated at the top in years) were hybridized with the probe indicated on the left. The signal
intensity for each sample was determined by densitometry, and a ratio of mucosaCaco2 (right) is indicated below each sample as a percentage.
In each case, methylation is more prominent in DNA from older individuals. (C) Southern blot analysis of clones showing type A methylation in
normal colon. DNA from normal colon mucosa from patients of various ages (top) was digested with HindIII and the methylation-sensitive enzyme
SmaI and probed with MINT 5 (Left) and MINT22 (Right). Arrows indicate the methylated alleles. The relative proportion of methylated alleles
was determined by densitometry and is indicated below each lane. (D) Bisulfite-PCR analysis of clones showing type A and type C methylation
in normal colon. DNA from normal colon mucosa from patients of various ages (top) was bisulfite treated, amplified by PCR, and digested with
restriction enzymes specific for sites that are created after bisulfite treatment (if the CpG sites are methylated). The arrows indicate the methylated
alleles. Percentage methylation was determined by densitometry and is indicated below each lane. In type A loci, the percentage of methylated
alleles from older people (70) was significantly higher than that of younger people (30) (6.4 1.1 vs. 22.9 1.2, P 0.000001 for MINT23;
1.3 0.3 vs. 16.1 4.0, P 0.01 for MINT32). In type C loci no methylation was detected in normal colon mucosa. The loci analyzed and restriction
enzymes used are indicated below each gel.
Medical Sciences: Toyota et al. Proc. Natl. Acad. Sci. USA 96 (1999) 8683
an adenoma and a cancer from the same patient were exam-
ined. In 1 of these, CIMP was detected in both the adenoma
and the cancer; in 3 cases, CIMP was detected in the cancer but
not in the adenoma; in 2 cases, CIMP was detected in neither
the adenoma nor the cancer. These results suggested that
CIMP is an early event in colorectal carcinogenesis.
To determine whether CIMP indeed affects the methylation
status of known tumor-suppressor genes important to tumor
progression, we studied the p16 gene (20), which is one of the
most frequently altered genes in human neoplasms, and the
THBS1 gene, which encodes for an angiogenesis-inhibitor with
tumor-suppressor properties (21, 22). p16 was studied by both
MCA and Southern blot analysis, and THBS1 was studied by
Southern blotting and bisulfite-PCR. There was an excellent
concordance between methylation of both genes and the
presence of CIMP: all 20 cancers and 3 adenomatous polyps
methylated at the p16 CpG island had been classified as
CIMP by using our type C MINT clones (Fig. 4). Similarly,
all 9 tumors hypermethylated at THBS1 also belonged to the
CIMP group (Fig. 4). Thus, CIMP is not limited to the clones
recovered by MCARDA, but truly reflects a methylator
phenotype in these tumors.
Microsatellite Instability Is Linked to CIMP in CRC. In a
previous study (12), we reported a link between microsatellite
instability and a hypermethylator phenotype in sporadic CRC.
Relatively few mutations in mismatch repair genes have been
reported in sporadic MSI cancers, but hMLH1 methylation
has recently been observed in some cases (23–25) and was
suggested to be a primary cause of the MSI phenotype in
sporadic cancers. To determine the relation between CIMP
and MSI in colorectal cancer, we measured hMLH1 methyl-
ation by using bisulfitePCR in our panel of CRC, which had
also been previously typed for the presence of MSI (Fig. 3).
hMLH1 was studied by bisulfite-PCR only because it does not
have 2 SmaI sites in its CGI. Overall, 16 of 50 (32%) cancers
had evidence of MSI. Among the 29 CIMP cases, 12 had
evidence of hMLH1 methylation, suggesting that hMLH1 is
one of the targets of global hypermethylation in CRC. All of
these 12 tumors had MSI. By contrast, hMLH1 methylation
was detected in only one of the 21 CIMPcases. These data
establish a strong link between the CIMP phenotype, hMLH1
methylation, and MSI in CRC. Two lines of evidence suggest
that MSI may follow, and be caused by, CIMP and hMLH1
methylation. First, CIMP is detectable in about half of colonic
adenomas, but none of these tumors have hMLH1 methylation,
and MSI is rare in this preneoplastic lesion (26, 27). Second,
CIMP is not simply caused by mismatch repair defects because,
MSI is absent in more than half of the CIMP cases, and
CIMP was absent in 4 of the 16 cancers with MSI. Overall,
these data suggest that, in sporadic CRC, the majority (1216,
75%) of cases with MSI may be caused by CIMP followed by
hMLH1 methylation, loss of hMLH1 expression, and resultant
mismatch repair deficiency (24). Interestingly, mismatch repair
deficiency in colorectal cancer is also clustered in proximal
tumors, similar to CIMP.
DISCUSSION
Recently, several reports have suggested that aberrant meth-
ylation of CGIs may play an important role in cancer devel-
opment (10, 11). However, there is little integrated informa-
tion on aberrant CGI methylation in cancer at multiple loci,
probably because of the lack of a method to detect methylation
in a large number of samples for unselected CGIs throughout
the genome. Furthermore, it has been shown that cultured cell
lines have a high degree of CGI methylation (28) but it was not
known to what extent this reflects methylation in primary
cancers. In the present study, we have been able to address
some of these issues by studying 30 differentially methylated
loci in a panel of colorectal cancers, a study facilitated by the
relatively quantitative and high-throughput features of MCA.
Despite the fact that all sequences were initially recovered
from a colon cancer cell line, 2627 nonrepeated CpG islands
proved to be methylated in some primary colon cancers.
