ArticlePDF Available

CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression

Authors:
  • Liquid Genomics, Inc.

Abstract and Figures

The molecular basis of aberrant hypermethylation of CpG islands observed in a subset of human colorectal tumors is unknown. One potential mechanism is the up-regulation of DNA (cytosine-5)-methyltransferases. Recently, two new mammalian DNA methyltransferase genes have been identified, which are referred to as DNMT3A and DNMT3B. The encoded proteins differ from the predominant mammalian DNA methyltransferase DNMT1 in that they have a substantially higher ratio of de novo to maintenance methyltransferase activity. We have used a highly quantitative 5' nuclease fluorogenic reverse transcription-PCR method (TaqMan) to analyze the expression of all three DNA methyltransferase genes in 25 individual colorectal adenocarcinoma specimens and matched normal mucosa samples. In addition, we examined the methylation patterns of four CpG islands [APC, ESR1 (estrogen receptor), CDKN2A (p16), and MLH1] to determine whether individual tumors show a positive correlation between the level of DNA methyltransferase expression and the frequency of CpG island hypermethylation. All three methyltransferases appear to be up-regulated in tumors when RNA levels are normalized using either ACTB (beta-actin) or POLR2A (RNA pol II large subunit), but not when RNA levels are normalized with proliferation-associated genes, such as H4F2 (histone H4) or PCNA. The frequency or extent of CpG island hypermethylation in individual tumors did not correlate with the expression of any of the three DNA methyltransferases. Our results suggest that deregulation of DNA methyltransferase gene expression does not play a role in establishing tumor-specific abnormal DNA methylation patterns in human colorectal cancer.
Content may be subject to copyright.
1999;59:2302-2306. Cancer Res
Cindy A. Eads, Kathleen D. Danenberg, Kazuyuki Kawakami, et al.
Not Associated with DNA Methyltransferase Overexpression
CpG Island Hypermethylation in Human Colorectal Tumors Is
Updated version
http://cancerres.aacrjournals.org/content/59/10/2302
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/59/10/2302.full.html#ref-list-1
This article cites by 29 articles, 16 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/59/10/2302.full.html#related-urls
This article has been cited by 66 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
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
[CANCER RESEARCH 59, 2302–2306, May 15, 1999]
Advances in Brief
CpG Island Hypermethylation in Human Colorectal Tumors Is Not Associated with
DNA Methyltransferase Overexpression
1
Cindy A. Eads, Kathleen D. Danenberg, Kazuyuki Kawakami, Leonard B. Saltz, Peter V. Danenberg, and
Peter W. Laird
2
Departments of Surgery [C. A. E., P. W. L.] and Biochemistry and Molecular Biology [C. A. E., K. D. D., K. K., P. V. D., P. W. L.], University of Southern California, School of
Medicine, Norris Comprehensive Cancer Center, Los Angeles, California 90033, and Memorial Sloan Kettering Cancer Center, New York, New York 10021-6094 [L. B. S.]
Abstract
The molecular basis of aberrant hypermethylation of CpG islands
observed in a subset of human colorectal tumors is unknown. One poten-
tial mechanism is the up-regulation of DNA (cytosine-5)-methyltrans-
ferases. Recently, two new mammalian DNA methyltransferase genes
have been identified, which are referred to as DNMT3A and DNMT3B.
The encoded proteins differ from the predominant mammalian DNA
methyltransferase DNMT1 in that they have a substantially higher ratio of
de novo to maintenance methyltransferase activity. We have used a highly
quantitative 5* nuclease fluorogenic reverse transcription-PCR method
(TaqMan) to analyze the expression of all three DNA methyltransferase
genes in 25 individual colorectal adenocarcinoma specimens and matched
normal mucosa samples. In addition, we examined the methylation pat-
terns offour CpG islands [APC, ESR1 (estrogen receptor), CDKN2A (p16),
and MLH1] to determine whether individual tumors show a positive
correlation between the level of DNA methyltransferase expression and
the frequency of CpG island hypermethylation. All three methyltrans-
ferases appear to be up-regulated in tumors when RNA levels are nor-
malized using either ACTB (
b
-actin) or POLR2A (RNA pol II large
subunit), but not when RNA levels are normalized with proliferation-
associated genes, such as H4F2 (histone H4) or PCNA. The frequency or
extent of CpG island hypermethylation in individual tumors did not
correlate with the expression of any of the three DNA methyltransferases.
Our results suggest that deregulation of DNA methyltransferase gene
expression does not play a role in establishing tumor-specific abnormal
DNA methylation patterns in human colorectal cancer.
Introduction
Vertebrate cytosine-5 DNA methylation occurs in the context of
CpG dinucleotides and is associated with transcriptional inactivity.
CpG islands are areas of high CpG density that are normally unmeth-
ylated. Aberrant de novo methylation of CpG islands is found in a
subset of human colorectal tumors. The abnormal methylation of CpG
islands associated with tumor suppressor genes can lead to transcrip-
tional silencing, inactivating the gene through epigenetic rather than
genetic means (1). When aberrant CpG island hypermethylation oc-
curs in colorectal tumors, it is frequently not restricted to a single CpG
island but affects multiple independent loci (2, 3), reflective of a
widespread deregulation of DNA methylation patterns (1). Although
the molecular basis of such a methylator phenotype is not known, its
global nature suggests a change in trans-acting factors that control the
pattern of distribution of methylated residues. It has recently been
suggested that the nuclear phosphoprotein FOS transforms cells in
part by up-regulating DNMT1 gene expression (4).
DNA methylation results from a methyl transfer reaction performed
by trans-acting enzymes known as DNA methyltransferases. Two
distinct methyl transfer activities can be distinguished, based on the
methylation status of the substrate. Maintenance DNA methyltrans-
ferase activity refers to the conversion of hemimethylated substrates
to a fully methylated state, whereas de novo methyltransferase activity
refers to the new addition of methyl groups at sites that were previ-
ously unmethylated. All known DNA (cytosine-5)-methyltransferases
are able to perform both reactions. The predominant mammalian DNA
(cytosine-5)-methyltransferase, DNMT1, is unusual in that its relative
de novo activity is 1–2 orders of magnitude lower than its mainte-
nance activity (5, 6) Recently, two additional mammalian DNA (cy-
tosine-5)-methyltransferase genes have been identified, which are
referred to as DNMT3A and DNMT3B. These genes differ from
DNMT1 in that the encoded polypeptides DNMT3
a
and DNMT3
b
have approximately equal ratios of de novo DNA methyltransferase
activity:maintenance DNA methyltransferase activity (7). An addi-
tional candidate DNA methyltransferase gene, DNMT2, has been
identified, but the encoded protein has not yet been shown to possess
methyltransferase activity (8–10). DNMT3
a
and DNMT3
b
are
thought to be responsible for the wave of de novo methylation that
occurs during embryogenesis (7). Because the abnormal hypermethy-
lation of CpG islands in colorectal tumors involves the new acquisi-
tion of DNA methylation (de novo methylation), we have investigated
whether transcriptional activation or up-regulation of either DNMT3A
or DNMT3B could be responsible for the methylator phenotype ob-
served in colorectal tumors.
