Sei sulla pagina 1di 12

Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.

CAPR-13-0264

Cancer
Prevention
Research Article Research

Folate Deficiency Induces Dysfunctional Long and


Short Telomeres; Both States Are Associated with
Hypomethylation and DNA Damage in Human
WIL2-NS Cells
Caroline F. Bull1,2, Graham Mayrhofer2, Nathan J. O'Callaghan1, Amy Y. Au3, Hilda A. Pickett3,4,
Grace Kah Mun Low5, Dimphy Zeegers5, M. Prakash Hande5,6, and Michael F. Fenech1

Abstract
The essential role of dietary micronutrients for genome stability is well documented, yet the effect of folate
deficiency or excess on telomeres is not known. Accordingly, human WIL2-NS cells were maintained in
medium containing 30, 300, or 3,000 nmol/L folic acid (FA) for 42 days to test the hypothesis that chronic
folate deficiency would cause telomere shortening and dysfunction. After 14 days, telomere length (TL) in
FA-deficient (30 nmol/L) cultures was 26% longer than that of 3,000 nmol/L FA cultures; however, this was
followed by rapid telomere attrition over the subsequent 28 days (P trend, P < 0.0001); both long and short
telomere status was positively correlated with biomarkers of chromosome instability (P  0.003) and
mitotic dysfunction (P ¼ 0.01), measured by the cytokinesis-block micronucleus cytome (CBMN-cyt) assay.
The early increase in TL was associated with FA-deficiency–induced global DNA hypomethylation (P ¼
0.05), with an effect size similar to that induced by the DNA methyltransferase inhibitor, 5-aza-20 -
deoxycytidine. Quantitative PCR analysis indicated a negative association between FA concentration and
uracil incorporation into telomeric DNA (r ¼ 0.47, P ¼ 0.1), suggesting a possible plausible mechanism for
uracil as a cause of folate deficiency–induced telomere dysfunction or deletion. Peptide nucleic acid-FISH
(PNA-FISH) analysis showed that FA deficiency resulted in 60% of micronuclei containing acentric terminal
fragments, an observation consistent with the 3-fold increase in terminal deletions (P ¼ 0.0001). Together,
these results demonstrate the impact of folate deficiency on biomarkers of telomere maintenance and
integrity, and provide evidence that dysfunctional long telomeres may be as important as critically short
telomeres as a cause of chromosomal instability. Cancer Prev Res; 7(1); 128–38. 2013 AACR.

Introduction ciated with unhealthy diet and lifestyle (2, 3), exposure to
Functionally intact telomeres are critical for protecting chronic stress (4), childhood violence (5), and negative
against degradation by nucleases, end-to-end fusions, and psychological states (6). Conversely, meditation, exercise,
structural rearrangement of chromosomes (1), each a and a healthy diet offer a protective effect against telomere
potential initiating factor for cancer and degenerative dis- erosion (7, 8). The essential role of dietary factors in
ease (1). Accelerated telomere shortening has been asso- maintenance of chromosome stability is well doc-
umented (9, 10); however, knowledge of the requirements
for individual micronutrients for maintenance of opti-
mal telomere length (TL) and functionality is extremely
Authors' Affiliations: 1Nutritional Genomics Laboratory, CSIRO Animal,
Food and Health Sciences, Adelaide, South Australia; 2Discipline of Micro- limited.
biology and Immunology, School of Molecular and Biomedical Sciences, Folate (vitamin B9) is increasingly consumed in the
University of Adelaide, South Australia; 3Children's Medical Research supplemental (monoglutamated) form of folic acid (FA).
Institute, Westmead, New South Wales, Australia; 4Sydney Medical
School, University of Sydney, New South Wales, Australia; 5Department Moderate folate deficiency can cause chromosomal insta-
of Physiology, Yong Loo Lin School of Medicine, National University of bility (CIN) comparable with that induced by carcino-
Singapore; and 6Tembusu College, National University of Singapore,
Singapore
genic doses of ionizing radiotherapy in vitro (10). In its
role as a one carbon (methyl)-group donor, folate is
Note: Supplementary data for this article are available at Cancer Prevention
Research Online (http://cancerprevres.aacrjournals.org/).
required for genomic stability via two distinct, yet inter-
dependent, biochemical pathways: (i) the synthesis of S-
Corresponding Authors: Caroline F. Bull, CSIRO Animal, Food and
Health Sciences, P.O. Box 10041, Adelaide BC, South Australia 5000, adenosyl methionine (SAM), essential for maintaining
Australia. Phone: 61-8-8303-8931; Fax: 61-8-8303-8899; E-mail: DNA cytosine methylation, epigenetic control of gene
caroline.bull@csiro.au; and Michael F. Fenech, michael.fenech@csiro.au expression, and structural integrity at regions of hyper-
doi: 10.1158/1940-6207.CAPR-13-0264 methylated tandem repeats (centromeres and the subte-
2013 American Association for Cancer Research. lomere; refs. 10–13); and (ii) methylation of dUMP to