Therefore, there appears to be relatively little ‘‘artifactual’’
methylation in cell lines. Nevertheless, cell lines often have
more frequent hypermethylation of selected loci than do
primary tumors, and it appears probable that this reflects
continuous selection in culture.
Analysis of the 26 clones methylated in primary tumors
revealed two distinct types of hypermethylation in cancer (type
A for aging-specific and type C for cancer-specific), which may
have distinct causes, and different roles in cancer development.
Type A methylation was seen in the majority of these clones:
19 of 26 (73%) clones were methylated in an age-related
manner in normal colon, and hypermethylated at high fre-
quency in colorectal cancer, as we have shown for the ER gene
(9) and others (19, 29). These results suggest that a large
number of CGIs in the human genome are incrementally
methylated during the aging process and, for many genes, this
methylation correlates with reduced gene expression as shown
for ER (9) and Versican (14). Although we do not know the
mechanism of type A methylation, it likely results from
physiological processes rather than a genetic alteration be-
cause (i) it is very frequent and affects large numbers of cells;
(ii) it is present in all individuals, not just patients with cancer;
and (iii) this process is gene and tissue specific (19). Because
the methylation status at a given CGI is thought to be related
to positive (methylator) factors (30–33) and negative (protec-
tor) factors (34–37), it is possible that, for some genes, this
balance slightly favors de novo methylation, and that this is
reflected by progressive hypermethylation after repeated cell
divisions.
In contrast to type A methylation, type C methylation is
relatively infrequent in primary colorectal cancer, and is never
observed in normal colon mucosa. Furthermore, detailed
analysis of type C methylation in CRC revealed a striking
pattern, suggesting the presence of a hypermethylator pheno-
type in a subset of these tumors: CIMPcases are character-
ized by frequent concordant methylation of the type C clones
examined. By contrast, type C methylation is virtually nonex-
istent in tumors without CIMP. This concordance cannot be
due to simple experimental variation or artifacts because (i)
methylation was verified by using separate methods (MCA,
bisulfite-PCR, and Southern blotting); (ii) the concordance
was not limited to the MINT clones, since it also affected the
FIG. 3. Methylation analysis of CpG islands in CRC by using
bisulfite-PCR. Bisulfite-treated DNA from CRC with (Left) or without
(Right) CIMP and paired normal colon mucosa were amplified and
digested with restriction enzymes that cleave only the methylated CpG
sites. The arrows indicate the methylated alleles. The loci analyzed and
restriction enzymes used are indicated below each gel. MINT6 is an
example of type A methylation, whereas hMLH1 exemplifies type C
methylation.
8684 Medical Sciences: Toyota et al. Proc. Natl. Acad. Sci. USA 96 (1999)
p16, THBS1, and hMLH1 genes; and (iii) there was no signif-
icant difference in type A methylation between CIMP and
CIMP tumors. Through its ability to silence multiple genes
simultaneously, CIMP would then be functionally equivalent
to genetic instability, resulting in the rapid accumulation of
molecular alterations with a potential to accelerate the neo-
plastic process. Because many genes are potential candidates
for inactivation through promoter methylation (10, 11), CIMP
may have profound pathophysiological consequences in neo-
plasia through inactivation of tumor-suppressor genes, metas-
tasis-suppressor genes, angiogenesis inhibitors, and others. In
fact, our data suggest that CIMP could also result in mismatch
repair deficiency through methylation and inactivation of the
hMLH1 promoter, and it may explain up to 75% of cases of
sporadic CRC with MSI.
The causes of type A and type C methylation are probably
different because the latter is detected only in a limited number
of cases and the genes affected are distinct. This defect could
be either aberrant de novo methylation (through a mutation in
DNA-methyltransferase for example), or loss of protection
against de novo methylation, through the loss of a trans-
activating factor (34, 35). Because DNA-methyltransferase
activity is similar in the two groups (data not shown), we favor
the latter hypothesis. Thus, at least in CRC, it appears likely
that type C methylation (an epigenetic error) is actually caused
by a genetic event that results in an increased chance of
methylating a subset of CGIs. Ironically, this epigenetic defect
may then result in additional genetic lesions through the
induction of mismatch-repair deficiency.
On the basis of these data, we propose the following
speculative model integrating CGI methylation into CRC
development (Fig. 5). In this model, CGI methylation plays
two distinct roles, and it appears to arise through distinct
mechanisms. Initially, type A methylation arises as a function
of age in normal colorectal epithelial cells. By potentially
affecting genes that regulate the growth andor differentiation
of these cells, such methylation could account, in part, for the
hyperproliferative state that is thought to precede tumor
formation in the colon (38). Such hyperproliferation is known
to arise with age in colorectal epithelium (39, 40) and to be
marked in patients with colorectal cancer. Furthermore, mod-
ulation of type A methylation may provide one possible
explanation for the reduction in CRC tumorigenesis by reduc-
ing levels of DNA-methyltransferase (41). A second major role
for CGI methylation appears later, possibly at the transition
between small and large adenomas in the colon (on the basis
of 15 adenomas presented here and preliminary data on 50
additional cases). This methylation (type C) affects only a
FIG. 4. Methylation profile of colorectal cancer. The methylation
status of all type C MINT clones, as well as p16, THBS1, and hMLH1,
was determined in a panel of primary CRC and adjacent mucosa. None
of the normal tissues studied showed any degree of methylation for
these genes. Also shown are representative type A clones (Right). Each
column represents a separate gene locus (indicated on top). Each row
is a primary CRC (samples above the bold solid line) or adenomatous
polyp (below the bold solid line). Black rectangles, methylated tumor;
FIG. 5. A model integrating CGI methylation in colorectal carci-
nogenesis. See text for details.
white rectangles, unmethylated tumor. MSI denotes the presence of
microsatellite instability and was not determined for the adenomas. In
the Center are known genes methylated in CRC (p16, THBS1,
hMLH1).