Previous studies have analyzed the expression levels of the DNMT1
gene in human tumors and cell lines, with somewhat conflicting
results (11–15). Some reports documented a substantial increase in the
expression of DNMT1 or of total DNA methyltransferase enzyme
activity in tumor cells, compared to normal counterparts. However,
Lee et al. (14) made the important observation that relative determi-
nations of expression levels can be affected by the gene used for the
normalization of RNA amounts. Normalization with a gene associated
with cell proliferation, such as histone H4 (H4F2) abolished any
statistically significantly higher mean expression of DNMT1 in colo-
rectal tumors compared to matched normal mucosa. One additional
caveat to these studies is that in all cases, mean expression levels were
determined for tumor tissues compared to normal specimens. How-
ever, substantial interindividual variability in DNA methyltransferase
expression exists. None of these studies analyzed CpG island hyper-
methylation frequencies in the same samples to correlate these with
DNA methyltransferase expression or activity.
In this study, we extend the analysis of DNA methyltransferase
expression levels in colorectal tumors in the following ways: (a)we
have analyzed the largest data set used thus far for such an analysis
(25 pairs of tumor and matched normal mucosa); (b) we have used a
Received 1/29/99; accepted 3/30/99.
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.
1
Supported by NIH/National Cancer Institute Grants R01 CA 71716 (to P. V. D.) and
R01 CA 75090 (to P. W. L.).
2
To whom requests for reprints should be addressed, at University of Southern
California/Norris Comprehensive Cancer Center, Mail Stop # 73,, Room 6418, 1441
Eastlake Avenue, Los Angeles, CA 90033. Phone: (323) 865-0650; Fax: (323) 865-0158;
E-mail: plaird@hsc.usc.edu.
2302
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
highly quantitative RT-PCR
3
method (Fig. 1) that is linearly accurate
in serial dilutions over 6 orders of magnitude; (c) we have used four
different genes [ACTB (
b
-actin), H4F2 (histone H4), PCNA, or
POLR2A (RNA pol II large subunit)] for normalization of RNA
levels; (d) we have analyzed the expression levels of two new DNA
methyltransferase genes, DNMT3A and DNMT3B, that have not been
investigated previously; (e) we investigated whether the individual
expression levels of any of the three DNA methyltransferases corre-
late with the de novo methylation of four commonly hypermethylated
CpG islands [APC, ESR1 (estrogen receptor), CDKN2A (p16), and
MLH1].
Materials and Methods
Sample Collection. A total of 25 paired tumor and normal mucosal tissue
samples were obtained from 25 patients with primary colorectal adenocarci-
noma. The patients comprised 16 males and 9 females, ranging in age from
3988 years, with a mean age of 68.8 years. The surgical procedures were
performed in Japan, and a preoperative diagnosis was obtained from a biopsy
and a histological examination. The mucosal distance from the tumor to the
normal specimens was between 10 and 20 cm. Approximately2gofthe
surgically removed tissue were frozen immediately in liquid nitrogen and
stored at 280°C until RNA and DNA isolation. The remaining section of the
sample was fixed with formalin and used for further histological examination
to confirm the diagnosis postoperatively. All histological examinations were
performed after staining with H&E.
Nucleic Acid Isolation. Genomic DNA was isolated by the standard
method of proteinase K digestion and phenol-chloroform extraction (16). Total
RNA was isolated by the single-step guanidinium isothiocyanate method (17).
Quantitative RT-PCR Analysis. The quantitation of mRNA levels was
carried out using a real-time fluorescence detection method as described
previously (18, 19). In brief, after RNA isolation, cDNA was prepared from
each sample as described previously (20). The specific cDNA of interest and
reference cDNA (ACTB, H4F2, PCNA, or POLR2A) were PCR-amplified
separately using an oligonucleotide probe with a 59 fluorescent reporter dye
(6FAM) and a 39 quencher dye (TAMRA; Ref. 21). The 59 to 39 nuclease
activity of Taq DNA polymerase cleaved the probe and released the reporter,
whose fluorescence could be detected by the laser detector of the ABI Prism
7700 Sequence Detection System (Perkin-Elmer Corp., Foster City, CA). After
crossing a fluorescence detection threshold, the PCR amplification results in a
fluorescent signal proportional to the amount of PCR product generated. Initial
template concentration was derived from the cycle number at which the
fluorescent signal crossed a threshold in the exponential phase of the PCR
reaction. Relative gene expression was determined based on the threshold
cycles of the gene of interest and of the internal reference gene. Use of a
reference gene avoids the need to directly quantitate the RNA, which could be
a major source of error for analysis. Several reference samples were included
on each assay plate to verify plate-to-plate consistency. Plates were normalized
to each other using these reference samples, if necessary. Contamination of the
RNA samples by genomic DNA was excluded by an analysis of all RNA
samples without prior cDNA conversion. The PCR amplification was per-
formed using a 96-well optical tray and caps with a 25-
m
l final reaction
mixture consisting of 600 nM each primer; 200 nM probe; 5 units of Ampli-Taq
Gold; 200
m
M each of dATP, dCTP, and dGTP; 400
m
M dUTP; 5.5 mM MgCl
2
;
1 unit of AmpErase uracil N-glycosylase; and 13 TaqMan buffer A containing
a reference dye at 50°C for 2 min and at 95°C for 10 min, followed by 40
cycles at 95°C for 15 s and at 60°C for 1 min. The primer and probe sequences
are listed below. In all cases, the first primer is the forward PCR primer, the
second primer is the TaqMan probe, and the third primer is the reverse PCR
primer.
The primer and probe sequences are as follows: (a) ACTB, TGAGCGCG-
GCTACAGCTT, 6FAM59-ACCACCACGGCCGAGCGG-39 TAMRA, and
CCTTAATGTCACACACGATT; (b) H4F2, CTTAGCCTCAGTGCGAAT-
GCT, 6FAM59-CAGAACCAGAGCACAGCCAAAGCCACTAC-39 TAMRA,
and ACGGTCCCCGGGAGAAT; (c) PCNA, GTGCAAAAGACGGAGT-
GAAATTT, 6FAM59-TGTTTCCATTTCCAAGTTCTCCACTTGCAG-39
3
The abbreviation used is: RT-PCR, reverse transcription-PCR.
Fig. 1. Schematic of quantitative RT-PCR (TaqMan) technology. A, in the PCR
reaction, an oligonucleotide probe tagged with a 59 fluorescent reporter and a 39 quencher
is added in addition to the standard PCR components. The probe is complementary to the
target sequence of interest and anneals during extension. The close proximity of the
quencher to the fluorescent reporter represses fluorescence in the intact probe. As the Taq
polymerase synthesizes the new strand, its 59 to 39 nuclease activity cleaves the probe,
separating the quencher and fluorescent reporter. The fluorescence emitted is proportional
to the amount of product accumulated with each cycle. B, the plot represents a sample
analysis from the experiments shown in Fig. 3. The plot shows the expression values
obtained for three genes (as indicated) in a matched tumor and normal tissue sample. The
horizontal bold line indicates the fluorescence level used for the threshold cycle deter-
mination in this particular example. DRn is defined as the cycle-to-cycle change in the
reporter fluorescence signal normalized to a passive reference fluorescence signal.