128 Cancer Prev Res; 7(1) January 2014

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Telomeres

dTTP, forming thymidine for DNA synthesis and repair in situ location of centromeric and telomeric DNA within
(14, 15). When folate is limiting, thymidine residues in biomarkers of CIN.
DNA can be replaced by uracil, initiating a cycle of base
excision repair, increased frequency of abasic sites, and Cell culture
heightened risk of double-strand breaks (DSB; ref. 16). In WIL2-NS cells (American Type Culture Collection; CRL-
addition, compelling evidence has shown that repair 8155) were cultured for 7 days in complete RPMI 1640
responses can be restricted at telomeric DNA, suggesting medium (Sigma) before seeding in treatment medium at
that damage may be prolonged (17–19). The telomere, day 0 of the experimental period. Cells were authenticated
comprised of a repeating DNA hexamer (TTAGGGn), is at the time of delivery (2002) to confirm p53-negative
bound by proteins of the shelterin complex to form the status, and in 2011 were confirmed as being karyotypically
"telosome," key components of which are "telomere consistent with previous reports (25). Duplicate 90 mL
repeat binding factors" (TRF1 and TRF2), the binding cultures were maintained in 75 cm2 vented-cap culture
kinetics for which are highly sensitive to substrate flasks (Becton Dickinson) at 37 C in a humidified atmo-
changes (20, 21). It is plausible, therefore, that the thy- sphere with 5% CO2. Medium was replaced twice weekly.
midine-rich nature of telomeres makes them vulnerable Complete RPMI medium (control 3,000 nmol/L FA condi-
to uracil misincorporation, incomplete excision repair of tion) was supplemented to contain 5% FBS (Thermo) and
uracil, leading to abasic sites, unrepaired DNA strand 1% penicillin/streptomycin (Sigma). L-Glutamine (1%;
breaks, and compromised telosome formation when Sigma) was added immediately before use. Medium con-
folate is deficient. Accordingly, we hypothesized that taining 30 or 300 nmol/L FA was prepared by dilution of
telomeres would shorten at an accelerated rate under complete RPMI medium with an appropriate volume of
FA-deficient conditions, and tested this in an in vitro supplemented FA-free RPMI (Sigma). For 5-aza-20 -deoxy-
human cell line model. cytidine (5azadC) studies, filter sterilized 5azadC (Sigma)
stock solution (21.9 mmol/L in PBS) was added to RPMI
Materials and Methods medium (3,000 nmol/L FA) to final concentrations of either
Design 0.2 or 1.0 mmol/L 5azadC. Control (0 mmol/L 5azadC)
An in vitro model of chronic FA deficiency was established medium was prepared by adding equivalent volume of
using human WIL2-NS cells. FA concentrations were deter- vehicle (1 PBS).
mined based on pilot growth data, and on previous studies
indicating that DNA damage occurs in WIL2-NS cells at Determination of TL
20 and 200 nmol/L FA (22). Cells were cultured for 42 TL was measured in viable single cells at G0–G1 using the
days in medium either deficient (30 nmol/L), replete (300 flow cytometric method detailed previously (26). TL was
nmol/L), or supraphysiological (control, 3,000 nmol/L) for expressed relative to a (long telomere) control cell line,
FA. The "replete" concentration of 300 nmol/L is one which 1301 European Collection of Cell Cultures (ECACC). Telo-
would only be achieved in serum of individuals supple- mere-specific peptide nucleic acid-fluorescein isothiocyan-
menting with several milligrams of FA daily, whereas the ate (PNA-FITC) labeling was conducted using kit K5327
"control" concentration of 3,000 nmol/L is not achievable (Dako). Doublets, debris (dead and dying cells), and cells at
in vivo in humans. We do, however, live in an era in which G2 were excluded based on cell cycle staining with propi-
cells are increasingly taken out of our bodies, grown in dium iodide. The coefficient of variation of replicate mea-
culture, and returned to the body (e.g., stem cell, bone surements was 13.5%.
marrow, and skin graft transplantation). Such culture sys-
tems routinely use media with supraphysiological concen- CBMN-cyt assay
trations of FA and yet it is not known whether this is optimal Chromosomal damage and cytostasis measures were
for DNA damage prevention. The human WIL2-NS (B- determined using the CBMN-cyt assay, using the standard
lymphoblastoid) cell line was selected because folate defi- protocol detailed previously (27), with additional scoring
ciency has been identified as an important risk factor for B- criteria for fused nuclei (FUS) morphologies as described
cell lymphoma (23), and because its p53-deficient status (9). See details in Supplementary Materials and Methods.
allowed observation of substantial genome damage events
without excessive cell death through apoptosis (24). Deter- Measurement of uracil incorporation into telomeric
mination was made of TL by flow cytometry, biomarkers DNA by quantitative real-time PCR
of DNA damage and cytostasis by cytokinesis-block micro- The amount of uracil present in telomeric repeat
nucleus cytome (CBMN-cyt) assay, expression of human sequences was measured using a modification of the quan-
telomerase reverse transcriptase [(hTERT); quantitative PCR titative real-time PCR (qPCR) method used to measure TL,
(qPCR)], uracil incorporation into telomeric DNA (qPCR), incorporating additional digestion steps. For details see
and global methylation status (ELISA). In addition, both Supplementary Materials and Methods.
metaphase and (induced) binucleated interphase cell pre- Detailed methods are provided in Supplementary
parations were used for molecular cytogenetic analysis of Materials and Methods for cell growth, the CBMN-cyt
chromosome aberrations, loss of chromosome terminal assay, binucleated cell preparations for PNA-fluorescence
ends, the presence of interstitial telomeric DNA, and the in situ hybridisation (FISH), metaphase chromosome

www.aacrjournals.org Cancer Prev Res; 7(1) January 2014 129

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Bull et al.

preparations, FISH with PNA probes, immunoaffinity


purification-telomeric repeat amplification protocol (IP-
TRAP), C-circle assay, terminal restriction fragment (TRF)
analysis, DNA isolation and global DNA methylation,
RNA isolation and expression analysis of hTERT, mea-
surement of uracil incorporation into telomeric DNA by
qPCR, and statistical analyses.

Results and Discussion


Culture in folate-deficient medium is associated with
rapid fluctuations in TL
WIL2-NS cells were maintained in medium deficient (30
nmol/L), replete (300 nmol/L), or supraphysiological
(3,000 nmol/L; control) for FA. The doubling time of the
cells in culture varied considerably for each FA condition.
The number of viable cells at each time point, indicating rate
of cell division, was reduced in both the 30 and 300 nmol/L
FA cultures, compared with the 3,000 nmol/L condition
(Supplementary Fig. S1a). Cell viability and nuclear divi-
sion index (NDI) were significantly lower at every time
point in 30 nmol/L compared with that of 3,000 nmol/L
FA (P < 0.0001), whereas the frequency of necrotic cells
significantly increased at all time points in the 30 nmol/L
condition (P < 0.0001; Supplementary Fig. S1b–S1d).
Pilot TL data from short-term (21 days) cultures indicated
TL significantly increased in the short term (7–14 days)
under FA-deficient conditions, followed by a modest
decline between day 14 and day 21 (Supplementary Fig.
S2). To examine the longer term chronic effects of FA
deficiency, cells were then maintained in culture for 42
days. TL of samples at baseline, and weekly or fortnightly
thereafter, were compared by flow cytometry and results
expressed relative to the TL of reference cell line 1301. At day
14, TL in 30 nmol/L FA was 18.1  2.1 (mean  SD), 1.4-
fold that of baseline (12.7  3.4), followed by rapid
attrition (P trend, P < 0.0001). By day 42, TL had shortened
to 12.6  1.2, comparable with cells grown for the same
length of time in 3,000 nmol/L FA (13.5  2.7; Fig. 1A). TL
of control cultures remained stable throughout the 42-day
period (mean, 13.7  0.6). Mean area under the curves
(AUC TL) for the 30, 300, and 3,000 nmol/L FA conditions
was 638, 628, and 576, respectively. Despite the rapid TL
shortening beyond day 14 in the 30 nmol/L condition, a
negative correlation was recorded between [FA] and AUC
TL, over the 42 days (r ¼ 0.47; P ¼ 0.008). These are new
and intriguing observations, which do not support the
original hypothesis that FA deficiency is associated with
Figure 1. TL and biomarkers of CIN. A, TL in WIL2-NS cells cultured for
telomere shortening. 42 days in medium containing 30, 300, or 3,000 nmol/L FA. (Mean 
SD; N ¼ 10 at each time point for each FA concentration; n ¼ 20 for
TL is positively associated with biomarkers of CIN in day 0). B–F, frequency per 1,000 binucleated (BN) cells displaying
the short term, but negatively associated in the longer biomarkers of CIN following culture in medium containing 30,
term 300, or 3,000 nmol/L FA over 42 days; B, 1 micronuclei (MN); C, 1
The relationships between [FA], TL, and CIN have not nucleoplasmic bridges (NPB); D, 1 nuclear buds (NBud); E, "fused"
(FUS) nuclei; F, combined total frequency of BN cells (per 1,000
previously been examined. Biomarkers of CIN were BN cells) displaying any CIN biomarker (MN or NPB or NBud or FUS;
scored in binucleated cells using the CBMN-cyt assay, mean  SD; N ¼ 2). Data at each time point not sharing the same
specifically; micronuclei (MN; broken fragments or lag- superscript letter differ significantly (P  0.05), as measured by the
ging chromosomes), nucleoplasmic bridges (NPB; indic- Bonferroni post-test. Two-way ANOVA analyses shown (P value;
% variance [in parentheses]; Int, interaction of [FA] with time).
ative of dicentric chromosome (DC) formation due to