Medical Sciences: Toyota et al. Proc. Natl. Acad. Sci. USA 96 (1999) 8685
subset of tumors, which then evolve along a pathway of global
hypermethylation. We propose then that CIMP leads to cancer
development through the simultaneous inactivation of multi-
ple tumor-suppressor genes such as p16, and induction of
mismatch repair deficiency through inactivation of hMLH1.
This model may be applicable to most human malignancies. In
fact, we have observed evidence for type A and type C
methylation in brain tumors (42), and we have preliminary
evidence suggesting the presence of CIMP in multiple types of
neoplasms, including stomach cancers, brain tumors, and
hematopoietic malignancies. Deciphering the mechanisms un-
derlying these phenomena should facilitate the early detection,
prevention, and therapy of various neoplasms.
We thank Dr. Stanley Hamilton for providing the colon tumor
specimens and Dr. Bert Vogelstein for reviewing the manuscript. This
work was supported by grants from the National Institutes of Health
(National Cancer Institute Grants CA77045 and CA54396 and Colon
Cancer Spore Grant CA62924). M.T. is a Postdoctoral Fellow of the
Japan Society for the Promotion of Science. N.A. is supported by
National Institutes of Health Training Grant 1-T32-DK07713. J.-P.J.I.
is a Kimmel Foundation Scholar.
1. Kinzler, K. W. & Vogelstein, B. (1996) Cell 87, 159–170.
2. White, R. L. (1998) Cell 92, 591–592.
3. Markowitz, S., Wang, J., Myeroff, L., Parsons, R., Sun, L.,
Lutterbaugh, J., Fan, R. S., Zborowska, E., Kinzler, K. W.,
Vogelstein, B., et al. (1994) Science 268, 1336–1338.
4. Rampino, N., Yamamoto, H., Ionov, Y., Li, Y., Sawai, H., Reed,
J. C. & Perucho, M. (1997) Science 275, 967–969.
5. Peltomaki, P. & de la Chapelle, A. (1997) Adv. Cancer Res. 71,
93–119.
6. Bird, A. P. (1986) Nature (London) 321, 209–213.
7. Latham, K. E. (1996) Trends Genet. 112, 134–138.
8. Hansen, R. S., Gartler, S. M., Scott, C. R., Chen, S. H. & Laird,
C. D. (1992) Hum. Mol. Genet. 8, 571–578.
9. Issa, J.-P., Ottaviano, Y. L., Celano, P., Hamilton, S. R., David-
son, N. E. & Baylin, S. B. (1994) Nat. Genet. 7, 536–540.
10. Baylin, S. B., Herman, J. G., Graff, J. R., Vertino, P. M. & Issa,
J.-P. (1998) Adv. Cancer Res. 72, 141–196.
11. Jones, P. A. (1996) Cancer Res. 56, 263–267.
12. Ahuja, N., Mohan, A. L., Li, Q., Stolker, J. M., Herman, J. G.,
Hamilton, S. R., Baylin, S. B. & Issa, J.-P. (1997) Cancer Res. 57,
3370–3374.
13. Issa, J.-P., Baylin, S. B. & Belinsky, S. A. (1996) Cancer Res. 56,
3655–3658.
14. Toyota, M., Ho, C., Ahuja, N., Jair, K.-W., Li, Q., Ohe-Toyota,
M., Baylin, S. B. & Issa, J.-P. J. (1999) Cancer Res. 59, 2307–2312.
15. Herman, J. G., Merlo, A., Mao, L., Lapidus, R. G., Issa, J.-P.,
Davidson, N. E., Sidransky, D. & Baylin, S. B. (1995) Cancer Res.
55, 4525–4530.
16. Sadri, R. & Hornsby, P. J. (1996) Nucleic Acids Res. 24, 558–559.
17. Xiong, Z. & Laird, P. W. (1997) Nucleic Acids Res. 25, 2532–2534.
18. Gardiner-Garden, M. & Frommer, M. (1987) J. Mol. Biol. 196,
261–282.
19. Ahuja, N., Li, Q., Mohan, A. L., Baylin, S. B. & Issa, J.-P. (1998)
Cancer Res. 58, 5489–5494.
20. Kamb, A. (1995) Trends Genet. 11, 136–140.
21. Dameron, K. M., Volpert, O. V., Tainsky, M. A. & Bouck, N.
(1994) Science 265, 1582–1584.
22. Sheibani, N. & Frazier, W. A. (1995) Proc. Natl. Acad. Sci. USA
92, 688692.
23. Kane, M. F., Loda, M., Gaida, G. M., Lipman, J., Mishra, R.,
Goldman, H., Jessup, J. M. & Kolodner, R. (1996) Cancer Res.
57, 808811.
24. Herman, J. G., Umar, A., Polyak, K., Graff, J. R., Ahuja, N., Issa,
J.-P. J., Markowitz, S., Willson, J. K., Hamilton, S. R., Kinzler,
K. W., et al. (1998) Proc. Natl. Acad. Sci. USA 95, 68706875.
25. Cunningham, J. M., Christensen, E. R., Tester, D. J., Kim, C. Y.,
Roche, P. C., Burgart, L. J. & Thibodeau, S. N. (1998) Cancer Res.