2303
DNA METHYLTRANSFERASE EXPRESSION AND HYPERMETHYLATION
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
TAMRA, and ATCGACATTACTTGTCTGTGACAATTTA; (d) POLR2A,
GCCACCCAGATGACCTTGAA, 6FAM59-CCTTCCACTATGCTGGTGT-
GTCTGCCA-39 TAMRA, and GCACACCCAGCGTCACATT; (e) DNMT1,
GGTTCTTCCTCCTGGAGAATGTC, 6FAM59-CCTTCAAGCGCTCCATG-
GTCCTGAA-39 TAMRA, and GGGCCACGCCGTACTG; (f) DNMT3A,
CAATGACCTCTCCATCGTCAAC, 6FAM59-AGCCGGCCAGTGCCCTC-
GTAG-39 TAMRA, and CATGCAGGAGGCGGTAGAA; and (g) DNMT3B,
CCATGAAGGTTGGCGACAA, 6FAM59-CACTCCAGGAACCGTGAGA-
TGTCCCT-39 TAMRA, and TGGCATCAATCATCACTGGATT.
DNA Methylation Analysis. Genomic DNA extracted from tumor and
normal samples was treated with sodium bisulfite as described previously (22).
After conversion, the DNA was amplified by fluorescence-based, real-time
quantitative PCR (as described above, but without the addition of AmpErase).
Two sets of primers and probes designed specifically for bisulfite-converted
DNA were used: (a) a methylated set for the gene of interest [APC, ESR1,
CDKN2A (p16), or MLH1]; and (b) an internal reference set (MYOD1)to
control for input DNA. The methylated primers and the probe were designed
to contain 1–5 CpG dinucleotides, which amplify only fully methylated mol-
ecules. Specificity of the reactions for methylated DNA was confirmed sepa-
rately using DNAs of known methylation status. The internal reference primers
and the probe were designed in a region of the MYOD1 gene that lacks any
CpG dinucleotides to allow for unbiased amplification. Parallel TaqMan PCR
reactions were performed with primers specific for the bisulfite-converted
methylated sequence for a particular locus and with the MYOD1 reference
primers. The ratio between the values obtained in these two TaqMan analyses
was used as a measure of the degree of methylation at that locus. A ratio
greater than or equal to four times the mean ratio for all normal mucosal
samples was classified as methylated (Figs. 2 and 3, F), and a ratio less than
four was regarded as unmethylated (Figs. 2 and 3, E). The primer and probe
sequences are listed below. In all cases, the first primer listed is the forward
PCR primer, the second primer is the TaqMan probe, and the third primer is the
reverse PCR primer.
The primer and probe sequences were as follows: (a) APC, TTATAT-
GTCGGTTACGTGCGTTTATAT, 6FAM59-CCCGTCGAAAACCCGC-
CGATTA-39 TAMRA, and GAACCAAAACGCTCCCCAT; (b) CDKN2A,
AACAACGTCCGCACCTCCT, 6FAM59-ACCCGACCCCGAACCGCG-39
TAMRA, and TGGAATTTTCGGTTGATTGGTT; (c) ESR1, GGCGT-
TCGTTTTGGGATTG, 6FAM59-CGATAAAACCGAACGACCCGACGA-39
TAMRA, and GCCGACACGCGAACTCTAA; (d) MLH1, CTATCGCCGCCT-
CATCGT, 6FAM59-CGCGACGTCAAACGCCACTACG-39 TAMRA, and
CGTTATATATCGTTCGTAGTATTCGTGTTT; and (e) MYOD1, CCAACTC-
CAAATCCCCTCTCTAT, 6FAM59-TCCCTTCCTATTCCTAAATCCAACCT-
AAATACCTCC-39 TAMRA, and TGATTAATTTAGATTGGGTTTAGAGA-
AGGA.
Statistics. TaqMan analyses performed as described above for either RT-
PCR or DNA methylation studies yield values that are expressed as ratios
between two absolute measurements (DNMT:normalization gene for RT-PCR
and CpG island:MYOD1 for DNA methylation analysis). The ratios for each
type of analysis were subsequently normalized such that the mean ratio of the
25 normal samples would equal a value of 1. Consequently, the values for the
tumor samples represent a fold increase or decrease relative to a mean normal
value of 1. Additional statistical manipulations are described in Tables 1 and 2.
Results and Discussion
DNMT1 Expression in Colorectal Tumors. Fig. 2 shows the
relative expression levels of DNMT1 in 25 individual colorectal
adenocarcinoma samples (f) and 25 matched normal mucosal sam-
ples (hatched bars). The expression levels are displayed as ratios
between DNMT1 and four reference genes (ACTB, Fig. 2A; POLR2A,
Fig. 2B; H4F2, Fig. 2C; and PCNA, Fig. 2D) to correct for variations
in the amounts of RNA. The mean expression levels of DNMT1 in
tumors versus normal mucosal samples were calculated using the four
normalization genes (Table 1; Fig. 2). DNMT1 appeared to be up-
regulated approximately 3.9-fold when ACTB or POLR2A were used
for normalization (Table 1; Fig. 2, A and B). However, this apparent
up-regulation was absent when the proliferation-associated genes
H4F2 or PCNA were used for normalization (Table 1; Fig. 2, C and
D). Similar results have been documented previously (14, 23). These
results indicate that either DNMT1 is truly up-regulated in tumors and
is otherwise normally expressed in a proliferation-independent fash-
ion or that DNMT1 gene expression is proliferation dependent, which
accounts for all of its apparent up-regulation in tumors. The latter
scenario seems more likely in view of the requirement for mainte-
nance DNA methylation during S-phase and in light of the reported
proliferation-dependent gene expression of DNMT1 (23–25).
DNMT3A and DNMT3B Expression in Colorectal Tumors.
Fig. 3 shows the relative expression levels of DNMT3A and DNMT3B
in the same samples as described above. The high values seen in some
of the samples have been reproduced multiple times and are therefore
thought to represent valid ratios. The advantage of the use of multiple
normalization genes is apparent from the variable ratios obtained
within individual samples. For instance, the apparent high value for
DNMT3/PCNA seen in sample 23N is due to an abnormally low
expression of PCNA compared to other reference genes in this
Fig. 2. DNMT1 expression in 25 colorectal tumors and matched normal mucosal
controls. RNA levels were measured by quantitative, real-time RT-PCR in 25 matched
normal (z) and tumor (f) colorectal samples. The expression levels are displayed as
ratios between DNMT1 and four reference genes (A, ACTB; B, POLR2A: C, H4F2, and D,
PCNA) to correct for variations in the amounts of RNA. The ratios for each type of
analysis have been normalized such that the mean ratio of the 25 normal samples equals
a value of 1. CpG island hypermethylation status for four different genes (APC, ESR1,
CDKN2A, and MLH1) is shown below each chart. F, the measured level of DNA
methylation at that CpG island was at least 4-fold higher than the mean level in the 25
normal samples; E, the measured level was below four times the mean level of the normal
samples.
2304
DNA METHYLTRANSFERASE EXPRESSION AND HYPERMETHYLATION
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
particular sample and is not due to a high level of DNMT3 gene
expression.
The analysis of DNMT3A and DNMT3B expression yielded similar
results to the analysis of DNMT1 expression. DNMT3A expression in
colorectal tumors appears to be elevated by an average of 2.9- and
2.8-fold when RNA levels are normalized using ACTB and POLR2A,
respectively (Table 1; Fig. 3, A and B). Likewise, DNMT3B levels
appear to be up-regulated by an average of 3.6- and 4.0-fold in a
similar analysis (Table 1; Fig. 3, E and F). However, this up-regula-
tion is absent for both genes when RNA levels are normalized using
Table 2 Analysis of the relationship between CpG island hypermethylation and DNA
methyltransferase expression in individual tumors (ANOVA)
ANOVA was used to compare mean DNMT gene expression between categories of
CpG island hypermethylation frequency. Twelve separate ANOVA calculations were
performed to investigate all combinations of the three DNMT genes and the four normal-
ization genes. The P value indicates the probability that the null hypothesis (no difference
in the mean DNMT gene expression levels in the different categories of CpG island
hypermethylation frequency) is correct.