130 Cancer Prev Res; 7(1) January 2014 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Telomeres

Table 1. Correlations between biomarkers of CIN, concentration of FA in culture medium, and TL

DNA damage
(all markers
MN NPB NBud FUS combined) TL day 14 TL day 42
Log [FA] P 0.0005 0.02 0.7 0.07 0.01 0.0003 0.065
r 0.98 0.88 0.21 0.77 0.9 0.98 0.8
MN P 0.01 0.9 0.05 0.005 0.005 0.03
r 0.9 0.04 0.81 0.94 0.94 0.85
NPB P 0.8 0.003 0.0002 0.004 0.002
r 0.11 0.96 0.98 0.95 0.97
NBud P 0.58 0.77 0.02 0.64
r 0.29 0.15 0.9 0.24
FUS P 0.003 0.01 0.01
r 0.95 0.9 0.91
DNA damage P 0.0002 0.003
(all markers combined) r 0.98 0.95

NOTE: Shaded values indicate statistically significant associations (P < 0.05), r ¼ Pearson correlation coefficient. DNA damage
(all markers combined), frequency of binucleated cells displaying one or more MN, NPB, NBud, or FUS.
All data are calculated using duplicate values at day 42, except TL data, which use mean TL values for each condition at two key time
points: days 14 and 42.

fusion of dysfunctional or critically short telomeres, or Hypomethylation of the subtelomeric promoter for "telo-
misrepair of DNA breaks), nuclear buds (NBud; gene meric repeat-containing RNA" (TERRA) may influence the
amplification; ref. 27), and fused nuclei (FUS; suggesting ALT phenotype, directly or indirectly (11, 31, 32). Accord-
failed chromatid separation and/or dysfunctional meta- ingly, we considered whether the increased TL and CIN in
phase/anaphase transition; ref. 9). FA deficiency induced WIL2-NS cells from FA-deficient cultures may be due to ALT-
highly significant increases in the frequency of all bio- associated recombination, the latter potentially inducing
markers over 42 days (P < 0.0001–P ¼ 0.0009; Fig. 1B–F). formation of DC containing interstitial telomeric DNA,
Consistent with previous (shorter term) findings, micro- initiating telomere amplification via breakage–fusion–
nuclei, NPBs, and FUS were positively correlated, yet fre- bridge (BFB) cycling.
quencies of each were strongly, negatively associated with To elucidate the mechanism of telomere elongation
[FA] (Table 1; refs. 9, 10). Relationships between TL and induced by FA deficiency, we explored (i) intrachromoso-
CIN biomarkers at day14 (the point at which the longest TL mal (interstitial) telomeric DNA to determine if amplifica-
was observed) were positive and significant. By day 42, this tion may have occurred as a result of BFB cycling; (ii) the
had reversed, with correlations between CIN and TL being ALT or telomerase status of WIL2-NS cells; (iii) whether FA
strongly negative (Table 1). deficiency reduced global DNA methylation, and the asso-
These data show that FA deficiency induces both shorter ciation between DNA methylation and TL; and (iv) the
and longer telomeres, and that both states are strongly impact of FA deficiency on expression of hTERT, the active
associated with CIN, indicating telomere dysfunction subunit of telomerase.
regardless of length.
BFB cycling is not responsible for amplification of
Possible mechanisms underlying the increase in TL telomeric DNA
Questions that arise from the above findings are the Numerous studies report increased incidence of ana-
following: (i) What mechanisms are responsible for the phase bridges when TL and/or integrity is compromised
increase in TL in folate-deficient cultures? and (ii) Does a (33–36); however, to our knowledge, evidence of telomeric
causal link exist between these and the evidence of CIN? DNA amplification through this mechanism has not been
Telomere shortening is a pivotal event early in the patho- reported. DC formation is a known initiator of BFB cycling,
genesis of cancer; however, in established cancers telome- gene amplification, and altered gene dosage (34, 36). We
rase activity (TA) can induce longer TL than that of adjacent speculated that random breakage of DCs formed by fusion
normal tissue (28). An alternative TA-independent, recom- of dysfunctional telomeres may result in daughter cells
bination-driven telomere maintenance mechanism (TMM), receiving uncapped (fusigenic) chromosomes; one contain-
called the alternative lengthening of telomeres (ALT), has ing telomere sequences of both fused chromosomes (inter-
been identified in approximately 10% of cancers (29, 30). stitially), and the other receiving a reduced telomere content

www.aacrjournals.org Cancer Prev Res; 7(1) January 2014 131

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Bull et al.

Figure 2. Investigations into mechanisms underlying transient telomere elongation in low FA culture conditions. A, proposed model for telomeric DNA
amplification via BFB cycling, wherein compromised (uncapped or broken) telomere ends fuse to form a dicentric chromosome containing intrachromosomal
telomeric DNA. Red squares, telomeric DNA; green circles, centromeres. B, southern (TRF) analysis of (day 0, untreated) WIL2-NS cells (reference cell lines;
 þ þ þ  þ
HeLa, ALT /TA , VA13, ALT ). C, detection of C-circles (CC) shows WIL2-NS cells to be CC-negative compared with ALT (IIICF/c) and ALT /TA (HeLa)
reference cells. D, WIL2-NS cells are positive (þ) for TA, determined using the IP-TRAP assay. Sizes of molecular weight markers are indicated to the
þ þ 
left of B and D. Buffer, buffer-only negative control; HCT116, telomerase-positive (TA ) reference cell line; GM847, ALT /TA reference cell line. E and F, global
methylation status of WIL2-NS cells cultured for 14 days and 42 days (E) in medium containing 30 or 3,000 nmol/L FA (a, P ¼ 0.046; b, P ¼ 0.06); and for 4 days
(F) in complete medium (CM; 3,000 nmol/L FA) containing 0, 0.2, or 1.0 mmol/L 5azadC (points not sharing the same letter differ significantly; P < 0.05).
%5-meC; percentage of cytosine residues methylated at carbon-5, as a percentage of total DNA; mean  SD; N ¼ 3. G, TL of WIL2-NS cells cultured
for 4 days in CM containing 0, 0.2, or 1.0 mmol/L 5azadC. Mean  SD; N ¼ 20 at day 0, n ¼ 10 at day 4 (points not sharing the same letter differ significantly:
P < 0.05).