58, 3455–3460.
26. Samowitz, W. S. & Slattery, M. L. (1997) Gastroenterology 112,
1516–1519.
27. Konishi, M., Kikuchi-Yanoshita, R., Tanaka, K., Muraoka, M.,
Onda, A., Okumura, Y., Kishi, N., Iwama, T., Mori, T., Koike, M.,
et al. (1997) Gastroenterology 111, 307–317.
28. Antequera, F., Boyes, J. & Bird, A. (1990) Cell 62, 503–514.
29. Issa, J.-P., Vertino, P. M., Boehm, C. D., Newsham, I. F. & Baylin,
S. B., (1996) Proc. Natl. Acad. Sci. USA 93, 1757–1762.
30. Mummaneni, P., Bishop, P. L. & Turker, M. S. (1993) J. Biol.
Chem. 268, 552–558.
31. Mummaneni, P., Walker, K. A., Bishop, P. L. & Turker, M. S.
(1995) J. Biol. Chem. 270, 788–792.
32. Magewu, A. N. & Jones, P. A. (1994) Mol. Cell. Biol. 14,
4225–4332.
33. Vertino, P. M., Yen, R. W., Gao, J. & Baylin, S. B. (1996) Mol.
Cell. Biol. 16, 4555–4565.
34. Macleod, D., Charlton, J., Mullins, J. & Bird, A. P. (1994) Genes
Dev. 8, 2282–2292.
35. Brandeis, M., Frank, D., Keshet, I., Siegfried, Z., Mendelsohn,
M., Nemes, A., Temper, V., Razin, A. & Cedar, H. (1994) Nature
(London) 371, 435–438.
36. Turker, M. S. & Bestor, T. H. (1997) Mutat. Res. 386, 119–130.
37. Chen, F. Y., Harris, L. C., Remack, J. S. & Brent, T. P. (1997)
Proc. Natl. Acad. Sci. USA 94, 43484353.
38. Lipkin, M. (1988) Cancer Res. 48, 235–245.
39. Holt, P. R. & Yeh, K. Y. (1988) Gastroenterology 95, 1556–1563.
40. Roncucci, L., Ponz de Leon, M., Scalmati, A., Malagoli, G.,
Pratissoli, S., Perini, M. & Chahin, N. J. (1988) Cancer 62,
2373–2377.
41. Laird, P. W., Jackson-Grusby, L., Fazeli, A., Dickinson, S. L.,
Jung, W. E., Li, E., Weinberg, R. A. & Jaenisch, R. (1995) Cell
81, 197–205.
42. Li, Q., Jedlicka, A., Ahuja, N., Gibbons, M. C., Baylin, S. B.,
Burger, P. C. & Issa, J.-P. J. (1998) Oncogene 16, 3197–3202.
8686 Medical Sciences: Toyota et al. Proc. Natl. Acad. Sci. USA 96 (1999)
... Recent reports have shown that aberrant methylation isn't confined to only a few genes or promoter areas and the majority of cancer cells exhibit abnormal patterns of DNA methylation, such as hypermethylation of gene promoter CpG islands and global demethylation of the genome, as seen in colon cancer. It also can result in the loss of genomic imprinting, which is an epigenetic phenomenon that occurs during the unequal allocation of chromosomes inherited from each parent during embryonic development [37,38,39]. A variety of genes have been demonstrated to be hypermethylated in colorectal tumors, including known TSGs [39] ( Table 1). ...
... It also can result in the loss of genomic imprinting, which is an epigenetic phenomenon that occurs during the unequal allocation of chromosomes inherited from each parent during embryonic development [37,38,39]. A variety of genes have been demonstrated to be hypermethylated in colorectal tumors, including known TSGs [39] ( Table 1). For example, the inactivation of the cyclin-dependent kinase inhibitor P16 through methylation can lead to the disruption of cell-cycle regulation, which may result in a growth advantage for the affected cells [40]. ...
... Age-related methylation? Source: [39]. ...
Article
Full-text available
In the modern age, colon cancer has attained a widespread status, affecting a considerable number of people. It develops due to the progressive accumulation of genetic and epigenetic alterations. While genetic mutations have been extensively studied in the context of colon cancer, emerging evidence highlights the pivotal role of epigenetic alterations in its pathogenesis. These alterations ultimately result in the transformation of normal colonic epithelium into colon adenocarcinoma. Key mechanisms of epigenetic modifications include DNA methylation, histone modification, and nucleosome positioning. Research findings have linked these modifications to the development, progression, or metastasis of tumors. Through the assessment of the colon cancer epigenome, it has been discovered that practically all colorectal cancers (CRCs) display gene methylation abnormalities and changes in miRNA expression. Advancements in this area indicate that epigenetic modifications will likely be commonly used in the near future to direct the prevention and treatment of CRC. The maintenance of genome stability is essential for preserving cellular integrity. The development of CRC is primarily influenced by the loss of genomic stability, which allows for the emergence of new mutations contributing to tumor characteristics. Although genetic mutations have been extensively researched in the realm of colon cancer, recent evidence underscores the pivotal role of epigenetic changes in its pathogenesis. The following types of genomic instability will be discussed: chromosomal instability, microsatellite instability, CpG island methylation phenotype, and aberrant DNA methylation.