Normalization gene DNMT1 DNMT3A DNMT3B
Nonproliferation-associated gene
ACTB P 5 0.0659 P 5 0.5288 P 5 0.2216
POLR2A P 5 0.2233 P 5 0.6959 P 5 0.3947
Proliferation-associated gene
H4F2 P 5 0.3603 P 5 0.4338 P 5 0.4500
PCNA P 5 0.7098 P 5 0.7209 P 5 0.8879
Fig. 3. DNMT3A and DNMT3B expression in 25 colorectal tumors and matched normal mucosal controls. The expression levels are displayed as ratios between DNMT3A (A–D)
or DNMT3B (E–H) and four reference genes (A and E, ACTB; B and F, POLR2A: C and G, H4F2; D and H, PCNA) to correct for variations in the amounts of RNA. The ratios for
each type of analysis have been normalized such that the mean ratio of the 25 normal samples equals a value of 1. The CpG island hypermethylation status for four different genes
(APC, ESR1, CDKN2A, and MLH1) is shown below each chart. F, the measured level of DNA methylation at that CpG island was at least 4-fold higher than the mean level in the
25 normal samples; E, the measured level was below four times the mean level of the normal samples.
Table 1 Relative mean expression of DNA methyltransferases in tumor versus normal tissue
The mean expression levels are shown of each of the DNMT genes in tumors versus
normal mucosal samples. The expression levels were first calculated as ratios between the
DNMT gene and one of four reference genes (ACTB, POLR2A, H4F2, and PCNA)as
indicated to correct for variations in the amounts of RNA. The ratios were then normal-
ized, such that the mean ratio of the 25 normal samples would equal a value of 1.
Subsequently, the ratios for the tumor samples were averaged to give the mean values
indicated in the table. Consequently, these values represent an average fold increase or
decrease of DNMT gene expression in tumors relative to a mean value of 1 in normal
mucosal samples. The P value for each average indicates the probability that the null
hypothesis (no difference between tumor and normal) is correct.
Normalization gene DNMT1 DNMT3A DNMT3B
Nonproliferation-associated gene
ACTB 3.9 (P , 0.0001) 2.9 (P 5 0.0006) 3.6 (P 5 0.0004)
POLR2A 3.9 (P , 0.0001) 2.8 (P 5 0.0015) 4.0 (P 5 0.0030)
Proliferation-associated gene
H4F2 0.9 (P 5 0.5517) 0.6 (P 5 0.0502) 0.8 (P 5 0.3823)
PCNA 0.8 (P 5 0.1920) 1.0 (P 5 0.9392) 0.8 (P 5 0.7210)
2305
DNA METHYLTRANSFERASE EXPRESSION AND HYPERMETHYLATION
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
either H4F2 or PCNA for normalization (Table 1; Fig. 3, C, D, G, and
H). The proliferation dependence of DNMT3A and DNMT3B gene
expression is not known. These results could indicate true up-regula-
tion of DNMT3A or DNMT3B in tumors, or the situation could be
analogous to that for DNMT1. Regardless of which of these two
scenarios is correct, if the apparent up-regulation of DNMT3A or
DNMT3B is responsible for the methylator phenotype in some human
colorectal tumors, then the extent of up-regulation should be greater in
tumors with frequent CpG island hypermethylation. We have inves-
tigated whether there is a direct link between expression of any of the
three DNA methyltransferases and the frequency of CpG island hy-
permethylation.
CpG Island Hypermethylation and DNA Methyltransferase Ex-
pression. We analyzed the methylation status of four CpG islands
known to undergo de novo methylation in human colorectal tumors in
all normal and tumor samples. These CpG islands are associated with
the genes APC (26), ESR1 (estrogen receptor; Ref. 27), CDKN2A
(p16) (2), and MLH1 (28, 29). We found that DNMT3A and DNMT3B
do not appear to be up-regulated in the two tumors (samples 10 and
17) with the most frequent (three of four) CpG island hypermethyla-
tion (Fig. 3). Tumors with relatively high DNMT3A or DNMT3B
expression levels tend to have at most one hypermethylated CpG
island. There is just one sample (26T) with high DNMT3A and/or
DNMT3B expression and two hypermethylated CpG islands. A similar
lack of concordance is apparent when CpG island hypermethylation
frequencies are compared to DNMT1 expression levels (Fig. 2).
ANOVA was used to compare the mean DNMT gene expression
between categories of CpG island hypermethylation frequency (Table
2). We performed 12 separate ANOVA calculations to investigate all
combinations of the three DNMT genes and the four normalization
genes. None of these combinations yielded a statistically significant P
value (Table 2), which would have indicated a correlation between the
frequency of CpG island hypermethylation and the level of DNMT
gene expression. Therefore, regardless of whether or not DNMT3A or
DNMT3B gene expression is proliferation dependent, as is DNMT1,
the RNA levels of neither of these two genes in individual tumors
correlate with CpG island hypermethylation frequency.
We conclude that most cases of frequent hypermethylation of CpG
islands in human colorectal tumors do not result from a simple
transcriptional up-regulation of any of the three known DNA meth-
yltransferase genes. This leaves open the possibility that one or more
of these genes are up-regulated posttranscriptionally. It is also con-
ceivable that other factors regulate the activity of the DNA methyl-
transferases, either by interacting with the enzymes themselves or by
regulating access to the DNA substrate. The molecular basis for the
methylator phenotype in human colorectal tumors could be found in
the disruption of the control mechanisms preventing access of the
DNA methyltransferases to CpG islands rather than in the mere
up-regulation of DNA methyltransferase levels in the cell.
Acknowledgments
We thank Dennis Salonga and Ji Min Park for help in generating cDNAs
and in designing TaqMan oligonucleotides.
References
1. Jones, P. A., and Laird, P. W. Cancer epigenetics comes of age. Nat. Genet., 21:
163–167, 1999.
2. Ahuja, N., Mohan, A. L., Li, Q., Stolker, J. M., Herman, J. G., Hamilton, S. R.,
Baylin, S. B., and Issa, J. P. Association between CpG island methylation and
microsatellite instability in colorectal cancer. Cancer Res., 57: 3370–3374, 1997.
3. Liang, G., Salem, C. E., Yu, M. C., Nguyen, H. D., Gonzales, F. A., Nguyen, T. T.,
Nichols, P. W., and Jones, P. A. DNA methylation differences associated with tumor
tissues identified by genome scanning analysis. Genomics, 53: 260–268, 1998.
4. Bakin, A. V., and Curran, T. Role of DNA 5-methylcytosine transferase in cell
transformation by fos. Science (Washington DC), 283: 387–390, 1999.
5. Bestor, T. H. Activation of mammalian DNA methyltransferase by cleavage of a Zn
binding regulatory domain. EMBO J., 11: 2611–2617, 1992.
6. Smith, S. S. Biological implications of the mechanism of action of human DNA
(cytosine-5)methyltransferase. Prog. Nucleic Acid Res. Mol. Biol., 49: 65–111, 1994.