(Fig. 2A). To test the hypothesis that BFB cycling was (APB), and C-circles (29, 30). C-circles are a specific, quan-
responsible for telomere amplification, metaphase prepara- tifiable marker of ALT (29, 30). To test whether WIL2-NS
tions from cells grown in control (3,000 nmol/L) or defi- cells are ALTþ, we examined untreated (day 0) cells first by
cient (30 nmol/L) FA for 0, 7, 14, 21, 28, or 35 days were Southern blot to assess the degree of TL heterogeneity in
probed using PNA-FISH for the presence of interstitial TRF, and second for the presence of C-circles by the C-circle
telomeric DNA (i.e., telomeric sequences at nonterminal assay (30). TRF analysis showed a comparable spread of TL
locations). Analysis showed no interstitial telomeric DNA relative to HeLa cells (ALT/TAþ), and smaller, less hetero-
in these metaphases (n ¼ 40 metaphases per condition, per geneous fragments than those from VA13 cells (ALTþ/
time point; data not shown), thus refuting this hypothesis. TA; Fig. 2B). WIL2-NS cells were then shown to be negative
for C-circles relative to ALTþ IIICF/c reference cells (Fig. 2C).
WIL2-NS cells are ALT-negative and telomerase- Together, these results suggest that WIL2-NS are ALT.
positive The IP-TRAP assay showed WIL2-NS are positive for TA
ALTþ cells are characterized by highly heterogeneous TL (Fig. 2D), consistent with an ALT phenotype (29), and
distribution (potentially ranging from <3 kb to >50 kb), the suggesting that the increase in TL observed at days 7 and 14
presence of ALT-associated promyelocytic leukemia bodies in the FA-deficient cultures may be mediated by telomerase.

132 Cancer Prev Res; 7(1) January 2014 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Telomeres

FA deficiency results in global DNA hypomethylation Relative to 3,000 nmol/L cultures, cells in 30 nmol/L FA
The relationship between DNA methylation status and TL had 74% less DNA methylation at day 14, and 26% longer
is currently unclear. Telomeres lack CpG sites, the substrate TL. Similarly, the 30% reduction in methylation in the 0.2
for DNA methyltransferase (DNMT) enzymes, and, as a mmol/L 5azadC culture had 30% longer TL, whereas the
consequence, are unmethylated. In contrast, the subtelo- 355% reduction in methylation in the 1.0 mmol/L condition
mere is normally heavily methylated, reduction in which was associated with 50% longer TL, relative to control
(in gene knockout studies) has been associated with cultures. From these data, we concluded that DNA hypo-
dramatically elongated telomeres, APBs, and telomeric methylation is a plausible candidate as the mechanism
sister chromatid exchange (TSCE; refs. 11, 32). Subtelo- underlying increased TL observed after 7 to 14 days of
meric and pericentromeric DNA hypomethylation was suboptimal FA.
also associated with increased TL and TSCE in a panel
of neoplastic cells (12), suggesting a plausible association Expression of hTERT is unaffected by FA deficiency
between global and subtelomeric methylation status, and TA is dependent upon presence of the active subunit,
telomere dysfunction. hTERT, which is regulated at the transcriptional level
The relationship between folate, DNA methylation, and (38). Animal models show that diets deficient for methyl-
TL has not previously been reported. We hypothesized that donors (including folate) increased DNMT expression,
under FA-deficient conditions, TL and global DNA methyl- resulting in CpG island hypermethylation, in certain
ation status would be negatively associated. An ELISA-based tissues (39), an effect that may plausibly impact hTERT
assay was used to determine the percentage of 5-methyl- expression. Accordingly, we determined whether hTERT
cytosine (%5me-C) in cells cultured in 30 nmol/L and 3,000 expression was altered in WIL2-NS under FA-deficient
nmol/L FA at day 14 and day 42. Results showed global conditions by quantifying RNA transcripts in cells
DNA methylation was lower in the 30 nmol/L compared following 14, 28, 35, and 42 days in 30, 300, or 3,000
with 3,000 nmol/L FA cultures, at both time points (Fig. nmol/L FA culture. qPCR results, expressed relative to
2E); 16% of variance was attributable to [FA] (ANOVA, P ¼ baseline (day 0), normalized against housekeeping gene
0.03), 43% to time (P ¼ 0.005), and 11% to their interac- glyceraldehyde-3-phosphate dehydrogenase (GAPDH),
tion (P ¼ 0.15). Methylation status and [FA] were positively showed no significant differences, with only 5.2% of
associated at day 14 (r ¼ 0.8, P ¼ 0.05) and day 42 (r ¼ 0.8, variance attributable to [FA] (P ¼ 0.77), and 18.2% of
P ¼ 0.06). In agreement with our hypothesis, at day 14, the variance due to time (P ¼ 0.75; data not shown).
point at which TL was longest in the 30 nmol/L FA culture, Although telomerase is active in WIL2-NS, it seems that
TL was negatively and significantly associated with global neither FA deficiency per se, nor an associated change in
methylation status (r ¼ 0.8, P ¼ 0.05). DNA methylation status, impacts transcription of hTERT,
thus increased TA is unlikely to have contributed to the
Global hypomethylation is associated with increased longer TL observed.
TL
To examine the impact of hypomethylation on TL, without Mechanisms for accelerated telomere loss
the potentially confounding effect of reduced intracellular Uracil incorporation into genomic DNA under folate-
thymidine through FA deprivation, the protocol was repeat- deficient culture conditions is well documented (14), but
ed using WIL2-NS in complete medium (3,000 nmol/L FA) whether uracil is incorporated into telomeres has not pre-
containing DNMT inhibitor, 5azadC. To minimize cytotoxic viously been examined. A functional telosome, key com-
effects, the lowest possible (bioefficacious) concentrations ponents of which are capping proteins TRF1 and TRF2,
(0.2 and 1.0 mmol/L) were used (37). Treatment could only inhibits nonhomologous end joining and end fusions (40).
be maintained for 4 days before cells became nonviable; The high specificity of capping proteins for their substrate
however, as TL increased in cells in 30 nmol/L FA within 7 was demonstrated in an elegant study showing substitution
days, 4 days exposure to 5azadC was deemed sufficient to of a single telomeric guanine with 8-oxo-guanine (8oxoG)
induce a comparable effect. resulted in 50% reduction in TRF1/2 binding, an abasic
We verified that methylation status of WIL2-NS exposed to site within the telomere reduced binding 3-fold, and a
0.2 and 1.0 mmol/L 5azadC was significantly lower than lesion within each hexamer repeat reduced bound TRF1
those from control (0 mmol/L) cultures; 21% of variance and TRF2 to barely detectable levels (21). Accordingly, we
attributable to [5azadC] (ANOVA, P ¼ 0.05; Fig. 2F). The hypothesized that uracil in telomeric DNA may impact
correlation between [5azadC] and DNA methylation at day 4 telosome integrity in a similar manner. Furthermore, uracil
was significant and negative (r ¼ 0.8, P ¼ 0.01). glycosylase (UDG), part of the base excision repair (BER)
At day 4, TL was 29% and 50% greater, in the 0.2 and 1.0 mechanism, removes uracil from genomic DNA, resulting
mmol/L 5azadC, respectively, than in untreated controls in a transient abasic site intermediate (15, 16). In low FA
(Fig. 2G). [5azadC] explained 27% of TL variance (P < (low thymidine) conditions, uracil can be reincorporated at
0.0001), 39% due to time (P < 0.0001) and 27% due to the "gap-filling" step, leading to futile cycling of BER and
their interaction (P < 0.0001). A significant, negative cor- abasic site generation, potentially compromising TRF1/2
relation was recorded between global DNA methylation binding, and increasing the risk of DSB (15, 16). Recent
and TL in these samples (r ¼ 0.75, P ¼ 0.02). findings indicate, however, that telomeric DNA damage is