... CpG island methylator phenotype (CIMP) is a distinct mechanism via which colorectal cancer advances, marked by an extremely high frequency of methylation at the promoter region of genes. 18 About 50% of human gene promoter regions contain CpG-islands, which are 200-500 base pair lengths of nucleotides. These segments are typically richer in the dinucleotide sequence that consists of cytosine and guanine and are known as CpG-residues. ...
... Due to the significant role CpG islands play in gene regulation, methylation of the CpG islands in promoter regions silences the corresponding genes. 18 The number of methylation markers detected in the colorectal tumor tissue can determine the CIMP status, which can be divided into 2 groups: CIMP-positive or CIMPnegative, or into 3 groups: CIMP-high, CIMP-low, and CIMP-negative. 19 In 20%-30% of all colorectal cancer patients, both MSI and CIN, widespread hypermethylation of the promoter regions of numerous tumor suppressor genes leads to epigenetic silencing. ...
Article
Full-text available
Background: Colorectal cancer is the second cause of cancer mortality and the third most commonly diagnosed cancer worldwide. Current data available implicate epigenetic modulations in colorectal cancer development. The health of the large bowel is impacted by gut microbiome dysbiosis, which may lead to colon and rectum cancers. The release of microbial metabolites and toxins by these microbiotas has been shown to activate epigenetic processes leading to colorectal cancer development. Increased consumption of a ‘Westernized diet’ and certain lifestyle factors such as excessive consumption of alcohol have been associated with colorectal cancer. Purpose: In this review, we seek to examine current knowledge on the involvement of gut microbiota, dietary factors, and alcohol consumption in colorectal cancer development through epigenetic modulations. Methods: A review of several published articles focusing on the mechanism of how changes in the gut microbiome, diet, and excessive alcohol consumption contribute to colorectal cancer development and the potential of using these factors as biomarkers for colorectal cancer diagnosis. Conclusions: This review presents scientific findings that provide a hopeful future for manipulating gut microbiome, diet, and alcohol consumption in colorectal cancer patients’ management and care.
... 3 Numerous studies have aimed to understand the epigenetic landscape of CRC, revealing a complex relationship between DNA methylation patterns and the pathogenesis of CRC. 4 CpG island methylator phenotype (CIMP) is characterized by a distinct pattern of hypermethylation in multiple CpG loci throughout the cancer genome. Initially identified in CRC patients, 5 CIMP has been observed in various cancers. In gliomas, CIMP is predominantly caused by IDH1/2 mutations, which lead to the accumulation of the oncometabolite 2-hydroxyglutarate, a competitive inhibitor of the TET family of DNA dioxygenases. ...
... Utilizing the DREAM platform, we detected the methylation status of 26,370 CpG sites following the initial quality filtering, ensuring a minimum of 30 reads/site in at least 85% of the 115 tumor samples (Table S1), including individual technical replicates to ensure high confidence and high coverage sites. Given that much of the methylation variation is attributed to aging, 5 we used the adjacent normal samples to eliminate agingspecific methylation sites. CpG sites were selected based on high variability (STDEV >20%) of methylation across tumor samples, coupled with low variability (STDEV <10%) and low average (average <2%) methylation in the adjacent normal samples, resulting in the identification of 1,317 cancerspecific CpG sites. ...
Article
Full-text available
The intestinal microbiota is an important environmental factor implicated in CRC development. Intriguingly, modulation of DNA methylation by gut microbiota has been reported in preclinical models, although the relationship between tumor-infiltrating bacteria and CIMP status is currently unexplored. In this study, we investigated tumor-associated bacteria in 203 CRC tumor cases and validated the findings using The Cancer Genome Atlas datasets. We assessed the abundance of Bacteroides fragilis, Escherichia coli, Fusobacterium nucleatum, and Klebsiella pneumoniae through qPCR analysis and observed enrichment of all four bacterial species in CRC samples. Notably, except for E. coli, all exhibited significant enrichment in cases of CIMP. This enrichment was primarily driven by a subset of cases distinguished by high levels of these bacteria, which we labeled as “Superhigh”. The bacterial Superhigh status showed a significant association with CIMP (odds ratio 3.1, p-value = 0.013) and with MLH1 methylation (odds ratio 4.2, p-value = 0.0025). In TCGA CRC cases (393 tumor and 45 adj. normal), bacterial taxa information was extracted from non-human whole exome sequencing reads, and the bacterial Superhigh status was similarly associated with CIMP (odds ratio 2.9, p < 0.001) and MLH1 methylation (odds ratio 3.5, p < 0.001). Finally, 16S ribosomal RNA gene sequencing revealed high enrichment of Bergeyella spp. C. concisus, and F. canifelinum in CIMP-Positive tumor cases. Our findings highlight that specific bacterial taxa may influence DNA methylation, particularly in CpG islands, and contribute to the development and progression of CIMP in colorectal cancer.
... These genes were carefully selected due to their significance in cancer biology and their potential usefulness as targets for diagnosis and treatment purposes [22]. Some of the genes included in the Weissenberg panel are APC (Adenomatous polyposis coli), BRCA1 (Breast cancer 1), CDH1 (Cadherin 1), CDKN2A (Cyclin-dependent kinase inhibitor 2 A), MLH1 (MutL homolog 1), PTEN (Phosphatase and tensin homolog), TP53 (Tumor protein p53) and VHL (Von Hippel-Lindau tumor suppressor) [22,23]. The Weissenberg panel was chosen based on its established significance in previous research related to CRC and SSAP. ...