7. Okano, M., Xie, S., and Li, E. Cloning and characterization of a family of novel
mammalian DNA (cytosine-5)methyltransferases. Nat. Genet., 19: 219–220, 1998.
8. Okano, M., Xie, S., and Li, E. Dnmt2 is not required for de novo and maintenance
methylation of viral DNA in embryonic stem cells. Nucleic Acids Res., 26: 2536
2540, 1998.
9. Van den Wyngaert, I., Sprengel, J., Kass, S. U., and Luyten, W. H. Cloning and
analysis of a novel human putative DNA methyltransferase. FEBS Lett., 426: 283–
289, 1998.
10. Yoder, J. A., and Bestor, T. H. A candidate mammalian DNA methyltransferase
related to pmt1p of fission yeast. Hum. Mol. Genet., 7: 279–284, 1998.
11. El-Deiry, W. S., Nelkin, B. D., Celano, P., Yen, R. W., Falco, J. P., Hamilton, S. R.,
and Baylin, S. B. High expression of the DNA methyltransferase gene characterizes
human neoplastic cells and progression stages of colon cancer. Proc. Natl. Acad. Sci.
USA, 88: 3470–3474, 1991.
12. Issa, J. P., Vertino, P. M., Wu, J., Sazawal, S., Celano, P., Nelkin, B. D., Hamilton,
S. R., and Baylin, S. B. Increased cytosine DNA-methyltransferase activity during
colon cancer progression. J. Natl. Cancer Inst., 85: 1235–1240, 1993.
13. Schmutte, C., Yang, A. S., Nguyen, T. T., Beart, R. W., and Jones, P. A. Mechanisms
for the involvement of DNA methylation in colon carcinogenesis. Cancer Res., 56:
2375–2381, 1996.
14. Lee, P. J., Washer, L. L., Law, D. J., Boland, C. R., Horon, I. L., and Feinberg, A. P.
Limited up-regulation of DNA methyltransferase in human colon cancer reflecting
increased cell proliferation. Proc. Natl. Acad. Sci. USA, 93: 10366–10370, 1996.
15. Kautiainen, T. L., and Jones, P. A. DNA methyltransferase levels in tumorigenic and
nontumorigenic cells in culture. J. Biol. Chem., 261: 1594–1598, 1986.
16. Wolff, R. K., Frazer, K. A., Jackler, R. K., Lanser, M. J., Pitts, L. H., and Cox, D. R.
Analysis of chromosome 22 deletions in neurofibromatosis type 2-related tumors.
Am. J. Hum. Genet., 51: 478485, 1992.
17. Chomczynski, P., and Sacchi, N. Single-step method of RNA isolation by acid
guanidinium thiocyanate-phenol-chloroform extraction. Anal. Biochem., 162: 156
159, 1987.
18. Gibson, U. E., Heid, C. A., and Williams, P. M. A novel method for real time
quantitative RT-PCR. Genome Res., 6: 995–1001, 1996.
19. Heid, C. A., Stevens, J., Livak, K. J., and Williams, P. M. Real time quantitative PCR.
Genome Res., 6: 986–994, 1996.
20. Bender, C. M., Pao, M. M., and Jones, P. A. Inhibition of DNA methylation by
5-aza-29-deoxycytidine suppresses the growth of human tumor cell lines. Cancer Res.,
58: 95–101, 1998.
21. Livak, K. J., Flood, S. J., Marmaro, J., Giusti, W., and Deetz, K. Oligonucleotides
with fluorescent dyes at opposite ends provide a quenched probe system useful for
detecting PCR product and nucleic acid hybridization. PCR Methods Applications, 4:
357–362, 1995.
22. Xiong, Z., and Laird, P. W. COBRA: a sensitive and quantitative DNA methylation
assay. Nucleic Acids Res., 25: 2532–2534, 1997.
23. Teubner, B., and Schulz, W. A. Regulation of DNA methyltransferase during differ-
entiation of F9 mouse embryonal carcinoma cells. J. Cell. Physiol., 165: 284–290,
1995.
24. Papadopoulos, T., Pfeifer, G. P., Hoppe, F., Drahovsky, D., and Muller-Hermelink,
H. K. Proliferation-associated expression of DNA methyltransferase in human em-
bryonic lung cells. Virchows Archiv. B Cell. Pathol. Incl. Mol. Pathol., 56: 371–375,
1989.
25. Szyf, M., Bozovic, V., and Tanigawa, G. Growth regulation of mouse DNA meth-
yltransferase gene expression. J. Biol. Chem., 266: 10027–10030, 1991.
26. Hiltunen, M. O., Alhonen, L., Koistinaho, J., Myohanen, S., Paakkonen, M., Marin,
S., Kosma, V. M., and Janne, J. Hypermethylation of the APC (adenomatous poly-
posis coli) gene promoter region in human colorectal carcinoma. Int. J. Cancer, 70:
644648, 1997.
27. Issa, J. P., Ottaviano, Y. L., Celano, P., Hamilton, S. R., Davidson, N. E., and Baylin,
S. B. Methylation of the oestrogen receptor CpG island links ageing and neoplasia in
human colon. Nat Genet., 7: 536–540, 1994.
28. Herman, J. G., Umar, A., Polyak, K., Graff, J. R., Ahuja, N., Issa, J. P., Markowitz,
S., Willson, J. K., Hamilton, S. R., Kinzler, K. W., Kane, M. F., Kolodner, R. D.,
Vogelstein, B., Kunkel, T. A., and Baylin, S. B. Incidence and functional conse-
quences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc. Natl.
Acad. Sci. USA, 95: 68706875, 1998.
29. Veigl, M. L., Kasturi, L., Olechnowicz, J., Ma, A. H., Lutterbaugh, J. D., Periyasamy,
S., Li, G. M., Drummond, J., Modrich, P. L., Sedwick, W. D., and Markowitz, S. D.
Biallelic inactivation of hMLH1 by epigenetic gene silencing, a novel mechanism
causing human MSI cancers. Proc. Natl. Acad. Sci. USA, 95: 86988702, 1998.
2306
DNA METHYLTRANSFERASE EXPRESSION AND HYPERMETHYLATION
on May 30, 2013. © 1999 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
... For these reasons, epigenetic therapies employing enzyme inhibitors always affect healthy, replicating cells, like conventional chemotherapy. This explains also why many enzymes involved in DNA and histone modifications are upregulated in tumor cells, and why many reports about specific activation of the encoding genes in tumor cells are false positive findings which disappear if proper controls for proliferation-associated effects are employed (already shown more than 20 years ago, e.g., [43]). ...