www.aacrjournals.org Cancer Prev Res; 7(1) January 2014 133

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Bull et al.

not repaired (18, 19), resulting in a persistent damage (n ¼ 4, P ¼ 0.3). The slightly greater amount of uracil
response (phosphorylated (g)-H2AX and/or ATM) at observed in the 300 nmol/L culture (compared with 30
lesions, prolonged cell cycle arrest, and potentially replica- nmol/L) is consistent with previous findings in genomic
tive senescence. DNA, an effect possibly due to the different rates of DNA
To examine the factors underlying the telomere attrition synthesis and cell division between the midrange and
observed in extended 30 nmol/L FA cultures, we tested (i) lowest [FA], thus influencing the rate of uracil incorporation
the relationship between [FA] and uracil within telomeres, (14). As predicted, [FA] and uracil content in telomeric DNA
(ii) the presence of telomeric DNA in biomarkers of DNA were negatively associated (r ¼ 0.47, P ¼ 0.1) after 42 days
damage in cytokinesis-blocked binucleated cells, and (iii) in culture.
"loss of telomere" events in metaphase preparations.
FA deficiency is associated with DSBs, loss of terminal
Uracil incorporation into telomeric DNA is negatively telomeric fragments, and mitotic dysfunction
associated with [FA] in medium To examine the in situ location of telomeric and centro-
Uracil content of telomeric DNA was measured in UDG- meric DNA in FA-deficient cultures, dual fluorochrome
digested and -undigested DNA using an adaptation of a PNA-FISH was conducted on cytokinesis-blocked binucle-
qPCR method developed in this laboratory to detect abnor- ated cells cultured in 30 nmol/L FA for 7, 14, 21, 28, or 35
mal bases by digesting DNA with lesion-specific enzymes days. Of 151 micronuclei scored (day 7, n ¼ 37; day 14, n ¼
(41). Results showed that, following 42 days of culture in 21; day 21, n ¼ 41; day 28, n ¼ 31; day 35, n ¼ 21), 38%
either 30 nmol/L or 300 nmol/L FA, the amount of uracil contained centromeric DNA (Cþ) and 79% contained
per kb of telomeric DNA was 2.9- and 3.4-fold greater, telomeric DNA (Tþ; Fig. 3A–C). Consistent with previous
respectively than at baseline, whereas no uracil was detect- data from 9-day FA-deficient cultures (42), only 25% of the
able in telomeres of cells from 3,000 nmol/L FA cultures 53 micronuclei containing 2 T signals were also Cþ

Figure 3. Representative PNA-FISH images of CIN biomarkers in cytokinesis-blocked binucleated (BN) WIL2-NS cells cultured in medium containing
30 nmol/L FA. Black and white images indicate 4',6-diamidino-2-phenylindole (DAPI) staining of DNA (white), with arrows indicating the location of CIN
þ
biomarkers. Corresponding color images show DAPI-stained DNA (blue), the location of 5-carboxy-fluorescein (FAM)-labeled centromeric probe (green; C )
þ
and/or Cy3-labeled telomeric probe (red; T ), visualized using PNA-FISH. A–C, BN cells after 7, 21, and 28 days in culture, respectively, each
þ þ þ þ  þ
containing a single C T micronucleus, the micronucleus in B contains 2C and 4T signals, and C also contains a single C T NPB. D (28 days culture),
   þ
E and F (35 days cultures), BN cells containing 1 or 2 NPB. All are C T , except for the left NPB in F which is C T . G, H, and I, fused (FUS) cells after
þ þ
(G, 28 days), (H and I, 35 days) in culture. Dashed lines indicate nuclear boundaries of fusion regions (FR) for scoring purposes. In each case, FR are C T .
þ þ þ þ þ þ þ
G, contains 4C , 1T , the FR of (H) contain 3T , 2C , and FR of (I) contain 1T , 1C . The large size of C signals suggests close proximity of more
than one hybridization site (indicated by arrows in H and I); magnification, 630.