Article
Full-text available
Colorectal cancer (CRC) originates from pre-existing polyps in the colon. The development of different subtypes of CRC is influenced by various genetic and epigenetic characteristics. CpG island methylator phenotype (CIMP) is found in about 15–20% of sporadic CRCs and is associated with hypermethylation of certain gene promoters. This study aims to find prognostic genes and compare their expression and methylation status as potential biomarkers in patients with serrated sessile adenomas/polyps (SSAP) and CRC, in order to evaluate which, one is a better predictor of disease. This study employed a multi-phase approach to investigate genes associated with CRC and SSAP. Initially, two gene expression datasets were analyzed using R and Limma package to identify differentially expressed genes (DEGs). Venn diagram analysis further refined the selection, revealing four genes from the Weissenberg panel with significant changes. These genes, underwent thorough in silico evaluations. Once confirmed, they proceeded to wet lab experimentation, focusing on expression and methylation status. This comprehensive methodology ensured a robust examination of the genes involved in CRC and SSAP. This study identified cancer-specific genes, with 8,351 and 1,769 genes specifically down-regulated in SSAP and CRC tissues, respectively. The down-regulated genes were associated with cell adhesion, negative regulation of cell proliferation, and drug response. Four highly downregulated genes in the Weissenberg panel, including CACNA1G, IGF2, MLH1, and SOCS1. In vitro analysis showed that they are hypermethylated in both SSAP and CRC samples while their expressions decreased only in CRC samples. This suggests that the decrease in gene expression could help determine whether a polyp will become cancerous. Using both methylation status and gene expression status of genes in the Weissenberg panel in prognostic tests may lead to better prognoses for patients.
... Aberrant DNA methylation, such as the CpG island methylator phenotype (CIMP), is an important oncogenic mechanism of colorectal cancer [19][20][21] and has been reported to be associated with molecular biological features [22,23] and prognosis [24,25]. We focused on this epigenetic factor and performed a comprehensive DNA methylation analysis of patients who received anti-EGFR antibodies as the third-line or later treatment [26]. ...
Article
Full-text available
Purpose The genome-wide DNA methylation status (GWMS) predicts of therapeutic response to anti-epidermal growth factor receptor (EGFR) antibodies in treating metastatic colorectal cancer. We verified the significance of GWMS as a predictive factor for the efficacy of anti-EGFR antibodies in the second-line treatment of metastatic colorectal cancer. Methods Clinical data were obtained from a prospective trial database, and a genome-wide DNA methylation analysis was performed. GWMS was classified into high-methylated colorectal cancer (HMCC) and low-methylated colorectal cancer (LMCC). The patients were divided into subgroups according to the treatment arm (cetuximab plus irinotecan or irinotecan alone) and GWMS, and the clinical outcomes were compared between the subgroups. Results Of the 112 patients, 58 (51.8%) were in the cetuximab plus irinotecan arm, and 54 (48.2%) were in the irinotecan arm; 47 (42.0%) were in the HMCC, and 65 (58.0%) were in the LMCC group regarding GWMS. Compared with the LMCC group, the progression-free survival (PFS) was significantly shortened in the HMCC group in the cetuximab plus irinotecan arm (median 1.4 vs. 4.1 months, p = 0.001, hazard ratio = 2.56), whereas no significant differences were observed in the irinotecan arm. A multivariate analysis showed that GWMS was an independent predictor of PFS and overall survival (OS) in the cetuximab plus irinotecan arm (p = 0.002, p = 0.005, respectively), whereas GWMS did not contribute to either PFS or OS in the irinotecan arm. Conclusions GWMS was a predictive factor for the efficacy of anti-EGFR antibodies in the second-line treatment of metastatic colorectal cancer.
... Epigenetic instability in this pathway is identified by the genome-wide simultaneous hypermethylation of promoter CpG island loci, leading to the inactivation of tumor-suppressor genes or genes related to tumors. The CIMP denotes a subset of CRCs that develop through this pathway of epigenetic instability [21,22]. ...
Article
Full-text available
Synchronous colorectal cancer (sCRC) is characterized by the occurrence of more than one tumor within six months of detecting the first tumor. Evidence suggests that sCRC might be more common in the serrated neoplasia pathway, marked by the CpG island methylator phenotype (CIMP), than in the chromosomal instability pathway (CIN). An increasing number of studies propose that CIMP could serve as a potential epigenetic predictor or prognostic biomarker of sCRC. Therapeutic drugs already used for treating CIMP-positive colorectal cancers (CRCs) are reviewed and drug selections for sCRC patients are discussed.
... The hallmark example of the former in CRC is hypermethylation of the MLH1 promoter, which leads to loss of MLH1 expression and tumour microsatellite instability [8,9]. On a more global scale, a specific and reproducible pattern of genome-wide hypermethylation can occur in CRC termed the CpG island methylator phenotype (CIMP) [10]. ...