Article
Full-text available
Simple Summary Triple-negative breast cancer is an aggressive subtype of this frequent malignancy in women, for which new therapeutic options are urgently needed. Changes in the packaging of the chromosomes within the cell nucleus, so called “epigenetic modifications”, cooperate with direct changes of the DNA sequence, called “genetic alterations”, in the development and progression of human tumors. The definition of epigenetic phenomena and the identification of the underlying molecular mechanisms is an active field of research with many open questions. So far, the best-defined players in the field of epigenetics are the methylation of DNA and the covalent modification of histone proteins around which the DNA is wrapped in the nucleus of human cells. Both modifications affect the expression of genes. The principal reversibility of these epigenetic modifications of the DNA strands and the histone proteins make them an attractive target for therapeutic intervention by so-called “epidrugs”, and several promising clinical trials are under way. However, many questions regarding specificity, safety, and efficiency of these drugs are still unresolved. Abstract Breast cancer, the most frequent malignancy in women worldwide, is a molecularly and clinically very heterogeneous disease. Triple-negative breast cancer is defined by the absence of hormone receptor and growth factor receptor ERBB2/HER2 expression. It is characterized by a more aggressive course of disease and a shortage of effective therapeutic approaches. Hallmarks of cancer cells are not only genetic alterations, but also epigenetic aberrations. The most studied and best understood alterations are methylation of the DNA base cytosine and the covalent modification of histone proteins. The reversibility of these covalent modifications make them attractive targets for therapeutic intervention, as documented in numerous ongoing clinical trials. Epidrugs, targeting DNA methylation and histone modifications, might offer attractive new options in treating triple-negative breast cancer. Currently, the most promising options are combination therapies in which the epidrug increases the efficiency of immuncheckpoint inhibitors. This review focusses exclusively on DNA methylation and histone modifications. In reviewing the knowledge about epigenetic therapies in breast cancer, and especially triple-negative breast cancer, the focus is on explaining concepts and raising awareness of what is not yet known and what has to be clarified in the future.
... Our results demonstrated that the levels of DNMTs mRNA were different in the ten colorectal tumors. In agreement with some previous reports [23,51], our findings also indicated no correlation between DNMTs overexpression and CpG islands hypermethylation in CRC patients. It could be concluded that in CRC patients, the gene-specific promoter hypermethylation is not dependent on the DNMTs transcript levels and is regulated through other processes. ...
Article
Full-text available
To investigate the effects of thymidylate synthase (TS) 3'UTR genotype on promotor methylation of tumor-related genes in 22 patients with sporadic colorectal cancer (CRC) from southern Iran. We evaluated the correlations of TS 3'UTR genotype with promoter methylation of hTERT, hMLH1, MSH2, MMP2, CDH1, p14, p16, and p21 genes in CRC patients. The polymorphism of TS 3′UTR was evaluated through mutagenically specific PCR. The genes promoter methylation was determined using methylation-specific PCR. For 10 patients, the gene expression profile of epigenetic regulating enzymes, histone deacetylases (HDACs) and DNA methyltransferases (DNMTs), was also examined in both tumor and normal adjacent tissues by quantitative real time PCR. There was a significant association between the hMLH1 methylation and age of patients (P= 0.039) and also between MSH2 methylation and tumor site (P= 0.036). There was insignificant association between gene-specific methylation and TS 3′UTR genotype. However, all polymorphic genotypes of TS were associated with higher methylation of hMLH1 and CDH1 and lower methylation of MSH2. The -6bp/+6bp (heterozygous mutant) and [-6bp/+6bp, +6bp/+6bp] (homozygous mutant) genotypes resulted in higher methylation of p16, and -6bp/+6bp and [-6bp/+6bp, +6bp/+6bp] genotypes were correlated with lower methylation of MMP2. The overexpression of epigenetic enzymes, HDACs and DNMTs, was also demonstrated. There was no association between DNMTs transcript levels and gene-specific hypermethylation. The polymorphic TS genotypes, especially -6bp/+6bp, could affect methylation frequencies of studied genes. Moreover, promoter methylation status was not dependent on DNMTs gene expression. Large sample size studies may contribute to validate these findings.
... Since DNA methylation is catalyzed by the family of enzymes known as DNA methyltransferases (DNMT1, DNMT3A, DNMT3B, and DNMT3L), various studies have been conducted to understand aberrant DNA methylation patterns in association with the expression of DNA methyltransferases in different types of cancer [64][65][66][67]. In general, the impact of DNMT aberrations in the promotion of tumorigenesis is still controversial, and relevant targeted therapies for DNMTs are still under exploration [68,69]. ...
Article
Full-text available
Epigenetic modifications are known to regulate cell phenotype during cancer progression, including breast cancer. Unlike genetic alterations, changes in the epigenome are reversible, thus potentially reversed by epi-drugs. Breast cancer, the most common cause of cancer death worldwide in women, encompasses multiple histopathological and molecular subtypes. Several lines of evidence demonstrated distortion of the epigenetic landscape in breast cancer. Interestingly, mammary cells isolated from breast cancer patients and cultured ex vivo maintained the tumorigenic phenotype and exhibited aberrant epigenetic modifications. Recent studies indicated that the therapeutic efficiency for breast cancer regimens has increased over time, resulting in reduced mortality. Future medical treatment for breast cancer patients, however, will likely depend upon a better understanding of epigenetic modifications. The present review aims to outline different epigenetic mechanisms including DNA methylation, histone modifications, and ncRNAs with their impact on breast cancer, as well as to discuss studies highlighting the central role of epigenetic mechanisms in breast cancer pathogenesis. We propose new research areas that may facilitate locus-specific epigenome editing as breast cancer therapeutics.
... First, deregulation of relevant methylation enzymes like DNA methyltransferases (DMNT) can kickstart aberrant methylation. While DMNT is rarely mutated in CRC (unlike other cancers), the protein is overexpressed but not related to a specific aberrant methylation phenotype [119,120]. TET1 methylation has been suggested in the progression of CIMP CRC [121]. Mechanisms that normally protect the genome from aberrant methylation (e.g., DNA-binding proteins, RNA polymerase, or histone binding) may be modified which allows nearby hypermethylated regions to affect previously nonmethylated areas [122]. ...
Chapter
Full-text available
This chapter describes genetics and epigenetics discoveries that have allowed investigators to better define cancer at the molecular level. Taking into consideration the expanse of the field of cancer, the focus will be on colon cancer as a platform to provide examples of techniques, recent discoveries, and translation of genetic studies to cancer care. In addition, this segment contributes to our understanding of racial and ethnic disparities in colon cancer and the use of -omic assessments as an application in cancer research. Thus, this section will provide an overarching view of cancer by defining the molecular characteristics of colon cancer; parameters of cancer disparities; and genetic factors that contribute to colon-tumor biology, specifically recent findings at the DNA, RNA, and protein levels. Importantly, the correlation of these factors with the immune system will be defined. This section ends with future directions for studying colon cancer in patients from medically underserved communities. In summary, this unit provides an introduction to how genetic and genomic investigations are helping to elucidate biological questions in an inclusive manner that will benefit patients on a global scale.
... Publicly available data from The Cancer Genome Atlas (TCGA) illustrates that significant alterations in DNMT mRNA expression are observed in a wide variety of cancers ( Figure 2). Although the gene expression data in Figure 2 are not normalized to proliferation-specific control (143), the overexpression of DNMTs in most of these cancers has been verified in several studies (reviewed in (144,145)). High DNMT3B expression levels correlate with a particular colorectal cancer phenotype characterized by high global DNA methylation levels, particularly at CpG islands (146). ...