134 Cancer Prev Res; 7(1) January 2014 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Telomeres

A C D E F
3,000 nmol/L
30 nmol/L
3.5
G H I
3.0
LOT (Relative to day 0)

2.5

2.0 J K L
1.5

1.0

0.5

0.0 M N
0 7 14 21 28 35
Day
B

O P

Day 14 Day 21 Day 28 Day 35

Figure 4. Chromosome aberrations in cells cultured in FA-deficient conditions. A, Loss of telomere (LOT) events in WIL2-NS metaphases cultured for 35 days
in medium containing 30 or 3,000 nmol/L FA. [Mean  SD, n ¼ 2, 20 metaphase spreads scored per duplicate, a total of 40 per condition per time point. Data
not sharing the same superscript letter at each time point differ significantly, measured by the Bonferroni post-test (P  0.05)]. B–P, PNA-FISH–stained partial
metaphase spreads from WIL2-NS cultured in 30 or 3,000 nmol/L FA; DAPI-stained DNA (blue), FAM-labeled centromeric DNA (green), Cy3-labeled
telomeric DNA (red). B, representative images of LOT events from 30 nmol/L FA cultures (yellow arrows, single LOT event; white arrows, double LOT event).
C–P, chromosome aberrations (yellow arrows, centromeres). C, a dicentric chromosome (DC) observed after 21 days in 3,000 nmol/L FA. (D–P are all from
30 nmol/L FA cultures); (D, E; day 14) ring chromosomes formed by end fusion (pink arrow) within the same chromosome; (F, day 21) DC; (G and I, day 14) DC;
(H, day 14) q-arm fusion (pink arrow); (J, K, day 28) DC; (L–P, day 35) DC. Image M also contains an acentric fragment (pink arrow), representing a
fusion between two broken chromosome ends; magnification, 630.

(indicating a lagging chromatid or chromosome), suggest- FA deficiency causes telomere loss and chromosome
ing that the majority of Tþ micronuclei represent acentric aberrations
fragments arising from DSB. Of 21 NPBs scored after days 7 "Loss of telomere" (LOT) events were scored in meta-
(n ¼ 6), 14 (n ¼ 1), 21 (n ¼ 2), 28 (n ¼ 8), or 35 (n ¼ 4) in 30 phase spreads from cells cultured in 30 or 3,000 nmol/L
nmol/L FA (Fig. 3D–F), 38% were Tþ, suggesting fusion FA for days 0, 7, 14, 21, 28, or 35 (20 metaphases/slide,
between dysfunctional or compromised telomeres, 43% n ¼ 2; Fig. 4A and B). Data were expressed relative to that of
were Cþ, and 24% contained both (CþTþ), which together baseline cultures (set arbitrarily at 1.0). Confirming indica-
suggest mitotic disruption (9). The greatest proportion tions from the above data, metaphases from 30 nmol/L FA
(43%) was for CT, suggesting that these NPBs arose from cultures had an approximately 3-fold increase in LOT from
fusion between uncapped chromosomes, or DSB misrepair day 14 to day 35, compared with cells maintained in 3,000
of breaks at nontelomeric chromosome regions. Cells dis- nmol/L FA. Of the variance in LOT, 43% was attributable to
playing FUS morphologies (n ¼ 196) were scored following [FA] (P ¼ 0.0001), 22% to time (P ¼ 0.04), and 18% to their
days 7 (n ¼ 37), 14 (n ¼ 21), 21 (n ¼ 40), 28 (n ¼ 30), or 35 interaction (P ¼ 0.08). At day 35, LOT was negatively
(n ¼ 21) in 30 nmol/L FA (Fig. 3G–I). The majority (78%) of correlated with log[FA] (r ¼ 0.96, P ¼ 0.03), and positively
fusion regions were Cþ, and/or Tþ (74%), with 61% exhi- correlated with binucleated cells containing 1 micronuclei
biting both (CþTþ) and only 8% displaying neither (CT). (r ¼ 0.96, P ¼ 0.04).
These results suggest that telomere end fusions or failure of These same spreads were analyzed for aberrations (DC,
sister chromatid separation could cause FUS morphologies acentric fragments, double minutes, chromosome or chro-
supporting our previous observations (9). matid breaks, ring chromosomes, q or p arm fusions). The

www.aacrjournals.org Cancer Prev Res; 7(1) January 2014 135

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Bull et al.

Figure 5. Possible pathways by which folate insufficiency could impact TL, telomere integrity, and chromosomal stability. SAM, methyl carrier/donor
S-adenosyl methionine; ALT, the telomerase-independent (alternative) mechanism for maintenance of TL; MRN complex, DNA repair complex associated
with ALT, consisting of Mre11, Rad50, and Nbs1.

presence of DC was of particular interest, as they represent term net gain in telomere content arising from hypomethy-
end fusions from telomere loss or dysfunction, or DSB lation, reduced capping and enhanced access by telomerase
misrepair. Only one aberration, a DC at day 21, was to its substrate. In the longer term, however, as telomere
observed in 3,000 nmol/L FA spreads (Fig. 4C). In contrast, maintenance and DNA repair mechanisms become over-
21 aberrations were recorded from 30 nmol/L FA cultures, whelmed (due to increasing uracil incorporation, BER pro-
10 of which were DC (2 at day 14, 1 at day 21, 2 at day 28, 5 cesses, abasic sites, and DSB), homeostatic balance is lost
at day 35; Fig. 4D–P). No aberrations were recorded at day 0 resulting in increasing telomere attrition with time. Fur-
or day 7, from 30 or 3,000 nmol/L FA cultures. thermore, we provide evidence that the rapid telomere loss
Together, these data show long-term FA deficiency is observed beyond day 14 in the 30 nmol/L FA condition
associated with micronuclei containing acentric fragments coincides with an increase in terminal deletions, leaving
and lagging chromosomes, terminal chromosome loss, exposed chromosome ends to which telomerase can no
fused chromosomes (including DC), apparent dysfunction- longer bind, and is thus unable to add telomeric repeats. A
al chromatid separation, and uracil in telomeric DNA. These diagrammatic summary of proposed pathways underlying
findings are consistent with those that may be expected FA-deficiency–induced telomeric and genomic instability is
from uracil incorporation into genomic and telomeric provided in Fig. 5.
DNA, and support our original hypothesis that FA deficien- Increased TL under hypomethylating conditions has only
cy could cause accelerated telomere erosion. been reported previously in mouse knockout models
(11, 32), or in cell lines exposed to cancer drugs such as
Conclusions 5azadC (12). The present data, however, show telomere
The novel data herein demonstrate that FA deficiency lengthening and DNA hypomethylation induced by FA
compromises telomere homeostasis, evidenced by substan- deficiency to a similar extent as that caused by 5azadC. As
tial fluctuations in TL, increased chromosome fusions epigenetic modifications are strongly associated with cancer
(NPBs and DCs), and terminal deletions. These effects initiation, these findings potentially have wide-reaching
coincide with global DNA hypomethylation, increased implications for disease risk minimization in populations
presence of uracil in telomeres, and increased CIN. We exposed to deficiency of folate, vitamins required for folate
propose a model whereby FA deficiency results in short- metabolism (such as vitamin B12), or essential factors in