Article
Full-text available
While epigenomic alterations are common in colorectal cancers (CRC), few epigenomic biomarkers that risk-stratify patients have been identified. We thus sought to determine the potential of ZNF331 promoter hypermethylation (mZNF331) as a prognostic and predictive marker in colon cancer. We examined the association of mZNF331 with clinicopathologic features, relapse, survival, and treatment efficacy in patients with stage III colon cancer treated within a randomized adjuvant chemotherapy trial (CALGB/Alliance89803). Residual tumour tissue was available for genomic DNA extraction and methylation analysis for 385 patients. ZNF331 promoter methylation status was determined by bisulphite conversion and fluorescence-based real-time polymerase chain reaction. Kaplan-Meier estimator and Cox proportional hazard models were used to assess the prognostic and predictive role of mZNF331 in this well-annotated dataset, adjusting for clinicopathologic features and standard molecular markers. mZNF331 was observed in 267/385 (69.4%) evaluable cases. Histopathologic features were largely similar between patients with mZNF331 compared to unmethylated ZNF331 (unmZNFF31). There was no significant difference in disease-free or overall survival between patients with mZNF331 versus unmZNF331 colon cancers, even when adjusting for clinicopathologic features and molecular marker status. Similarly, there was no difference in disease-free or overall survival across treatment arms when stratified by ZNF331 methylation status. While ZNF331 promoter hypermethylation is frequently observed in CRC, our current study of a small subset of patients with stage III colon cancer suggests limited applicability as a prognostic marker. Larger studies may provide more insight and clarity into the applicability of mZNF331 as a prognostic and predictive marker.
... In addition to hypomethylation, gene hypermethylation can contribute to colorectal tumorigenesis. In 1999, Toyota et al. proposed the role of CIMP in CRC (Toyota et al., 1999). Three main classical pathways have been proposed for the genetic including chromosomal instability (CIN, approximately 80%), microsatellite instability (MSI), and CpG island methylator phenotype (Armaghany et al., 2012). ...
Article
Full-text available
CpG islands near promoters are normally unmethylated despite being surrounded by densely methylated regions. Aberrant hypermethylation of these CpG islands has been associated with the development of various human diseases. Although local genetic elements have been speculated to play a role in protecting promoters from methylation, only a limited number of methylation barriers have been identified. In this study, we conducted an integrated computational and experimental investigation of colorectal cancer methylomes. Our study revealed 610 genes with disrupted methylation barriers. Genomic sequences of these barriers shared a common 41-bp sequence motif (MB-41) that displayed homology to the chicken HS4 methylation barrier. Using the CDKN2A (P16) tumor suppressor gene promoter, we validated the protective function of MB-41 and showed that loss of such protection led to aberrant hypermethylation. Our findings highlight a novel sequence signature of cis-acting methylation barriers in the human genome that safeguard promoters from silencing.
Article
Full-text available
DNA methylation of promoter-associated CpG islands may function as an alternate mechanism of silencing tumor suppressor genes in multiple neoplasias including colorectal cancer. De novo methylation of genes appears to be an early and frequent event in most neoplasias. For the ER and IGF2 genes, we have previously shown that methylation actually begins in the normal colon mucosa as an age-related event and progresses to hypermethylation in cancer. In this study, we have determined the frequency of age-related methylation in normal colonie mucosa among the genes hypermethylated in colorectal cancer. We studied six genes, includ ing N33, MYOD,p¡6, HIC-l, THBSI, and CALCA. The N33 gene showed partial methylation in normal colon mucosa, which was age-related (r = 0.7; P = 0.003 using regression analysis). Adenomas and cancers showed further hypermethylation at this locus. Similarly, the MYOD gene showed age-related methylation in normal colon mucosa (r = 0.7; /' < 0.00001 using regression analysis) and hypermethylation in cancers. Age-related methylation seems to be gene specific, because pI6, THBSI, HIC-l, and CALCA were not affected. Furthermore, this process may also be modulated by tissue-specific factors. Our study suggests that aging is a major contributing factor to hypermethylation in cancer.
Article
Full-text available
Inactivation of the genes involved in DNA mismatch repair is associated with microsatellite instability (MSI) in colorectal cancer. We report that hypermethylation of the 5′ CpG island of hMLH1 is found in the majority of sporadic primary colorectal cancers with MSI, and that this methylation was often, but not invariably, associated with loss of hMLH1 protein expression. Such methylation also occurred, but was less common, in MSI− tumors, as well as in MSI+ tumors with known mutations of a mismatch repair gene (MMR). No hypermethylation of hMSH2 was found. Hypermethylation of colorectal cancer cell lines with MSI also was frequently observed, and in such cases, reversal of the methylation with 5-aza-2′-deoxycytidine not only resulted in reexpression of hMLH1 protein, but also in restoration of the MMR capacity in MMR-deficient cell lines. Our results suggest that microsatellite instability in sporadic colorectal cancer often results from epigenetic inactivation of hMLH1 in association with DNA methylation.