Article
Full-text available
In mammals, DNA methyltransferases DNMT1 and DNMT3’s (A, B and L) deposit and maintain DNA methylation in dividing and nondividing cells. Although these enzymes have an unremarkable DNA sequence specificity (CpG), their regional specificity is regulated by interactions with various protein factors, chromatin modifiers, and post-translational modifications of histones. Changes in the DNMT expression or interacting partners affect DNA methylation patterns. Consequently, the acquired gene expression may increase the proliferative potential of cells, often concomitant with loss of cell identity as found in cancer. Aberrant DNA methylation, including hypermethylation and hypomethylation at various genomic regions, therefore, is a hallmark of most cancers. Additionally, somatic mutations in DNMTs that affect catalytic activity were mapped in Acute Myeloid Leukemia cancer cells. Despite being very effective in some cancers, the clinically approved DNMT inhibitors lack specificity, which could result in a wide range of deleterious effects. Elucidating distinct molecular mechanisms of DNMTs will facilitate the discovery of alternative cancer therapeutic targets. This review is focused on: (i) the structure and characteristics of DNMTs, (ii) the prevalence of mutations and abnormal expression of DNMTs in cancer, (iii) factors that mediate their abnormal expression and (iv) the effect of anomalous DNMT-complexes in cancer.
... 3 Sin embargo, se ha visto que se sobreexpresan en diferentes tipos de cáncer como: colorrectal, próstata, ovario, endometrio y mama. [4][5][6][7][8][9] Con respecto al cáncer de seno, en un trabajo en el cual se evaluaron 130 carcinomas de mama no metastásicos unilaterales primarios, la cantidad de mRNA de las enzimas DNMT1, DNMT3a y DNMT3b se correlacionaron positivamente entre sí, esto sugiere una regulación coordinada. En 30% de estas pacientes se detectó sobreexpresión de DNMT3b y sólo se identificó sobreexpresión de 5.4% para DNMT1 y de 3.1% para DNMT3a. ...
... However, the gains observed were not specific to the orthologues of those genes methylated in human tumours and the relative degree of Dnmt3b targeting of H3K36me3 marked loci was not assessed 50 . The relevance of DNMT3B overexpression in human cancer has also been questioned and apparent upregulation is suggested to reflect the greater proportion of cycling cells in tumour tissues 51,52 . The mechanism by which polycomb marked CGIs aberrantly gain methylation in cancer remains unclear 2 . ...
Article
Full-text available
The aberrant gain of DNA methylation at CpG islands is frequently observed in colorectal tumours and may silence the expression of tumour suppressors such as MLH1 . Current models propose that these CpG islands are targeted by de novo DNA methyltransferases in a sequence-specific manner, but this has not been tested. Using ectopically integrated CpG islands, here we find that aberrantly methylated CpG islands are subject to low levels of de novo DNA methylation activity in colorectal cancer cells. By delineating DNA methyltransferase targets, we find that instead de novo DNA methylation activity is targeted primarily to CpG islands marked by the histone modification H3K36me3, a mark associated with transcriptional elongation. These H3K36me3 marked CpG islands are heavily methylated in colorectal tumours and the normal colon suggesting that de novo DNA methyltransferase activity at CpG islands in colorectal cancer is focused on similar targets to normal tissues and not greatly remodelled by tumourigenesis.
Article
Full-text available
The measurement of DNA methyltransferase (MTase) activity and screening of DNA MTase inhibitors holds significant importance for the diagnosis and therapy of methylation-related illness. Herein, we developed a colorimetric biosensor (PER-FHGD nanodevice) to detect DNA MTase activity by integrating the primer exchange reaction (PER) amplification and functionalized hemin/G-quadruplex DNAzyme (FHGD). By replacing the native hemin cofactor into the functionalized cofactor mimics, FHGD has exhibited significantly improved catalytic efficiency, thereby enhancing the detection performance of the FHGD-based system. The proposed PER-FHGD system is capable of detecting Dam MTase with excellent sensitivity, exhibiting a limit of detection (LOD) as low as 0.3 U/mL. Additionally, this assay demonstrates remarkable selectivity and ability for Dam MTase inhibitors screening. Furthermore, using this assay, we successfully detect the Dam MTase activity both in serum and in E. coli cell extracts. Importantly, this system has the potential to serve as a universal strategy for FHGD-based diagnosis in point-of-care (POC) tests, by simply altering the recognition sequence of the substrate for other analytes.
Article
Subclonal loss of mismatch repair (MMR) proteins has been described in a small subset of endometrial carcinomas (ECs), but the genomic basis for this phenomenon has received limited attention. Herein, we retrospectively evaluated all ECs with MMR immunohistochemistry (n=285) for subclonal loss, and in those (n=6), performed a detailed clinicopathologic and genomic comparison of the MMR-deficient and MMR-proficient components. Three tumors were FIGO stage IA, and one each stage IB, II, and IIIC2. Patterns of subclonal loss were as follows: (1) 3 FIGO grade 1 endometrioid carcinomas with subclonal MLH1/PMS2, MLH1 promoter hypermethylation, and no MMR gene mutations; (2) POLE-mutated FIGO grade 3 endometrioid carcinoma with subclonal PMS2, and PMS2 and MSH6 mutations limited to the MMR-deficient component; (3) dedifferentiated carcinoma with subclonal MSH2/MSH6, as well as complete loss of MLH1/PMS2, MLH1 promoter hypermethylation, and PMS2 and MSH6 mutations in both components; (4) dedifferentiated carcinoma with subclonal MSH6, and somatic and germline MSH6 mutations in both components, but with a higher allele frequency in MMR-deficient foci. Recurrences occurred in 2 patients, one consisted of the MMR-proficient component from a FIGO 1 endometrioid carcinoma, while the other was from the MSH6-mutated dedifferentiated endometrioid carcinoma. At the last follow-up (median: 44 mo), 4 patients were alive and disease-free and 2 were alive with disease. In summary, subclonal MMR loss reflects subclonal and often complex genomic and epigenetic alterations, which may have therapeutic implications and therefore must be reported when present. In addition, subclonal loss can occur in both POLE-mutated and Lynch syndrome-associated ECs.
Chapter
Full-text available
Epigenetics “above or over genetics” is the term used for processes that result in modifications which are stably inherited through cell generations, without changing the underlying DNA sequence of the cell. These include DNA methylation, Post-translational histone modification and non-coding RNAs. Over the last two decades, interest in the field of epigenetics has grown manifold because of the realization of its involvement in key cellular and pathological processes beyond what was initially anticipated. Epigenetics and chromatin biology have been underscored to play key roles in diseases like cancer. The landscape of different epigenetic signatures can vary considerably from one cancer type to another, and even from one ethnic group to another in the case of same cancer. This chapter discusses the emerging role of epigenetics and chromatin biology in the field of cancer research. It discusses about the different forms of epigenetic mechanisms and their respective role in carcinogenesis in the light of emerging research.
Article
Full-text available
A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described. The method provides a pure preparation of undegraded RNA in high yield and can be completed within 4 h. It is particularly useful for processing large numbers of samples and for isolation of RNA from minute quantities of cells or tissue samples.
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
Full-text available
The steady state level of DNA methyltransferase mRNA is markedly increased as growth-arrested Balb/c 3T3 cells progress into the S phase of the cell cycle. mRNA abundance is reduced to the basal level before termination of DNA synthesis activity. Maintenance DNA methylation activity in nuclear extracts follows a similar pattern with two exceptions. (a) A small peak of DNA methylation activity is detected in early G1 phase. (b) The extinction of DNA methylation activity lags behind the termination of DNA synthesis. Nuclear runon experiments demonstrate that the gene is transcribed in growth-arrested cells, and expression of the gene is post-transcriptionally regulated. We suggest that this mode of regulation of the DNA methyltransferase gene might play an important role in determining and maintaining DNA methylation patterns.