136 Cancer Prev Res; 7(1) January 2014 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Telomeres

one-carbon (methyl) metabolism (e.g., choline; ref. 43). Authors' Contributions


Until testing has been undertaken in nontransformed cells, Conception and design: C.F. Bull, G. Mayrhofer, M.F. Fenech
Development of methodology: C.F. Bull, N.J. O’Callaghan, M.F. Fenech
however, these data can only be considered directly relevant Acquisition of data (provided animals, acquired and managed patients,
to p53-negative cells with active telomerase, and can not be provided facilities, etc.): C.F. Bull, A.Y. Au, H.A. Pickett, M.P. Hande
Analysis and interpretation of data (e.g., statistical analysis, biosta-
extrapolated to primary lymphocytes. As such, these obser- tistics, computational analysis): C.F. Bull, G. Mayrhofer, N.J. O’Calla-
vations are relevant to cancer cells, and highlight how ghan, H.A. Pickett, G.K.M. Low, D. Zeegers, M.P. Hande, M.F. Fenech
periods of FA deficiency may compromise TL homeostasis Writing, review, and/or revision of the manuscript: C.F. Bull, G. Mayr-
hofer, N.J. O’Callaghan, G.K.M. Low, D. Zeegers, M.P. Hande, M.F. Fenech
and functional integrity. We also can not exclude the pos- Study supervision: C.F. Bull, G. Mayrhofer, M.F. Fenech
sibility that some of the effects we observed are specific to
FA, as opposed to methyltetrahydrofolate, a form of the
cofactor to which lymphoid cells would also be exposed in Acknowledgments
vivo. Furthermore, senescence-induced inflammation has The authors thank M. Hor for technical assistance for the 5azadC work,
and S. Mitchell [the Commonwealth Scientific and Industrial Research
been associated with enhanced cancer promotion (44), an Organisation (CSIRO), Sydney, Australia] and V. Dhillon (CSIRO, Adelaide,
effect likely to be increased when telomeres are damaged Australia) for generating preliminary methylation status data for these
samples.
and/or become dysfunctional. Together, these mechanisms
may provide plausible bases for recent prospective epide-
miologic studies showing positive associations between TL Grant Support
and cancer risk (28). C.F. Bull was supported by an Australian Postgraduate Award (APA)
Together, these results demonstrate the impact of methyl Scholarship through the University of Adelaide, CSIRO’s Preventative Health
Flagship, and a Science Industry Endowment Fund (SIEF) John Stocker
insufficiency on telomere homeostasis and provide evi- Postdoctoral Research Fellowship.
dence that dysfunctional long telomeres may be as impor- The costs of publication of this article were defrayed in part by the
tant as critically short telomeres as a cause of chromosomal payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
instability in p53-negative cells. this fact.

Disclosure of Potential Conflicts of Interest Received July 14, 2013; revised October 30, 2013; accepted November 1,
No potential conflicts of interest were disclosed. 2013; published OnlineFirst November 19, 2013.

References
1. Blackburn EH. Telomeres: no end in sight. Cell 1994;77:621–3. 13. Jaco I, Canela A, Vera E, Blasco MA. Centromere mitotic recombina-
2. Lin J, Epel E, Blackburn E. Telomeres and lifestyle factors: roles in tion in mammalian cells. J Cell Biol 2008;181:885–92.
cellular aging. Mutat Res 2012;730:85–9. 14. Mashiyama ST, Courtemanche C, Elson-Schwab I, Crott J, Lee BL,
3. Sun Q, Shi L, Prescott J, Chiuve SE, Hu FB, De Vivo I, et al. Healthy Ong CN, et al. Uracil in DNA, determined by an improved assay, is
lifestyle and leukocyte telomere length in U.S. women. PLoS One increased when deoxynucleosides are added to folate-deficient cul-
2012;7:e38374. tured human lymphocytes. Anal Biochem 2004;330:58–69.
4. Epel ES, Blackburn EH, Lin J, Dhabhar FS, Adler NE, Morrow JD, et al. 15. Blount BC, Mack MM, Wehr CM, MacGregor JT, Hiatt RA, Wang G,
Accelerated telomere shortening in response to life stress. Proc Natl et al. Folate deficiency causes uracil misincorporation into human DNA
Acad Sci U S A 2004;101:17312–5. and chromosome breakage: implications for cancer and neuronal
5. Shalev I. Early life stress and telomere length: investigating the con- damage. Proc Natl Acad Sci U S A 1997;94:3290–5.
nection and possible mechanisms: a critical survey of the evidence 16. Dianov GL, Timchenko TV, Sinitsina OI, Kuzminov AV, Medvedev OA,
base, research methodology and basic biology. Bioessays 2012;34: Salganik RI. Repair of uracil residues closely spaced on the opposite
943–52. strands of plasmid DNA results in double-strand break and deletion
6. Wolkowitz OM, Mellon SH, Epel ES, Lin J, Dhabhar FS, Su Y, et al. formation. Mol Gen Genet 1991;225:448–52.
Leukocyte telomere length in major depression: correlations with 17. Michelson RJ, Rosenstein S, Weinert T. A telomeric repeat sequence
chronicity, inflammation and oxidative stress–preliminary findings. adjacent to a DNA double-stranded break produces an anticheck-
PLoS One 2011;6:e17837. point. Genes Dev 2005;19:2546–59.
7. Ornish D, Lin J, Daubenmier J, Weidner G, Epel E, Kemp C, et al. 18. Hewitt G, Jurk D, Marques FD, Correia-Melo C, Hardy T, Gackowska A,
Increased telomerase activity and comprehensive lifestyle changes: a et al. Telomeres are favoured targets of a persistent DNA damage
pilot study. Lancet Oncol 2008;9:1048–57. response in ageing and stress-induced senescence. Nat Commun
8. Puterman E, Lin J, Blackburn E, O'Donovan A, Adler N, Epel E. The 2012;3:708.
power of exercise: buffering the effect of chronic stress on telomere 19. Fumagalli M, Rossiello F, Clerici M, Barozzi S, Cittaro D, Kaplunov
length. PLoS One 2010;5:e10837. JM, et al. Telomeric DNA damage is irreparable and causes per-
9. Bull CF, Mayrhofer G, Zeegers D, Mun GL, Hande MP, Fenech MF. sistent DNA-damage-response activation. Nat Cell Biol 2012;14:
Folate deficiency is associated with the formation of complex nuclear 355–65.
anomalies in the cytokinesis-block micronucleus cytome assay. Envi- 20. de Lange T. How telomeres solve the end-protection problem. Science
ron Mol Mutagen 2012;53:311–23. 2009;326:948–52.
10. Fenech M. Folate (vitamin B9) and vitamin B12 and their function in the 21. Opresko P, Fan J, Danzy S, Wilson D, Bohr V. Oxidative damage in
maintenance of nuclear and mitochondrial genome integrity. Mutat telomeric DNA disrupts recognition by TRF1 and TRF2. Nucleic Acids
Res 2012;733:21–33. Res 2005;33:1230–9.
11. Blasco MA. The epigenetic regulation of mammalian telomeres. Nat 22. Beetstra S, Thomas P, Salisbury C, Turner J, Fenech M. Folic acid
Rev Genet 2007;8:299–309. deficiency increases chromosomal instability, chromosome 21 aneu-
12. Vera E, Canela A, Fraga MF, Esteller M, Blasco MA. Epigenetic ploidy and sensitivity to radiation-induced micronuclei. Mutat Res
regulation of telomeres in human cancer. Oncogene 2008;27:6817–33. 2005;578:317–26.

www.aacrjournals.org Cancer Prev Res; 7(1) January 2014 137

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Bull et al.