Article
Cytosine methylation at CpG dinucleotides is thought to cause more than one-third of all transition mutations responsible for human genetic diseases and cancer. We investigated the methylation status of the CpG dinucleotide at codon 248 in exon 7 of the p53 gene because this codon is a hot spot for inactivating mutations in the germ line and in most human somatic tissues examined. Codon 248 is contained within an HpaII site (CCGG), and the methylation status of this and flanking CpG sites was analyzed by using the methylation-sensitive enzymes CfoI (GCGC) and HpaII. Codon 248 and the CfoI and HpaII sites in the flanking introns were methylated in every tissue and cell line examined, indicating extensive methylation of this region in the p53 gene. Exhaustive treatment of an osteogenic sarcoma cell line, TE85, with the hypomethylating drug 5-aza-2'-deoxycytidine did not demethylate codon 248 or the CfoI sites in intron 6, although considerable global demethylation of the p53 gene was induced. Constructs containing either exon 7 alone or exon 7 and the flanking introns were transfected into TE85 cells to determine whether de novo methylation would occur. The presence of exon 7 alone caused some de novo methylation to occur at codon 248. More extensive de novo methylation of the CfoI sites in intron 6, which contains an Alu sequence, occurred in cells transfected with a vector containing exon 7 and flanking introns. With longer time in culture, there was increased methylation at the CfoI sites, and de novo methylation of codon 248 and its flanking HpaII sites was observed. These de novo-methylated sites were also resistant to 5-aza-2'-deoxycytidine-induced demethylation. The frequent methylation of codon 248 and adjacent Alu sequence may explain the enhanced mutability of this site as a result of the deamination of the 5-methylcytosine.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Article
The fragile-X syndrome of mental retardation is associated with an expansion in the number of CGG repeats present in the FMR1 gene. The repeat region is within sequences characteristic of a CpG island. Methylation of CpG dinucleotides that are 5' to the CGG repeat has been shown to occur on the inactive X chromosome of normal females and on the X chromosome of affected fragile-X males, and is correlated with silencing of the FMR1 gene. The methylation status of CpG sites 3' to the repeat and within the repeat itself has not previously been reported. We have used two methylation-sensitive restriction enzymes, AciI and Fnu4HI, to further characterize the methylation pattern of the FMR1 CpG island in normal individuals and in those carrying fragile-X mutations. Our results indicate that: (i) CpG dinucleotides on the 3' side of the CGG repeat are part of the CpG island that is methylated during inactivation of a normal X chromosome in females; (ii) the CGG repeats are also part of the CpG island and are extensively methylated as a result of normal X-chromosome inactivation; (iii) similar to normal males, unaffected fragile-X males with small CGG expansions are unmethylated in the CpG island; for affected males, the patterns of methylation are similar to those of a normal, inactive X chromosome; (iv) in contrast to the partial methylation observed for certain sites in lymphocyte DNA, complete methylation was observed in DNA from cell lines containing either a normal inactive X chromosome or a fragile-X chromosome from an affected male.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
CpG islands are normally methylation free in cells of the animal, even when the associated gene is transcriptionally silent. In mouse NIH 3T3 and L cells, however, over half of the islands are heavily methylated. Near identity of the methylated subset in the two cell lines suggested that methylation is confined to genes that are nonessential in culture. In agreement with this, islands at several tissue-specific genes, but not at housekeeping genes, have become methylated in many human and mouse cell lines. At the chromatin level, methylated islands are Mspl resistant compared with their nonmethylated counterparts. We suggest that mutation-like gene inactivation due to CpG island methylation is widespread in many cell lines and could explain the loss of cell type-specific functions in culture.
Article
It is likely that most vertebrate genes are associated with 'HTF islands'--DNA sequences in which CpG is abundant and non-methylated. Highly tissue-specific genes, though, usually lack islands. The contrast between islands and the remainder of the genome may identify sequences that are to be constantly available in the nucleus. DNA methylation appears to be involved in this function, rather than with activation of tissue specific genes.
Article
Cancer of the large bowel is relatively rare in persons younger than 50 years of age, but its incidence increases sharply in persons older than 60 years of age. We thought that the evaluation of colonic cell proliferation, an accurate biomarker of predisposition to colorectal cancer, might help to elucidate the susceptibility of elderly persons to this common malignancy. Accordingly, 30 persons with normal lower endoscopy results were divided into three age groups (30 to 50,51 to 65, and 66 to 90 years of age; Groups 1, 2, and 3, respectively). Samples of rectal mucosa were taken at endoscopic examination, incubated with [3H]thymidine, and processed with standard autoradiographic techniques. At histologic examination, each intestinal hemicrypt was divided into five equal longitudinal compartments from the fundus (compartment 1) to the surface (compartment 5). The number and the position of labeled cells along the crypt were recorded. The total labeling index (LI) (the ratio of labeled cells to total cells) was significantly higher in Group 3 than in the two other groups. Similarly, the LI per crypt compartment in the most superficial portions of the crypts was consistently higher in persons older than 65 years of age (P less than 0.01 at least), indicating an expansion of the proliferative zone to the most superficial portion of the colonic glands. When the proliferative profiles of the three groups of subjects investigated were compared with those of patients with polyps, an almost complete overlap of values was observed between this population at increased risk for cancer and the subjects in Group 3. We conclude that aging is characterized by an overall increase of epithelial cell proliferation in colorectal mucosa and by an upwards expansion of the proliferative compartment, similar to that observed in a population at risk for cancer of the large bowel.
Article
Our previous studies suggested that crypt size enlarged and that proliferation rate might be greater in the small intestine of rats during senescence. Crypt cell numbers and crypt cell proliferation rates, using the vincristine-induced metaphase arrest technique, now have been measured in the colon of aging and young Fischer 344 rats. The proximal colon of 26-28-mo-old unfasted rats had 10% more crypt cells and a higher proliferative rate than 3-4-mo-old young controls. In the distal colon, the crypt cell proliferation rate in aging rats was 56% greater than in the young. A 3-day fast reduced crypt cell proliferation about fourfold in young rats but only by 20% in aging rats. One-day refeeding abruptly increased the crypt cell population and proliferation rate in rats of both age groups. The crypt zone of proliferating cells from aging rats was broader than that seen in young rats. In addition, starvation lowered colonic crypt cell cycling rate much less in aging than in young animals. We conclude that the colons of aging rats demonstrate a hyperproliferative state and a failure to adapt appropriately to changes in food intake. These observations may be relevant to states of altered proliferation that occur in the premalignant colon.