Article
Full-text available
The levels of DNA methyltransferase in nuclei from 9 tumorigenic and 9 nontumorigenic cell lines were examined. In all but 2 cases, the extractable methyltransferase activity was 4-3000-fold higher in tumorigenic than in nontumorigenic cells. Tumorigenic and nontumorigenic cells from four species were grown in the presence of various concentrations (10(-8)-10(-6) M) of an inhibitor of the methylase enzyme, 5-aza-2'-deoxycytidine (5-aza-dCyd). The reduction of 5-methylcytosine content in newly replicated DNA in the presence of 5-aza-dCyd was used to determine the relative methylase activity in each cell line. In all 4 cases, tumorigenic cells required larger doses of drug to inhibit DNA methylation to the same extent as their nontumorigenic counterparts. The relative rates of incorporation of [3H]5-aza-dCyd were determined for each cell line, and tumorigenic cells were shown to incorporate equal or greater amounts of 5-aza-dCyd into DNA compared to nontumorigenic cells. These results showed that the differences in the inhibition of DNA methylation in response to 5-aza-dCyd were not due to differences in the ability of these cells to incorporate the drug. Thus, it was demonstrated by two independent methods that tumorigenic cells contained higher levels of methylating capacity than nontumorigenic cells. This overabundance of methyltransferase may alter DNA methylation patterns and affect phenotypic stability.
Article
Full-text available
A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described. The method provides a pure preparation of undegraded RNA in high yield and can be completed within 4 h. It is particularly useful for processing large numbers of samples and for isolation of RNA from minute quantities of cells or tissue samples.
Article
Full-text available
The 5' nuclease PCR assay detects the accumulation of specific PCR product by hybridization and cleavage of a double-labeled fluorogenic probe during the amplification reaction. The probe is an oligonucleotide with both a reporter fluorescent dye and a quencher dye attached. An increase in reporter fluorescence intensity indicates that the probe has hybridized to the target PCR product and has been cleaved by the 5'-->3' nucleolytic activity of Taq DNA polymerase. In this study, probes with the quencher dye attached to an internal nucleotide were compared with probes with the quencher dye attached to the 3'-end nucleotide. In all cases, the reporter dye was attached to the 5' end. All intact probes showed quenching of the reporter fluorescence. In general, probes with the quencher dye attached to the 3'-end nucleotide exhibited a larger signal in the 5' nuclease PCR assay than the internally labeled probes. It is proposed that the larger signal is caused by increased likelihood of cleavage by Taq DNA polymerase when the probe is hybridized to a template strand during PCR. Probes with the quencher dye attached to the 3'-end nucleotide also exhibited an increase in reporter fluorescence intensity when hybridized to a complementary strand. Thus, oligonucleotides with reporter and quencher dyes attached at opposite ends can be used as homogeneous hybridization probes.
Article
The cell cycle-dependent and proliferation-associated expression of the enzyme DNA methyltransferase has been evaluated immunocytochemically in synchronized L-132 human embryonic lung cells, using the anti-DNA methyltransferase monoclonal antibody M1F6D7/5C10. DNA methyltransferase-reactivity was firstly seen in mid-G1 cells. An intense and granular reaction in the cell nuclei with a sparing of the nucleoli was observed in addition to a homogenous and faint cytoplasmic staining. The staining intensity in the cell nuclei increased progressively up to mitosis. In early mitotic cells an intense perichromosomal staining was observed in addition to a homogenous staining of cyto- and karyoplasm after the resolving of the core membrane. In late mitosis the staining intensity decreased rapidly. Early G1 cells and density inhibited, resting G0 cells showed no DNA methyltransferase reactivity at all. Our results indicate that anti-DNA methyltransferase monoclonal antibodies could become valuable tools to detect proliferating cells in cell cultures and tissues.
Article
The neurofibromatosis type 2 (NF2) gene has been hypothesized to be a recessive tumor suppressor, with mutations at the same locus on chromosome 22 that lead to NF2 also leading to sporadic tumors of the types seen in NF2. Flanking markers for this gene have previously been defined as D22S1 centromeric and D22S28 telomeric. Identification of subregions of this interval that are consistently rearranged in the NF2-related tumors would aid in better defining the disease locus. To this end, we have compared tumor and constitutional DNAs, isolated from 39 unrelated patients with sporadic and NF2-associated acoustic neuromas, meningiomas, schwannomas, and ependymomas, at eight polymorphic loci on chromosome 22. Two of the tumors studied revealed loss-of-heterozygosity patterns, which is consistent with the presence of chromosome 22 terminal deletions. By using additional polymorphic markers, the terminal deletion breakpoint found in one of the tumors, an acoustic neuroma from an NF2 patient, was mapped within the previously defined NF2 region. The breakpoint occurred between the haplotyped markers D22S41/D22S46 and D22S56. This finding redefines the proximal flanking marker and localizes the NF2 gene between markers D22S41/D22S46 and D22S28. In addition, we identified a sporadic acoustic neuroma that reveals a loss-of-heterozygosity pattern consistent with mitotic recombination or deletion and reduplication, which are mechanisms not previously seen in studies of these tumors. This finding, while inconsistent with models of tumorigenesis that invoke single deletions and their gene-dosage effects, lends further support to the recessive tumor-suppressor model.
Article
Mammalian DNA (cytosine-5) methyltransferase contains a C-terminal domain that is closely related to bacterial cytosine-5 restriction methyltransferase. This methyltransferase domain is linked to a large N-terminal domain. It is shown here that the N-terminal domain contains a Zn binding site and that the N- and C-terminal domains can be separated by cleavage with trypsin or Staphylococcus aureus protease V8; the protease V8 cleavage site was determined by Edman degradation to lie 10 residues C-terminal of the run of alternating lysyl and glycyl residues which joins the two domains and six residues N-terminal of the first sequence motif conserved between the mammalian and bacterial cytosine methyltransferases. While the intact enzyme had little activity on unmethylated DNA substrates, cleavage between the domains caused a large stimulation of the initial velocity of methylation of unmethylated DNA without substantial change in the rate of methylation of hemimethylated DNA. These findings indicate that the N-terminal domain of DNA methyltransferase ensures the clonal propagation of methylation patterns through inhibition of the de novo activity of the C-terminal domain. Mammalian DNA methyltransferase is likely to have arisen via fusion of a prokaryotic-like restriction methyltransferase and an unrelated DNA binding protein. Stimulation of the de novo activity of DNA methyltransferase by proteolytic cleavage in vivo may contribute to the process of ectopic methylation observed in the DNA of aging animals, tumors and in lines of cultured cells.
Article
DNA methylation abnormalities occur consistently in human neoplasia including widespread hypomethylation and more recently recognized local increases in DNA methylation that hold potential for gene inactivation events. To study this imbalance further, we have cloned and localized to chromosome 19 a portion of the human DNA methyltransferase gene that codes for the enzyme catalyzing DNA methylation. Expression of this gene is low in normal human cells, significantly increased (30- to 50-fold by PCR analysis) in virally transformed cells, and strikingly elevated in human cancer cells (several hundredfold). In comparison to colon mucosa from patients without neoplasia, median levels of DNA methyltransferase transcripts are 15-fold increased in histologically normal mucosa from patients with cancers or the benign polyps that can precede cancers, 60-fold increased in the premalignant polyps, and greater than 200-fold increased in the cancers. Thus, increases in DNA methyltransferase gene expression precede development of colonic neoplasia and continue during progression of colonic neoplasms. These increases may play a role in the genetic instability of cancer and mark early events in cell transformation.