23. Koutros S, Zhang Y, Zhu Y, Mayne ST, Zahm SH, Holford TR, et al. 34. Gisselsson D, Hoglund M. Connecting mitotic instability and chromo-
Nutrients contributing to one-carbon metabolism and risk of non- some aberrations in cancer–can telomeres bridge the gap? Semin
Hodgkin lymphoma subtypes. Am J Epidemiol 2008;167:287–94. Cancer Biol 2005;15:13–23.
24. Umegaki K, Fenech M. Cytokinesis-block micronucleus assay in 35. Stewenius Y, Gorunova L, Jonson T, Larsson N, Hoglund M, Mandahl
WIL2-NS cells: a sensitive system to detect chromosomal damage N, et al. Structural and numerical chromosome changes in colon
induced by reactive oxygen species and activated human neutrophils. cancer develop through telomere-mediated anaphase bridges, not
Mutagenesis 2000;15:261–9. through mitotic multipolarity. Proc Natl Acad Sci U S A 2005;102:
25. Amundson SA, Xia F, Wolfson K, Liber HL. Different cytotoxic and 5541–6.
mutagenic responses induced by X-rays in two human lymphoblastoid 36. Albertson DG. Gene amplification in cancer. Trends Genet 2006;22:
cell lines derived from a single donor. Mutat Res 1993;286:233–41. 447–55.
26. Bull CF, O'Callaghan NJ, Mayrhofer G, Fenech MF. Telomere length in 37. Stresemann C, Brueckner B, Musch T, Stopper H, Lyko F. Functional
lymphocytes of older South Australian men may be inversely associ- diversity of DNA methyltransferase inhibitors in human cancer cell
ated with plasma homocysteine. Rejuvenation Res 2009;12:341–9. lines. Cancer Res 2006;66:2794–800.
27. Fenech M. Cytokinesis-block micronucleus cytome assay. Nat Protoc 38. Aisner DL, Wright WE, Shay JW. Telomerase regulation: not just
2007;2:1084–104. flipping the switch. Curr Opin Genet Dev 2002;12:80–5.
28. Svenson U, Roos G. Telomere length as a biological marker in malig- 39. Ghoshal K, Li X, Datta J, Bai S, Pogribny I, Pogribny M, et al. A
nancy. Biochim Biophys Acta 2009;1792:317–23. folate- and methyl-deficient diet alters the expression of DNA
29. Henson JD, Reddel RR. Assaying and investigating alternative length- methyltransferases and methyl CpG binding proteins involved in
ening of telomeres activity in human cells and cancers. FEBS Lett epigenetic gene silencing in livers of F344 rats. J Nutr 2006;136:
2010;584:3800–11. 1522–7.
30. Henson JD, Cao Y, Huschtscha LI, Chang AC, Au AY, Pickett HA, et al. 40. van Steensel B, Smogorzewska A, de Lange T. TRF2 protects human
DNA C-circles are specific and quantifiable markers of alternative- telomeres from end-to-end fusions. Cell 1998;92:401–13.
lengthening-of-telomeres activity. Nat Biotechnol 2009;27:1181–5. 41. O'Callaghan N, Baack N, Sharif R, Fenech M. A qPCR-based assay to
31. Ng LJ, Cropley JE, Pickett HA, Reddel RR, Suter CM. Telomerase quantify oxidized guanine and other FPG-sensitive base lesions within
activity is associated with an increase in DNA methylation at the telomeric DNA. Biotechniques 2011;51:403–11.
proximal subtelomere and a reduction in telomeric transcription. 42. Lindberg HK, Wang X, Jarventaus H, Falck GC, Norppa H, Fenech M.
Nucleic Acids Res 2009;37:1152–9. Origin of nuclear buds and micronuclei in normal and folate-deprived
32. Gonzalo S, Jaco I, Fraga MF, Chen T, Li E, Esteller M, et al. DNA human lymphocytes. Mutat Res 2007;617:33–45.
methyltransferases control telomere length and telomere recombina- 43. Lu L, Ni J, Zhou T, Xu W, Fenech M, Wang X. Choline and/or folic acid
tion in mammalian cells. Nat Cell Biol 2006;8:416–24. deficiency is associated with genomic damage and cell death in human
33. Slijepcevic P, Hande MP, Bouffler SD, Lansdorp P, Bryant PE. Telo- lymphocytes in vitro. Nutr Cancer 2012;64:481–7.
mere length, chromatin structure and chromosome fusigenic potential. 44. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol
Chromosoma 1997;106:413–21. 2013;75:685–705.

138 Cancer Prev Res; 7(1) January 2014 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.
Published OnlineFirst November 19, 2013; DOI: 10.1158/1940-6207.CAPR-13-0264

Folate Deficiency Induces Dysfunctional Long and Short


Telomeres; Both States Are Associated with Hypomethylation and
DNA Damage in Human WIL2-NS Cells
Caroline F. Bull, Graham Mayrhofer, Nathan J. O'Callaghan, et al.

Cancer Prev Res 2014;7:128-138. Published OnlineFirst November 19, 2013.

Updated version Access the most recent version of this article at:
doi:10.1158/1940-6207.CAPR-13-0264

Supplementary Access the most recent supplemental material at:


Material http://cancerpreventionresearch.aacrjournals.org/content/suppl/2013/11/20/1940-6207.CAPR-13-0264.DC
1.html

Cited articles This article cites 44 articles, 12 of which you can access for free at:
http://cancerpreventionresearch.aacrjournals.org/content/7/1/128.full.html#ref-list-1

Citing articles This article has been cited by 7 HighWire-hosted articles. Access the articles at:
http://cancerpreventionresearch.aacrjournals.org/content/7/1/128.full.html#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Subscriptions pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications Department at
permissions@aacr.org.

Downloaded from cancerpreventionresearch.aacrjournals.org on March 31, 2016. © 2014 American Association for Cancer
Research.

Potrebbero piacerti anche